1
|
Gebrie A. The melanocortin receptor signaling system and its role in neuroprotection against neurodegeneration: Therapeutic insights. Ann N Y Acad Sci 2023; 1527:30-41. [PMID: 37526975 DOI: 10.1111/nyas.15048] [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] [Indexed: 08/02/2023]
Abstract
The melanocortin signaling system consists of the melanocortin peptides, their distinctive receptors, accessory proteins, and endogenous antagonists. Melanocortin peptides are small peptide hormones that have been studied in a variety of physiological and pathological conditions. There are five types of melanocortin receptors, and they are distributed within the central nervous system and in several tissues of the periphery. The G protein-coupled melanocortin receptors typically signal through adenylyl cyclase and other downstream signaling pathways. Depending on the ligand, surface expression of melanocortin receptor, receptor occupancy period, related proteins, the type of cell, and other parameters, the signaling pathways are complicated and pleiotropic. While it is known that all five melanocortin receptors are coupled to Gs, they can also occasionally couple to Gq or Gi. Both direct and indirect neuroprotection are induced by the melanocortin signaling system. Targeting several of the components of the melanocortin signaling system (ligands, receptors, accessory proteins, signaling effectors, and regulators) may provide therapeutic opportunities. Activation of the melanocortin system improves different functional traits in neurodegenerative diseases. There is a potential for additional melanocortin system interventions by interfering with dimerization or dissociation. This review aims to discuss the melanocortin receptor signaling system and its role in neuroprotection, as well as its therapeutic potential.
Collapse
Affiliation(s)
- Alemu Gebrie
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
2
|
Stanislaus V, Kam A, Murphy L, Wolgen P, Walker G, Bilbao P, Cloud GC. A feasibility and safety study of afamelanotide in acute stroke patients - an open label, proof of concept, phase iia clinical trial. BMC Neurol 2023; 23:281. [PMID: 37496004 PMCID: PMC10373257 DOI: 10.1186/s12883-023-03338-9] [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: 12/06/2022] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Neuroprotective agents have the potential to improve the outcomes of revascularisation therapies in acute ischemic stroke patients (AIS) and in those unable to receive revascularisation. Afamelanotide, a synthetic α-melanocyte stimulating hormone analogue, is a potential novel neuroprotective agent. We set out to assess the feasibility and safety of afamelanotide for the first time in AIS patients. METHODS AIS patients within 24 h of onset, with perfusion abnormality on imaging (Tmax) and otherwise ineligible for revascularisation therapies were enrolled. Afamelanotide 16 mg implants were administered subcutaneously on Day 0 (D0, day of recruitment), D1 and repeated on D7 and D8, if not well recovered. Treatment emergent adverse events (TEAEs) and neurological assessments were recorded regularly up to D42. Magnetic resonance imaging (MRI) with FLAIR sequences were also performed on D3 and D9. RESULTS Six patients (5 women, median age 81, median NIHSS 6) were recruited. Two patients received 4 doses and four patients received 2. One patient (who received 2 doses), suffered a fatal recurrent stroke on D9 due to a known complete acute internal carotid artery occlusion, assessed as unrelated to the study drug. There were no other local or major systemic TEAEs recorded. In all surviving patients, the median NIHSS improved from 6 to 2 on D7. The median Tmax volume on D0 was 23 mL which was reduced to a FLAIR volume of 10 mL on D3 and 4 mL on D9. CONCLUSIONS Afamelanotide was well tolerated and safe in our small sample of AIS patients. It also appears to be associated with good recovery and radiological improvement of salvageable tissue which needs to be tested in randomized studies. CLINICALTRIALS GOV IDENTIFIER NCT04962503, First posted 15/07/2021.
Collapse
Affiliation(s)
- Vimal Stanislaus
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
- Alfred Hospital, Melbourne, Australia
| | | | | | | | - Gill Walker
- CLINUVEL Pharmaceuticals, Melbourne, Australia
| | | | - Geoffrey C Cloud
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia.
- Alfred Hospital, Melbourne, Australia.
| |
Collapse
|
3
|
Hu Q, Wu X, Wang Z, Yan T, Wang L, Yu W, Du Q, Hu W, Zheng Y, Wang K, Dong X, Yang D. α-MSH as a potential biomarker of severity and prognosis after intracerebral hemorrhage: A prospective cohort study. Clin Chim Acta 2023; 538:131-138. [PMID: 36402174 DOI: 10.1016/j.cca.2022.11.004] [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: 10/13/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND α-melanocyte-stimulating hormone (α-MSH) exerts anti-inflammatory and brain protective effects. We determined plasma α-MSH concentrations and discovered the relationship between plasma α-MSH concentrations and severity plus clinical outcome after intracerebral hemorrhage (ICH). METHODS A total of 117 ICH patients and 117 healthy controls were included in this study. Glasgow coma scale (GCS) score and hematoma volume were recorded to assess disease severity. We used Glasgow outcome scale (GOS) score to evaluate the 3-month clinical prognosis. And multivariate analysis was done to discern the relation of plasma α-MSH concentrations to disease severity plus poor prognosis. Receiver operating characteristic curve (ROC) was built to evaluate the prognostic predictive capability. RESULTS Plasma α-MSH concentrations in ICH patients, compared with healthy controls, were significantly decreased (median, 25.37 vs 46.80 pg/ml; P < 0.001), and were independently correlated with GCS score (t = 4.091, P < 0.001). Plasma α-MSH concentrations were highly correlated with GOS scores (ρ = 0.548, P < 0.001), were substantially lower with poor prognosis (GOS scores 1-3) than good prognosis, and efficiently discriminated patients at risk of poor prognosis (AUC ROC, 0.793; 95 % CI: 0.709-0.863). Using Youden method, plasma α-MSH concentrations < 23.63 pg/ml predicted poor prognosis with sensitivity of 72.7 % and specificity of 82.2 %. Alternatively, plasma α-MSH concentrations emerged as an independent predictor of poor prognosis with odds ratio of 0.888 (95 % CI: 0.793-0.995; P = 0.040). CONCLUSION Plasma α-MSH concentrations are significantly associated with disease severity and poor 3-month prognosis in patients with ICH, indicating that plasma α-MSH may serve as a useful potential prognostic biomarker for ICH.
Collapse
Affiliation(s)
- Qiang Hu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou 310006, China
| | - Xiaoyu Wu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou 310053, China
| | - Zefan Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou 310053, China
| | - Tian Yan
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou 310053, China
| | - Linfeng Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou 310053, China
| | - Wenhua Yu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou 310006, China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou 310006, China
| | - Wei Hu
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou 310006, China
| | - Yongke Zheng
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou 310006, China
| | - Keyi Wang
- Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou 310006, China
| | - Xiaoqiao Dong
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou 310006, China
| | - Dingbo Yang
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261 Huansha Road, Hangzhou 310006, China.
| |
Collapse
|
4
|
Villarreal-Reyna G, Garza-Morales R, Soto-Domínguez A, Montañez-Guerrero L, Saucedo-Cárdenas O, Gómez-Flores M, Ocampo-Garza JA, Pérez-Trujillo JJ, Montes-de-Oca-Luna R. Cerebrolysin induces hair repigmentation associated to MART-1/Melan-A reactivation. Eur J Med Res 2022; 27:257. [PMID: 36411485 PMCID: PMC9677656 DOI: 10.1186/s40001-022-00889-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 11/06/2022] [Indexed: 11/22/2022] Open
Abstract
Hair graying, a prototypical sign of human aging, is a progressive loss of pigmentation from growing hair shafts caused by disease and as a side effect of medications. Cerebrolysin is a neuropeptide preparation that mimics the effect of endogenous neurotrophic factors. Cerebrolysin has been widely used in neurologic conditions, such as cerebral stroke, Alzheimer's disease, and dementia, among others. Cerebrolysin treatment has achieved to regain or maintain the cognitive ability of affected patients; however, up to date, there are no reports about the reactivation of hair pigmentation. We describe a previously not described effect occurring on patients receiving Cerebrolysin treatment for neurologic diseases and whether this effect is associated in reactivation of melanocytes and melanin expression. Here, we report five patients (mean age, 70.6 years), who also had age-related hair graying and scalp hair repigmentation during Cerebrolysin treatment. Macroscopic analysis revealed hair repigmentation consisted in diffuse darkening of the scalp hair. Impregnation and immunostaining analysis were performed on scalp biopsies taken before and after Cerebrolysin treatment; the results showed greater melanin and melanocyte marker MART-1/Melan-A staining following Cerebrolysin treatment. We present, to our knowledge, the first report on hair repigmentation is a previously not described effect occurring following Cerebrolysin treatment.
