1
|
Guan D, Li Y, Chen B, Wang K, Guo Y, Dong N, Cui Y, Gao Y, Wang M, Wang J, Ren Y, Shang P, Liu Y. CBL/Cbl-b mediates Fas degradation to reduce neuronal damage in experimental autoimmune encephalomyelitis. Scand J Immunol 2025; 101:e13430. [PMID: 39726027 DOI: 10.1111/sji.13430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/15/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024]
Abstract
Fas has been shown to positively regulate the differentiation of T helper 17 (Th17) cells in mouse models of experimental autoimmune encephalomyelitis (EAE). Fas protein expression is regulated by ubiquitination but has not been further studied. In this study, we investigated the role of the Fas ubiquitin ligase in Th17 cell differentiation and highlighted its potential as a therapeutic target for EAE. The E3 ubiquitin ligase CBL of Fas was predicted using the online prediction software ubibrowser. Overexpression of CBL significantly reduced Fas protein levels but did not affect mRNA levels. The decrease in Fas protein mediated by CBL overexpression was rescued by the proteasome inhibitor MG132. Co-IP analysis revealed that CBL interacted with Fas. Further results suggested that CBL regulated Fas expression through ubiquitination, thereby affecting Th17 cell differentiation. Cbl-b, a homologue of CBL, significantly promoted the degradation of Fas protein and increased its ubiquitination modification. Furthermore, CBL and Cbl-b could synergistically regulate Fas to influence Th17 cell differentiation. The same conclusion was also reached in animal models. Luxol Fast Blue staining showed that myelinated fibres in the spinal cord were significantly increased after CBL/