Collapse
Affiliation(s)
| | - Rodolfo Garza-Morales
- grid.411455.00000 0001 2203 0321Department of Histology, School of Medicine, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Nuevo Leon Mexico
| | - Adolfo Soto-Domínguez
- grid.411455.00000 0001 2203 0321Department of Histology, School of Medicine, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Nuevo Leon Mexico
| | | | - Odila Saucedo-Cárdenas
- grid.411455.00000 0001 2203 0321Department of Histology, School of Medicine, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Nuevo Leon Mexico
| | - Minerva Gómez-Flores
- grid.464574.00000 0004 1760 058XService of Dermatology, Hospital Universitario Jose Eleuterio Gonzalez, 64460 Monterrey, Nuevo Leon Mexico
| | - Jorge Alejandro Ocampo-Garza
- grid.464574.00000 0004 1760 058XService of Dermatology, Hospital Universitario Jose Eleuterio Gonzalez, 64460 Monterrey, Nuevo Leon Mexico
| | - José Juan Pérez-Trujillo
- grid.411455.00000 0001 2203 0321Department of Histology, School of Medicine, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Nuevo Leon Mexico
| | - Roberto Montes-de-Oca-Luna
- grid.411455.00000 0001 2203 0321Department of Histology, School of Medicine, Universidad Autonoma de Nuevo Leon, 64460 Monterrey, Nuevo Leon Mexico
| |
Collapse
|
5
|
Neuropeptide α-Melanocyte-Stimulating Hormone Promotes Neurological Recovery and Repairs Cerebral Ischemia/Reperfusion Injury in Type 1 Diabetes. Neurochem Res 2021; 47:394-408. [PMID: 34586586 DOI: 10.1007/s11064-021-03453-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022]
Abstract
Persons with type 1 diabetes have an increased risk of stroke compared with the general population. α-Melanocyte-stimulating hormone (α-MSH) is a neuropeptide that has protective effects against ischemia/reperfusion (I/R) induced organ damages. In this study, we aimed to investigate the neuroprotective role of this peptide on I/R induced brain damage after experimental stroke associated with hyperglycemia using C57BL/6J Ins2Akita/+ mice. Experimental stroke was induced by blocking the right middle cerebral artery for 2 h with reperfusion for 2 and 22 h, respectively using the intraluminal method. Animals were treated intraperitoneally with or without α-MSH at 1 h after ischemia and 1 h after reperfusion. Significantly higher survival rate and lower neurological scores were recorded in animals injected with α-MSH. Similarly, neuron death, glial cells activation as well as oxidative and nitrosative stress were significantly decreased in α-MSH treated group. Relative intensities of matrix metallopeptidases 9, cyclooxygenase 2 and nuclear factor-κB were significantly decreased while intensities of Akt, heme oxygenase (HO) 1, HO-2 and B-cell lymphoma 2 were significantly increased after α-MSH treatment. In addition, gene expressions of monocarboxylate transporter (MCT) 1, MCT-2 and activity-regulated cytoskeleton-associated protein were significantly higher in brain samples treated with α-MSH, suggesting this peptide may have role in neuron survival by an involvement of lactate metabolism. In conclusion, α-MSH is neuroprotective under hyperglycemic condition against I/R induced brain damage by its anti-inflammatory, anti-oxidative and anti-apoptotic properties. The use of α-MSH analogues may be potential therapeutic agents for diabetic stroke.
Collapse
|
6
|
Zheng Y, Wu S, Yang Q, Xu Z, Zhang S, Fan S, Liu C, Li X, Ma C. Trigeminal nerve electrical stimulation: An effective arousal treatment for loss of consciousness. Brain Res Bull 2021; 169:81-93. [PMID: 33453332 DOI: 10.1016/j.brainresbull.2021.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/16/2020] [Accepted: 01/08/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND To determine if trigeminal nerve electrical stimulation (TNS) would be an effective arousal treatment for loss of consciousness (LOC), we applied neuroscientific methods to investigate the role of potential brain circuit and neuropeptide pathway in regulating level of consciousness. METHODS Consciousness behavioral analysis, Electroencephalogram (EEG) recording, Chemogenetics, Microarray analysis, Milliplex MAP rat peptide assay, Chromatin immune-precipitation (ChIP), Dual-luciferase reporter experiment, Western blot, PCR and Fluorescence in situ hybridization (FISH). RESULTS TNS can markedly activate the neuronal activities of the lateral hypothalamus (LH) and the spinal trigeminal nucleus (Sp5), as well as improve rat consciousness level and EEG activities. Then we proved that LH activation and upregulated neuropeptide hypocretin are beneficial for promotion of consciousness recovery. We then applied gene microarray experiment and found hypocretin might be mediated by a well-known transcription factor Early growth response gene 1 (EGR1), and the results were confirmed by ChIP and Dual-luciferase reporter experiment. CONCLUSION This study illustrates that TNS is an effective arousal strategy Treatment for LOC state via the activation of Sp5 and LH neurons and upregulation of hypocretin expression.
Collapse
Affiliation(s)
- Yaochao Zheng
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510030, China
| | - Shaoling Wu
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510030, China
| | - Qian Yang
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510030, China
| | - Zhen Xu
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510030, China
| | - Subo Zhang
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510030, China
| | - Shengnuo Fan
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510030, China
| | - Cuicui Liu
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510030, China
| | - Xiao Li
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510030, China
| | - Chao Ma
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510030, China.
| |
Collapse
|
7
|
Sharma A, Muresanu DF, Sahib S, Tian ZR, Castellani RJ, Nozari A, Lafuente JV, Buzoianu AD, Bryukhovetskiy I, Manzhulo I, Patnaik R, Wiklund L, Sharma HS. Concussive head injury exacerbates neuropathology of sleep deprivation: Superior neuroprotection by co-administration of TiO 2-nanowired cerebrolysin, alpha-melanocyte-stimulating hormone, and mesenchymal stem cells. PROGRESS IN BRAIN RESEARCH 2020; 258:1-77. [PMID: 33223033 DOI: 10.1016/bs.pbr.2020.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sleep deprivation (SD) is common in military personnel engaged in combat operations leading to brain dysfunction. Military personnel during acute or chronic SD often prone to traumatic brain injury (TBI) indicating the possibility of further exacerbating brain pathology. Several lines of evidence suggest that in both TBI and SD alpha-melanocyte-stimulating hormone (α-MSH) and brain-derived neurotrophic factor (BDNF) levels decreases in plasma and brain. Thus, a possibility exists that exogenous supplement of α-MSH and/or BDNF induces neuroprotection in SD compounded with TBI. In addition, mesenchymal stem cells (MSCs) are very portent in inducing neuroprotection in TBI. We examined the effects of concussive head injury (CHI) in SD on brain pathology. Furthermore, possible neuroprotective effects of α-MSH, MSCs and neurotrophic factors treatment were explored in a rat model of SD and CHI. Rats subjected to 48h SD with CHI exhibited higher leakage of BBB to Evans blue and radioiodine compared to identical SD or CHI alone. Brain pathology was also exacerbated in SD with CHI group as compared to SD or CHI alone together with a significant reduction in α-MSH and BDNF levels in plasma and brain and enhanced level of tumor necrosis factor-alpha (TNF-α). Exogenous administration of α-MSH (250μg/kg) together with MSCs (1×106) and cerebrolysin (a balanced composition of several neurotrophic factors and active peptide fragments) (5mL/kg) significantly induced neuroprotection in SD with CHI. Interestingly, TiO2 nanowired delivery of α-MSH (100μg), MSCs, and cerebrolysin (2.5mL/kg) induced enhanced neuroprotection with higher levels of α-MSH and BDNF and decreased the TNF-α in SD with CHI. These observations are the first to show that TiO2 nanowired administration of α-MSH, MSCs and cerebrolysin induces superior neuroprotection following SD in CHI, not reported earlier. The clinical significance of our findings in light of the current literature is discussed.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
8
|
Sharma A, Muresanu DF, Ozkizilcik A, Tian ZR, Lafuente JV, Manzhulo I, Mössler H, Sharma HS. Sleep deprivation exacerbates concussive head injury induced brain pathology: Neuroprotective effects of nanowired delivery of cerebrolysin with α-melanocyte-stimulating hormone. PROGRESS IN BRAIN RESEARCH 2019; 245:1-55. [DOI: 10.1016/bs.pbr.2019.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
9
|
Feng G, Feng J, Zhang S, Tong Y, Zhang Q, Yang X, Zhang H. Altered levels of α-melanocyte stimulating hormone in cerebrospinal fluid and plasma of patients with traumatic brain injury. Brain Res 2018; 1696:22-30. [PMID: 29859146 DOI: 10.1016/j.brainres.2018.05.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/07/2018] [Accepted: 05/27/2018] [Indexed: 10/16/2022]
Abstract
Traumatic brain injury (TBI) is closely associated with marked inflammation. Although alpha-Melanocyte-Stimulating Hormone (α-MSH) exerts powerful anti-inflammatory effects, changes in endogenous α-MSH levels following TBI remain poorly understood. We investigated the changes of α-MSH levels in the cerebrospinal fluid (CSF) and plasma of post-TBI patients and the association of these changes with the severity of TBI and inflammation. TBI severity was assessed by the GCS coma scale from which, patients were separated into three groups. Clinical data were collected on days 1, 3, 5, and 7 including levels of α-MSH, tumor necrosis factor (TNF-α), and intracranial pressure (ICP). α-MSH levels in CSF steadily increased for one week (peak at day 5) but plasma α-MSH decreased and remained low. These changes were more substantial in the Severe Group of TBI with lower GCS. TNF-α levels were similarly increased in both CSF and plasma (peak at day 3). In the early phase of TBI elevated TNF-α and ICP dominated, and CSF α-MSH displayed a slow and insufficient increase. In later phases of TBI, TNF-α and ICP levels were alleviated concordantly with sustained increases in central α-MSH, wherein an anti-inflammatory environment might predominate. The relationship between plasma α-MSH and TNF-α showed significant negative correlation, and the relationship between CSF α-MSH and TNF-α showed significant positive correlation with a two-day lag. In conclusion, plasma α-MSH levels decreased, but CSF levels increased slowly following TBI. These changes were more substantial in severe patients with a lower GCS. Increases in central α-MSH paralleled alleviation of inflammation.
Collapse
Affiliation(s)
- Guilong Feng
- Department of Emergency, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Jie Feng
- Department of Emergency, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shaojun Zhang
- Department of Endocrinology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Yuping Tong
- Department of Emergency, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qiang Zhang
- Department of Emergency, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoming Yang
- Department of Emergency, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huaping Zhang
- Molecular Biology Laboratories, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
10
|
Harazin A, Bocsik A, Barna L, Kincses A, Váradi J, Fenyvesi F, Tubak V, Deli MA, Vecsernyés M. Protection of cultured brain endothelial cells from cytokine-induced damage by α-melanocyte stimulating hormone. PeerJ 2018; 6:e4774. [PMID: 29780671 PMCID: PMC5958884 DOI: 10.7717/peerj.4774] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022] Open
Abstract
The blood–brain barrier (BBB), an interface between the systemic circulation and the nervous system, can be a target of cytokines in inflammatory conditions. Pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) induce damage in brain endothelial cells and BBB dysfunction which contribute to neuronal injury. The neuroprotective effects of α-melanocyte stimulating hormone (α-MSH) were investigated in experimental models, but there are no data related to the BBB. Based on our recent study, in which α-MSH reduced barrier dysfunction in human intestinal epithelial cells induced by TNF-α and IL-1β, we hypothesized a protective effect of α-MSH on brain endothelial cells. We examined the effect of these two pro-inflammatory cytokines, and the neuropeptide α-MSH on a culture model of the BBB, primary rat brain endothelial cells co-cultured with rat brain pericytes and glial cells. We demonstrated the expression of melanocortin-1 receptor in isolated rat brain microvessels and cultured brain endothelial cells by RT-PCR and immunohistochemistry. TNF-α and IL-1β induced cell damage, measured by impedance and MTT assay, which was attenuated by α-MSH (1 and 10 pM). The peptide inhibited the cytokine-induced increase in brain endothelial permeability, and restored the morphological changes in cellular junctions visualized by immunostaining for claudin-5 and β-catenin. Elevated production of reactive oxygen species and the nuclear translocation of NF-κB were also reduced by α-MSH in brain endothelial cells stimulated by cytokines. We demonstrated for the first time the direct beneficial effect of α-MSH on cultured brain endothelial cells, indicating that this neurohormone may be protective at the BBB.
Collapse
Affiliation(s)
- András Harazin
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Alexandra Bocsik
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Lilla Barna
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - András Kincses
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Judit Váradi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Ferenc Fenyvesi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | | | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Miklós Vecsernyés
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
11
|
Du X, Dong B, Li C, Zhang F, Ji Y, Zhang J, Yin C. Dynamic changes of α-melanocyte-stimulating hormone levels in the serum of patients with craniocerebral trauma. Exp Ther Med 2017; 14:2511-2516. [PMID: 28962188 PMCID: PMC5609151 DOI: 10.3892/etm.2017.4793] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/04/2017] [Indexed: 12/20/2022] Open
Abstract
The aim of the study was to investigate dynamic changes in α-melanocyte-stimulating hormone (α-MSH) levels in the serum of patients with craniocerebral trauma. Forty-eight patients with acute craniocerebral injury were selected between January 2015 and October 2016. The patients were divided into three groups: severe (18 cases), moderate (16 cases) and mild (14 cases), according to the Glasgow Coma Scale (GCS) score at the time of admission. At the same time, 10 adults with a similar age distribution to the patients were also selected as a control group. Venous blood was extracted from patients at 1, 3, 5 and 7 days after injury. Serum α-MSH and tumor necrosis factor (TNF)-α levels were measured using an enzyme-linked immunosorbent assay (ELISA). The correlation between α-MSH and TNF-α was analyzed using Pearson's correlation analysis. Serum α-MSH levels in patients with craniocerebral injury were lower than those in the healthy control group (P<0.05). Decreased serum α-MSH levels were usually accompanied with higher degrees of craniocerebral injury. Serum α-MSH levels initially decreased and then later increased, with the lowest α-MSH levels in the mild at 5 days, moderate at 5 days, and severe groups at 3 days after injury (P<0.05). Serum TNF-α levels in all the patient groups were higher than those in the control group at different time points after injury, with higher TNF-α serum levels accompanying higher degrees of brain injury. In all three groups, serum TNF-α levels initially increased and then decreased post-injury, with peak serum TNF-α levels found at 3-day post-injury in all the patient groups (P<0.05). A negative correlation between serum α-MSH content and serum TNF-α levels in patients with craniocerebral trauma at different time points, was noted (P<0.05). Serum α-MSH content in the survival group was higher than that in the death group (P<0.05). Serum α-MSH levels in patients with non-systemic inflammatory response syndrome (SIRS) were higher than in patients with SIRS (P<0.05). Serum α-MSH levels during the early stages after craniocerebral trauma can be used as a factor for the prediction of secondary SIRS, with constant low levels of serum α-MSH suggest poor prognosis.
Collapse
Affiliation(s)
- Xiaoguang Du
- Department of Neurosurgery, Shandong Jiaotong Hospital, Jinan, Shandong 250031, P.R. China
| | - Baozhong Dong
- Department of Neurosurgery, Shandong Jiaotong Hospital, Jinan, Shandong 250031, P.R. China
| | - Chen Li
- Department of Neurosurgery, Shandong Jiaotong Hospital, Jinan, Shandong 250031, P.R. China
| | - Faxue Zhang
- Department of Neurosurgery, Shandong Jiaotong Hospital, Jinan, Shandong 250031, P.R. China
| | - Yanwei Ji
- Department of Neurosurgery, Shandong Jiaotong Hospital, Jinan, Shandong 250031, P.R. China
| | - Jianbin Zhang
- Department of Neurosurgery, Shandong Jiaotong Hospital, Jinan, Shandong 250031, P.R. China
| | - Changjiang Yin
- Department of Neurosurgery, Shandong Jiaotong Hospital, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
12
|
Váradi J, Harazin A, Fenyvesi F, Réti-Nagy K, Gogolák P, Vámosi G, Bácskay I, Fehér P, Ujhelyi Z, Vasvári G, Róka E, Haines D, Deli MA, Vecsernyés M. Alpha-Melanocyte Stimulating Hormone Protects against Cytokine-Induced Barrier Damage in Caco-2 Intestinal Epithelial Monolayers. PLoS One 2017; 12:e0170537. [PMID: 28103316 PMCID: PMC5245816 DOI: 10.1371/journal.pone.0170537] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Alpha-melanocyte-stimulating hormone (α-MSH) is a potent anti-inflammatory peptide with cytoprotective effect in various tissues. The present investigation demonstrates the ability of α-MSH to interact with intestinal epithelial cell monolayers and mitigate inflammatory processes of the epithelial barrier. The protective effect of α-MSH was studied on Caco-2 human intestinal epithelial monolayers, which were disrupted by exposure to tumor necrosis factor-α and interleukin-1β. The barrier integrity was assessed by measuring transepithelial electric resistance (TEER) and permeability for marker molecules. Caco-2 monolayers were evaluated by immunohistochemistry for expression of melanocortin-1 receptor and tight junction proteins ZO-1 and claudin-4. The activation of nuclear factor kappa beta (NF-κB) was detected by fluorescence microscopy and inflammatory cytokine expression was assessed by flow cytometric bead array cytokine assay. Exposure of Caco-2 monolayers to proinflammatory cytokines lowered TEER and increased permeability for fluorescein and albumin, which was accompanied by changes in ZO-1 and claudin-4 immunostaining. α-MSH was able to prevent inflammation-associated decrease of TEER in a dose-dependent manner and reduce the increased permeability for paracellular marker fluorescein. Further immunohistochemistry analysis revealed proinflammatory cytokine induced translocation of the NF-κB p65 subunit into Caco-2 cell nuclei, which was inhibited by α-MSH. As a result the IL-6 and IL-8 production of Caco-2 monolayers were also decreased with different patterns by the addition of α-MSH to the culture medium. In conclusion, Caco-2 cells showed a positive immunostaining for melanocortin-1 receptor and α-MSH protected Caco-2 cells against inflammatory barrier dysfunction and inflammatory activation induced by tumor necrosis factor-α and interleukin-1β cytokines.
Collapse
Affiliation(s)
- Judit Váradi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
- * E-mail:
| | - András Harazin
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Ferenc Fenyvesi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Katalin Réti-Nagy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Péter Gogolák
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Vámosi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ildikó Bácskay
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Pálma Fehér
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Zoltán Ujhelyi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Gábor Vasvári
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Eszter Róka
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - David Haines
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Miklós Vecsernyés
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
13
|
Multiple beneficial effects of melanocortin MC 4 receptor agonists in experimental neurodegenerative disorders: Therapeutic perspectives. Prog Neurobiol 2016; 148:40-56. [PMID: 27916623 DOI: 10.1016/j.pneurobio.2016.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022]
Abstract
Melanocortin peptides induce neuroprotection in acute and chronic experimental neurodegenerative conditions. Melanocortins likewise counteract systemic responses to brain injuries. Furthermore, they promote neurogenesis by activating critical signaling pathways. Melanocortin-induced long-lasting improvement in synaptic activity and neurological performance, including learning and memory, sensory-motor orientation and coordinated limb use, has been consistently observed in experimental models of acute and chronic neurodegeneration. Evidence indicates that the neuroprotective and neurogenic effects of melanocortins, as well as the protection against systemic responses to a brain injury, are mediated by brain melanocortin 4 (MC4) receptors, through an involvement of the vagus nerve. Here we discuss the targets and mechanisms underlying the multiple beneficial effects recently observed in animal models of neurodegeneration. We comment on the potential clinical usefulness of melanocortin MC4 receptor agonists as neuroprotective and neuroregenerative agents in ischemic stroke, subarachnoid hemorrhage, traumatic brain injury, spinal cord injury, and Alzheimer's disease.
Collapse
|
14
|
Leone S, Noera G, Bertolini A. Developments and new vistas in the field of melanocortins. Biomol Concepts 2016; 6:361-82. [PMID: 26479351 DOI: 10.1515/bmc-2015-0023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/25/2015] [Indexed: 12/14/2022] Open
Abstract
Melanocortins play a fundamental role in several basic functions of the organism (sexual activity, feeding, inflammation and immune responses, pain sensitivity, response to stressful situations, motivation, attention, learning, and memory). Moreover, a large body of animal data, some of which were also confirmed in humans, unequivocally show that melanocortins also have impressive therapeutic effects in several pathological conditions that are the leading cause of mortality and disability worldwide (hemorrhagic, or anyway hypovolemic, shock; septic shock; respiratory arrest; cardiac arrest; ischemia- and ischemia/reperfusion-induced damage of the brain, heart, intestine, and other organs; traumatic injury of brain, spinal cord, and peripheral nerves; neuropathic pain; toxic neuropathies; gouty arthritis; etc.). Recent data obtained in animal models seem to moreover confirm previous hypotheses and preliminary data concerning the neurotrophic activity of melanocortins in neurodegenerative diseases, in particular Alzheimer's disease. Our aim was (i) to critically reconsider the established extrahormonal effects of melanocortins (on sexual activity, feeding, inflammation, tissue hypoperfusion, and traumatic damage of central and peripheral nervous system) at the light of recent findings, (ii) to review the most recent advancements, particularly on the effects of melanocortins in models of neurodegenerative diseases, (iii) to discuss the reasons that support the introduction into clinical practice of melanocortins as life-saving agents in shock conditions and that suggest to verify in clinical setting the impressive results steadily obtained with melanocortins in different animal models of tissue ischemia and ischemia/reperfusion, and finally, (iv) to mention the advisable developments, particularly in terms of selectivity of action and of effects.
Collapse
|
15
|
Kooijman E, Nijboer CH, van Velthoven CTJ, Kavelaars A, Kesecioglu J, Heijnen CJ. The rodent endovascular puncture model of subarachnoid hemorrhage: mechanisms of brain damage and therapeutic strategies. J Neuroinflammation 2014; 11:2. [PMID: 24386932 PMCID: PMC3892045 DOI: 10.1186/1742-2094-11-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 12/17/2013] [Indexed: 01/05/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) represents a considerable health problem. To date, limited therapeutic options are available. In order to develop effective therapeutic strategies for SAH, the mechanisms involved in SAH brain damage should be fully explored. Here we review the mechanisms of SAH brain damage induced by the experimental endovascular puncture model. We have included a description of similarities and distinctions between experimental SAH in animals and human SAH pathology. Moreover, several novel treatment options to diminish SAH brain damage are discussed.SAH is accompanied by cerebral inflammation as demonstrated by an influx of inflammatory cells into the cerebral parenchyma, upregulation of inflammatory transcriptional pathways and increased expression of cytokines and chemokines. Additionally, various cell death pathways including cerebral apoptosis, necrosis, necroptosis and autophagy are involved in neuronal damage caused by SAH.Treatment strategies aiming at inhibition of inflammatory or cell death pathways demonstrate the importance of these mechanisms for survival after experimental SAH. Moreover, neuroregenerative therapies using stem cells are discussed as a possible strategy to repair the brain after SAH since this therapy may extend the window of treatment considerably. We propose the endovascular puncture model as a suitable animal model which resembles the human pathology of SAH and which could be applied to investigate novel therapeutic therapies to combat this debilitating insult.
Collapse
Affiliation(s)
- Elke Kooijman
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cindy TJ van Velthoven
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annemieke Kavelaars
- Division of Internal Medicine, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jozef Kesecioglu
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cobi J Heijnen
- Division of Internal Medicine, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
16
|
Schaible EV, Steinsträßer A, Jahn-Eimermacher A, Luh C, Sebastiani A, Kornes F, Pieter D, Schäfer MK, Engelhard K, Thal SC. Single administration of tripeptide α-MSH(11-13) attenuates brain damage by reduced inflammation and apoptosis after experimental traumatic brain injury in mice. PLoS One 2013; 8:e71056. [PMID: 23940690 PMCID: PMC3733710 DOI: 10.1371/journal.pone.0071056] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 06/24/2013] [Indexed: 12/29/2022] Open
Abstract
Following traumatic brain injury (TBI) neuroinflammatory processes promote neuronal cell loss. Alpha-melanocyte-stimulating hormone (α-MSH) is a neuropeptide with immunomodulatory properties, which may offer neuroprotection. Due to short half-life and pigmentary side-effects of α-MSH, the C-terminal tripeptide α-MSH(11-13) may be an anti-inflammatory alternative. The present study investigated the mRNA concentrations of the precursor hormone proopiomelanocortin (POMC) and of melanocortin receptors 1 and 4 (MC1R/MC4R) in naive mice and 15 min, 6, 12, 24, and 48 h after controlled cortical impact (CCI). Regulation of POMC and MC4R expression did not change after trauma, while MC1R levels increased over time with a 3-fold maximum at 12 h compared to naive brain tissue. The effect of α-MSH(11-13) on secondary lesion volume determined in cresyl violet stained sections (intraperitoneal injection 30 min after insult of 1 mg/kg α-MSH(11-13) or 0.9% NaCl) showed a considerable smaller trauma in α-MSH(11-13) injected mice. The expression of the inflammatory markers TNF-α and IL-1β as well as the total amount of Iba-1 positive cells were not reduced. However, cell branch counting of Iba-1 positive cells revealed a reduced activation of microglia. Furthermore, tripeptide injection reduced neuronal apoptosis analyzed by cleaved caspase-3 and NeuN staining. Based on the results single α-MSH(11-13) administration offers a promising neuroprotective property by modulation of inflammation and prevention of apoptosis after traumatic brain injury.
Collapse
Affiliation(s)
- Eva-Verena Schaible
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Arne Steinsträßer
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Antje Jahn-Eimermacher
- Institute of Medical Biostatistics, Epidemiology and Informatics, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Clara Luh
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anne Sebastiani
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Frida Kornes
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Dana Pieter
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael K. Schäfer
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kristin Engelhard
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Serge C. Thal
- Department of Anesthesiology, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- * E-mail:
| |
Collapse
|
17
|
Giuliani D, Minutoli L, Ottani A, Spaccapelo L, Bitto A, Galantucci M, Altavilla D, Squadrito F, Guarini S. Melanocortins as potential therapeutic agents in severe hypoxic conditions. Front Neuroendocrinol 2012; 33:179-93. [PMID: 22531139 DOI: 10.1016/j.yfrne.2012.04.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 03/26/2012] [Accepted: 04/09/2012] [Indexed: 01/14/2023]
Abstract
Melanocortin peptides with the adrenocorticotropin/melanocyte-stimulating hormone (ACTH/MSH) sequences and synthetic analogs have protective and life-saving effects in experimental conditions of circulatory shock, myocardial ischemia, ischemic stroke, traumatic brain injury, respiratory arrest, renal ischemia, intestinal ischemia and testicular ischemia, as well as in experimental heart transplantation. Moreover, melanocortins improve functional recovery and stimulate neurogenesis in experimental models of cerebral ischemia. These beneficial effects of ACTH/MSH-like peptides are mostly mediated by brain melanocortin MC(3)/MC(4) receptors, whose activation triggers protective pathways that counteract the main ischemia/reperfusion-related mechanisms of damage. Induction of signaling pathways and other molecular regulators of neural stem/progenitor cell proliferation, differentiation and integration seems to be the key mechanism of neurogenesis stimulation. Synthesis of stable and highly selective agonists at MC(3) and MC(4) receptors could provide the potential for development of a new class of drugs for a novel approach to management of severe ischemic diseases.
Collapse
Affiliation(s)
- Daniela Giuliani
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gatti S, Lonati C, Acerbi F, Sordi A, Leonardi P, Carlin A, Gaini SM, Catania A. Protective action of NDP-MSH in experimental subarachnoid hemorrhage. Exp Neurol 2012; 234:230-8. [DOI: 10.1016/j.expneurol.2011.12.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 12/10/2011] [Accepted: 12/22/2011] [Indexed: 12/22/2022]
|
19
|
Zierath D, Tanzi P, Cain K, Shibata D, Becker K. Plasma α-melanocyte stimulating hormone predicts outcome in ischemic stroke. Stroke 2011; 42:3415-20. [PMID: 21960572 DOI: 10.1161/strokeaha.111.627331] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE α-Melanocyte stimulating hormone (α-MSH) is an endogenously produced neuropeptide derived from the same precursor as adrenocorticotropic hormone. α-MSH has profound immunomodulatory properties and may also be neuroprotective. Nothing is known about α-MSH and changes in its plasma concentrations in patients with acute ischemic stroke. METHODS In this prospective observational study, plasma concentrations of α-MSH, adrenocorticotropic hormone, cortisol, and interleukin 6 were assessed longitudinally over the course of 1 year after stroke onset in 111 patients. Logistic regression was used to the effect of initial plasma α-MSH, adrenocorticotropic hormone, cortisol, and interleukin 6 on long-term outcome. RESULTS There was an early decrease in plasma α-MSH in patients with severe stroke (National Institutes of Health Stroke Scale≥17) that normalized over the course of the year; these same patients evidenced elevations in plasma cortisol and interleukin 6. Higher initial plasma α-MSH, but not adrenocorticotropic hormone, cortisol, or interleukin 6, was independently predictive of good long-term outcome. CONCLUSIONS This research is the first to study endogenous changes in plasma α-MSH after stroke. The independent effect of early plasma α-MSH on stroke outcome, as well as a growing body of experimental data demonstrating improved stroke outcome with exogenous α-MSH administration, suggests a potential therapeutic role for α-MSH in the treatment of stroke.
Collapse
Affiliation(s)
- Dannielle Zierath
- Department of Neurology, University of Washington School of Medicine, Box 359775, Harborview Medical Center, 325 9th Ave, Seattle, WA 98104-2499, USA
| | | | | | | | | |
Collapse
|
20
|
Spaccapelo L, Bitto A, Galantucci M, Ottani A, Irrera N, Minutoli L, Altavilla D, Novellino E, Grieco P, Zaffe D, Squadrito F, Giuliani D, Guarini S. Melanocortin MC₄ receptor agonists counteract late inflammatory and apoptotic responses and improve neuronal functionality after cerebral ischemia. Eur J Pharmacol 2011; 670:479-86. [PMID: 21946115 DOI: 10.1016/j.ejphar.2011.09.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 08/29/2011] [Accepted: 09/07/2011] [Indexed: 01/30/2023]
Abstract
Indirect evidence indicates that, in cerebral ischemia, melanocortins have neuroprotective effects likely mediated by MC₄ receptors. To gain direct insight into the role of melanocortin MC₄ receptors in ischemic stroke, we investigated the effects of a highly selective MC₄ receptor agonist. Gerbils were subjected to transient global cerebral ischemia by occluding both common carotid arteries for 10 min. In saline-treated stroke animals, an impairment in learning and memory occurred that, at day 11 after stroke, was associated with hippocampus up-regulation of tumor necrosis factor-α (TNF-α), BAX, activated extracellular signal-regulated kinases (ERK1/2), c-jun N-terminal kinases (JNK1/2) and caspase-3, down-regulation of Bcl-2, and neuronal loss. Treatment for 11days with the selective melanocortin MC₄ receptor agonist RO27-3225, as well as with the well known non-selective [Nle⁴,D-Phe⁷]α-melanocyte-stimulating hormone (NDP-α-MSH) as a reference non-selective melanocortin, counteracted the inflammatory and apoptotic responses, as indicated by the changes in TNF-α, BAX, ERK1/2, JNK1/2, caspase-3 and Bcl-2 protein expression. Furthermore, melanocortin treatment reduced neuronal loss and dose-dependently improved learning and memory. These positive effects were associated with overexpression of Zif268, an immediate early gene involved in injury repair, synaptic plasticity and memory formation. Pharmacological blockade of MC₄ receptors with the selective MC₄ receptor antagonist HS024 prevented all effects of RO27-3225 and NDP-α-MSH. These data give direct evidence that stimulation of MC₄ receptors affords neuroprotection and promotes functional recovery from stroke, by counteracting prolonged and/or recurrent inflammatory and apoptotic responses, and likely by triggering brain repair pathways.
Collapse
Affiliation(s)
- Luca Spaccapelo
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia, Via G. Campi 287, 41125, Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bitto A, Polito F, Altavilla D, Irrera N, Giuliani D, Ottani A, Minutoli L, Spaccapelo L, Galantucci M, Lodi R, Guzzo G, Guarini S, Squadrito F. Melanocortins protect against multiple organ dysfunction syndrome in mice. Br J Pharmacol 2011; 162:917-28. [PMID: 21039420 PMCID: PMC3042202 DOI: 10.1111/j.1476-5381.2010.01098.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 09/17/2010] [Accepted: 10/09/2010] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Melanocortins reverse circulatory shock and improve survival by counteracting the systemic inflammatory response, and through the activation of the vagus nerve-mediated cholinergic anti-inflammatory pathway. To gain insight into the potential therapeutic value of melanocortins against multiple organ damage following systemic inflammatory response, here we investigated the effects of the melanocortin analogue [Nle⁴ D-Phe⁷]α-MSH (NDP-α-MSH) in a widely used murine model of multiple organ dysfunction syndrome (MODS). EXPERIMENTAL APPROACH MODS was induced in mice by a single intraperitoneal injection of lipopolysaccharide followed, 6 days later (= day 0), by zymosan. After MODS or sham MODS induction, animals were randomized to receive intraperitoneally NDP-α-MSH (340 µg·kg⁻¹ day) or saline for up to 16 days. Additional groups of MODS mice were concomitantly treated with the melanocortin MC₄ receptor antagonist HS024, or the nicotinic acetylcholine receptor antagonist chlorisondamine, and NDP-α-MSH. KEY RESULTS At day 7, in the liver and lung NDP-α-MSH, significantly reduced mRNA expression of tumour necrosis factor-α (TNF-α), increased mRNA expression of interleukin-10 and improved the histological picture, as well as reduced TNF-α plasma levels; furthermore, NDP-α-MSH dose-dependently increased survival rate, as assessed throughout the 16 day observation period. HS024 and chlorisondamine prevented all the beneficial effects of NDP-α-MSH in MODS mice. CONCLUSIONS AND IMPLICATIONS These data indicate that NDP-α-MSH protects against experimental MODS by counteracting the systemic inflammatory response, probably through brain MC₄ receptor-triggered activation of the cholinergic anti-inflammatory pathway. These findings reveal previously undescribed effects of melanocortins and could have clinical relevance in the MODS setting.
Collapse
Affiliation(s)
- Alessandra Bitto
- Department of Clinical and Experimental Medicine and Pharmacology, Section of Pharmacology, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Savos AV, Gee JM, Zierath D, Becker KJ. α-MSH: a potential neuroprotective and immunomodulatory agent for the treatment of stroke. J Cereb Blood Flow Metab 2011; 31:606-13. [PMID: 20700130 PMCID: PMC3049515 DOI: 10.1038/jcbfm.2010.130] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Alpha-melanocyte-stimulating hormone (MSH) is a neuropeptide with profound immunomodulatory properties; we evaluated the effects of α-MSH on stroke outcome and its ability to modulate the postischemic immune response. In Lewis rats subjected to 3 hours of middle cerebral artery occlusion (MCAO), plasma concentrations of α-MSH rapidly decreased and returned to baseline over the course of days. Exogenous administration of α-MSH (100 or 500 μg/kg) improved 24 hour outcome in animals subjected to 2 hours MCAO; α-MSH 500 μg/kg also decreased infarct volume at this time point. Both doses of α-MSH were ineffective in improving outcome or decreasing infarct volume in animals subjected to 3 hours MCAO. The splenocyte response to phytohemagglutin in animals treated with α-MSH was attenuated at 24 hours after MCAO. At 1 month after MCAO, treatment with α-MSH 500 μg/kg at the time of stoke was associated with a decrease in TH1 response to myelin basic protein (MBP) in animals subjected to 2 hours MCAO, although treatment was not associated with improved outcome at this time point. Given the early benefits of α-MSH treatment and its effect on immunologic outcome, further studies to evaluate the utility of α-MSH for the treatment of cerebral ischemia are warranted.
Collapse
Affiliation(s)
- Anna V Savos
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | | |
Collapse
|
23
|
Defining the neurotoxin derived illness chronic ciguatera using markers of chronic systemic inflammatory disturbances: A case/control study. Neurotoxicol Teratol 2010; 32:633-9. [DOI: 10.1016/j.ntt.2010.05.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 05/25/2010] [Accepted: 05/26/2010] [Indexed: 12/31/2022]
|
24
|
Szmydynger-Chodobska J, Strazielle N, Zink BJ, Ghersi-Egea JF, Chodobski A. The role of the choroid plexus in neutrophil invasion after traumatic brain injury. J Cereb Blood Flow Metab 2009; 29:1503-16. [PMID: 19471279 PMCID: PMC2736364 DOI: 10.1038/jcbfm.2009.71] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Traumatic brain injury (TBI) frequently results in neuroinflammation, which includes the invasion of neutrophils. After TBI, neutrophils infiltrate the choroid plexus (CP), a site of the blood-cerebrospinal fluid (CSF) barrier (BCSFB), and accumulate in the CSF space near the injury, from where these inflammatory cells may migrate to brain parenchyma. We have hypothesized that the CP functions as an entry point for neutrophils to invade the injured brain. Using the controlled cortical impact model of TBI in rats and an in vitro model of the BCSFB, we show that the CP produces CXC chemokines, such as cytokine-induced neutrophil chemoattractant (CINC)-1 or CXCL1, CINC-2alpha or CXCL3, and CINC-3 or CXCL2. These chemokines are secreted both apically and basolaterally from the choroidal epithelium, a prerequisite for neutrophil migration across epithelial barriers. Consistent with these findings, we also provide electron microscopic evidence that neutrophils infiltrate the choroidal stroma and subsequently reach the intercellular space between choroidal epithelial cells. This is the first detailed analysis of the BCSFB function related to neutrophil trafficking. Our observations support the role of this barrier in posttraumatic neutrophil invasion.
Collapse
Affiliation(s)
- Joanna Szmydynger-Chodobska
- Department of Emergency Medicine, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA.
| | | | | | | | | |
Collapse
|
25
|
Abstract
BACKGROUND Alpha-melanocyte stimulating hormone (alpha-MSH) is a neuropeptide which modulates inflammation. Prior studies have documented decreased alpha-MSH concentrations in patients with acute traumatic brain injury and subarachnoid hemorrhage. We hypothesized that alpha-MSH levels would be decreased in critically injured patients and that this would correlate with poor outcome. METHODS We performed a retrospective review of prospectively collected data more than 12 months ending December 2005. alpha-MSH concentrations were measured in major torso trauma patients (excluding severe head injuries) who underwent standardized shock resuscitation. alpha-MSH concentrations were measured every 4 hours for the first 24 hours of intensive care unit admission and daily thereafter for hospital days 2 to 5. Controls were similarly aged, healthy volunteers. Outcomes measured included lengths of stay, infectious morbidity, and the incidence of multiple organ failure (MOF) and mortality. RESULTS Fifty-one trauma patients were studied with a median age of 33 (22-54) years. Seventy-five percent were male and 82% sustained blunt trauma. The median Injury Severity Score was 25 (16-34). Eighteen percent of the patients developed MOF, 18% died, and 24% developed MOF and died. The mean initial (first value on the first day) alpha-MSH concentration was significantly lower than in controls (15.9 pg/mL +/- 7.6 pg/mL vs. 26.1 pg/mL +/- 7.4 pg/mL, p = 0.0008) and did not change significantly during the 5-day study period. On univariate and adjusted multivariate analyses, initial alpha-MSH concentrations did not predict either MOF or mortality. CONCLUSIONS The current study is the first to document significantly decreased alpha-MSH concentrations in critically injured trauma patients as compared with controls. Furthermore, alpha-MSH concentrations remained so throughout the study period.
Collapse
|
26
|
Reductions in qEEG slowing over 1 year and after treatment with Cerebrolysin in patients with moderate-severe traumatic brain injury. J Neural Transm (Vienna) 2008; 115:683-92. [PMID: 18273537 DOI: 10.1007/s00702-008-0024-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2007] [Accepted: 01/18/2008] [Indexed: 10/22/2022]
Abstract
Changes in quantitative EEG (qEEG) recordings over a 1-year period and the effects of Cerebrolysin (Cere) on qEEG slowing and cognitive performance were investigated in postacute moderate-severe traumatic brain injury (TBI) patients. Time-related changes in qEEG activity frequency bands (increases of alpha and beta, and reductions of theta and delta relative power) and in qEEG slowing (reduction of EEG power ratio) were statistically significant in patients with a disease progress of less than 2 years at baseline, but not in those patients having a longer disease progress time. Slowing of qEEG activity was also found to be significantly reduced in TBI patients after 1 month of treatment with Cere and 3 months later. Therefore, Cere seems to accelerate the time-related reduction of qEEG slowing occurring in untreated patients. The decrease of qEEG slowing induced by Cere correlated with the improvement of attention and working memory. Results of this exploratory study suggest that Cere might improve the functional recovery after brain injury and encourage the conduction of further controlled clinical trials.
Collapse
|
27
|
Giuliani D, Mioni C, Bazzani C, Zaffe D, Botticelli AR, Capolongo S, Sabba A, Galantucci M, Iannone A, Grieco P, Novellino E, Colombo G, Tomasi A, Catania A, Guarini S. Selective melanocortin MC4 receptor agonists reverse haemorrhagic shock and prevent multiple organ damage. Br J Pharmacol 2007; 150:595-603. [PMID: 17245369 PMCID: PMC2189765 DOI: 10.1038/sj.bjp.0707115] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE In circulatory shock, melanocortins have life-saving effects likely to be mediated by MC4 receptors. To gain direct insight into the role of melanocortin MC4 receptors in haemorrhagic shock, we investigated the effects of two novel selective MC4 receptor agonists. EXPERIMENTAL APPROACH Severe haemorrhagic shock was produced in rats under general anaesthesia. Rats were then treated with either the non-selective agonist [Nle4, D-Phe7]-melanocyte-stimulating hormone (NDP--MSH) or with the selective MC4 agonists RO27-3225 and PG-931. Cardiovascular and respiratory functions were continuously monitored for 2 h; survival rate was recorded up to 24 h. Free radicals in blood were measured using electron spin resonance spectrometry; tissue damage was evaluated histologically 25 min or 24 h after treatment. KEY RESULTS All shocked rats treated with saline died within 30-35 min. Treatment with NDP--MSH, RO27-3225 and PG-931 produced a dose-dependent (13-108 nmol kg-1 i.v.) restoration of cardiovascular and respiratory functions, and improved survival. The three melanocortin agonists also markedly reduced circulating free radicals relative to saline-treated shocked rats. All these effects were prevented by i.p. pretreatment with the selective MC4 receptor antagonist HS024. Moreover, treatment with RO27-3225 prevented morphological and immunocytochemical changes in heart, lung, liver, and kidney, at both early (25 min) and late (24 h) intervals. CONCLUSIONS AND IMPLICATIONS Stimulation of MC4 receptors reversed haemorrhagic shock, reduced multiple organ damage and improved survival. Our findings suggest that selective MC4 receptor agonists could have a protective role against multiple organ failure following circulatory shock.
Collapse
MESH Headings
- Animals
- Blood Pressure/drug effects
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Free Radicals/blood
- Heart Rate/drug effects
- Kidney/drug effects
- Kidney/pathology
- Liver/drug effects
- Liver/pathology
- Lung/drug effects
- Lung/pathology
- Multiple Organ Failure/metabolism
- Multiple Organ Failure/pathology
- Multiple Organ Failure/physiopathology
- Multiple Organ Failure/prevention & control
- Myocardium/pathology
- Peptides, Cyclic/pharmacology
- Peptides, Cyclic/therapeutic use
- Rats
- Rats, Wistar
- Receptor, Melanocortin, Type 4/agonists
- Receptor, Melanocortin, Type 4/metabolism
- Respiratory Mechanics
- Severity of Illness Index
- Shock, Hemorrhagic/drug therapy
- Shock, Hemorrhagic/metabolism
- Shock, Hemorrhagic/pathology
- Shock, Hemorrhagic/physiopathology
- Time Factors
- alpha-MSH/analogs & derivatives
- alpha-MSH/pharmacology
- alpha-MSH/therapeutic use
Collapse
Affiliation(s)
- D Giuliani
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia Modena, Italy
| | - C Mioni
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia Modena, Italy
| | - C Bazzani
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia Modena, Italy
| | - D Zaffe
- Department of Anatomy and Histology, University of Modena and Reggio Emilia Modena, Italy
| | - A R Botticelli
- Department of Human Pathology, University of Pavia Pavia, Italy
| | - S Capolongo
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia Modena, Italy
| | - A Sabba
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia Modena, Italy
| | - M Galantucci
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia Modena, Italy
| | - A Iannone
- Department of Biomedical Sciences, Section of General Pathology, University of Modena and Reggio Emilia Modena, Italy
| | - P Grieco
- Department of Pharmaceutical Chemistry and Toxicology, University of Napoli ‘Federico II' Napoli, Italy
| | - E Novellino
- Department of Pharmaceutical Chemistry and Toxicology, University of Napoli ‘Federico II' Napoli, Italy
| | - G Colombo
- Center for Preclinical Investigation, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena Milano, Italy
| | - A Tomasi
- Department of Diagnostic Services, University of Modena and Reggio Emilia Modena, Italy
| | - A Catania
- Center for Preclinical Investigation, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena Milano, Italy
| | - S Guarini
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia Modena, Italy
- Author for correspondence:
| |
Collapse
|
28
|
Colombo G, Gatti S, Turcatti F, Lonati C, Sordi A, Rossi G, Bonino F, Catania A. Alteration in the Transcriptional Profile of Livers from Brain-dead Organ Donors. Transplantation 2006; 82:69-79. [PMID: 16861944 DOI: 10.1097/01.tp.0000225829.07208.58] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND There is evidence that brain death causes changes in peripheral organs. Marked inflammation is found in organs collected during experimental brain death and clinical studies indicate that, despite genetic mismatch, organs obtained from living donors show improved survival over those from brain-dead donors. The aim of the present clinical research was to explore changes in the transcriptional profile of livers from brain-dead organ donors. METHODS Using the cDNA macroarray technique, we compared gene expression in liver biopsies from 21 brain-dead organ donors and in normal liver tissue obtained during resection of benign focal lesions. RESULTS Analysis of gene expression showed significant differences in the mRNA levels of 117 genes. There was reduced expression of 93 genes whereas expression of 24 genes was enhanced. Downregulated pathways included transcripts related to morphogenesis, blood coagulation, complement cascade, amine metabolism, lipid metabolism, nucleic acid metabolism, biodegradation of xenobiotics, signal transduction, and transcription. Conversely, there was induction of genes related to acute phase response, damage-related response, electron transport, and energy metabolism. CONCLUSIONS The present research demonstrates major changes in the transcriptional profile of livers from brain-dead organ donors. The presence of both down- and upregulated gene families suggests that the alteration in transcriptional profile is not a consequence of death-associated organ failure, but rather, an active change in regulatory mechanisms.
Collapse
Affiliation(s)
- Gualtiero Colombo
- Center for Preclinical Investigation, Fondazione Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Forslin Aronsson S, Spulber S, Popescu LM, Winblad B, Post C, Oprica M, Schultzberg M. alpha-Melanocyte-stimulating hormone is neuroprotective in rat global cerebral ischemia. Neuropeptides 2006; 40:65-75. [PMID: 16414116 DOI: 10.1016/j.npep.2005.10.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Revised: 10/17/2005] [Accepted: 10/19/2005] [Indexed: 02/04/2023]
Abstract
The aim of the study was to investigate the effects of alpha-melanocyte-stimulating hormone (alpha-MSH), a tridecapeptide derived from proopiomelanocortin (POMC), on the neurodegeneration following global cerebral ischemia and reperfusion in the rat. The biological activities of alpha-MSH include inhibition of inflammatory responses and anti-pyretic effects. Male Sprague-Dawley rats were subjected to four-vessel occlusion (4-VO) global cerebral ischemia followed by reperfusion, and treated with alpha-MSH (intraperitoneally, i.p.) at 30 min, and 24, 48, 72 and 96 h post-ischemia. Stereological quantification of the pyramidal cells in the CA1 area of the hippocampus showed that the number of viable neurons in ischemic rats was 96,945+/-18,610 (means+/-SD) as compared to 183,156+/-49,935 in sham-operated rats (P<0.05). The number of viable neurons after treatment of ischemic rats with alpha-MSH was 162,829+/-34,757, i.e. significantly different from the number of viable neurons in ischemic rats injected with saline (P<0.01). Astrocyte proliferation due to the ischemic insult was markedly reduced by the treatment with alpha-MSH, and the loss in body weight was reduced by alpha-MSH. In conclusion, post-ischemic administration of alpha-MSH was found to provide neuroprotection in the CA1 pyramidal cell layer in the hippocampus, concomitant with a reduction in glial activation, indicating that alpha-MSH or mimetics thereof may have a potential in the treatment of stroke or other neurodegenerative diseases. Further studies will be required to define the post-ischemic time window for administration of alpha-MSH.
Collapse
Affiliation(s)
- Sa Forslin Aronsson
- Division of Experimental Geriatrics, Neurotec Department, Karolinska Institutet, Karolinska University Hospital, Novum, KFC, Level 4, SE-141 86 Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
30
|
Gatti S, Colombo G, Turcatti F, Lonati C, Sordi A, Bonino F, Lipton JM, Catania A. Reduced expression of the melanocortin-1 receptor in human liver during brain death. Neuroimmunomodulation 2006; 13:51-5. [PMID: 16864968 DOI: 10.1159/000094513] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Accepted: 06/07/2006] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE There is evidence that brain death has detrimental effects on peripheral organs. Clinical and experimental studies on organ donors showed marked inflammation in tissue samples of livers and kidneys collected during brain death. The inflammatory reaction is characterized by release of cytokines and inflammatory cell infiltration. Because melanocortins and their receptors are significant modulators of inflammation, we hypothesized that downregulation of melanocortin receptors during brain death could contribute to enhance inflammation. METHODS Using real-time polymerase chain reaction (PCR) analysis, we determined expression of melanocortin receptors in liver biopsies obtained from brain-dead organ donors before cold ischemia and in normal liver tissue during resection of benign focal lesions of the liver. Tissue biopsies were also analyzed for expression of intercellular adhesion molecule-1 (ICAM-1), which has a central function in inflammatory cell migration. RESULTS Expression of melanocortin-1 receptor (MC1R) mRNA was markedly reduced in liver samples obtained from brain-dead organ donors compared to hepatic tissue collected during resection of benign focal lesions of the liver. Conversely, expression of the adhesion molecule ICAM-1 was significantly increased in livers of brain-dead organ donors. CONCLUSIONS Disruption of the endogenous anti-inflammatory circuit based on MC1R could contribute to tissue damage during brain death.
Collapse
Affiliation(s)
- Stefano Gatti
- Liver Transplantation Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, Milano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Huleihel M, Golan H, Hallak M. Intrauterine infection/inflammation during pregnancy and offspring brain damages: possible mechanisms involved. Reprod Biol Endocrinol 2004; 2:17. [PMID: 15104793 PMCID: PMC411057 DOI: 10.1186/1477-7827-2-17] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2004] [Accepted: 04/22/2004] [Indexed: 11/17/2022] Open
Abstract
Intrauterine infection is considered as one of the major maternal insults during pregnancy. Intrauterine infection during pregnancy could lead to brain damage of the developmental fetus and offspring. Effects on the fetal, newborn, and adult central nervous system (CNS) may include signs of neurological problems, developmental abnormalities and delays, and intellectual deficits. However, the mechanisms or pathophysiology that leads to permanent brain damage during development are complex and not fully understood. This damage may affect morphogenic and behavioral phenotypes of the developed offspring, and that mice brain damage could be mediated through a final common pathway, which includes over-stimulation of excitatory amino acid receptor, over-production of vascularization/angiogenesis, pro-inflammatory cytokines, neurotrophic factors and apoptotic-inducing factors.
Collapse
Affiliation(s)
- Mahmoud Huleihel
- Department of Microbiology and Immunology and the BGU Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hava Golan
- Department of Microbiology and Immunology and the BGU Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Development and Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Mordechai Hallak
- Department of Microbiology and Immunology and the BGU Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Obstetrics & Gynecology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
32
|
Catania A, Gatti S, Colombo G, Lipton JM. Targeting Melanocortin Receptors as a Novel Strategy to Control Inflammation. Pharmacol Rev 2004; 56:1-29. [PMID: 15001661 DOI: 10.1124/pr.56.1.1] [Citation(s) in RCA: 337] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Adrenocorticotropic hormone and alpha-, beta-, and gamma-melanocyte-stimulating hormones, collectively called melanocortin peptides, exert multiple effects upon the host. These effects range from modulation of fever and inflammation to control of food intake, autonomic functions, and exocrine secretions. Recognition and cloning of five melanocortin receptors (MCRs) has greatly improved understanding of peptide-target cell interactions. Preclinical investigations indicate that activation of certain MCR subtypes, primarily MC1R and MC3R, could be a novel strategy to control inflammatory disorders. As a consequence of reduced translocation of the nuclear factor kappaB to the nucleus, MCR activation causes a collective reduction of the major molecules involved in the inflammatory process. Therefore, anti-inflammatory influences are broad and are not restricted to a specific mediator. Short half-life and lack of selectivity could be an obstacle to the use of the natural melanocortins. However, design and synthesis of new MCR ligands with selective chemical properties are already in progress. This review examines how marshaling MCR could control inflammation.
Collapse
Affiliation(s)
- Anna Catania
- Division of Internal Medicine, Ospedale Maggiore di Milano, Instituto di Ricovero e Cura a Caraterre Scientifico, Milano, Italy.
| | | | | | | |
Collapse
|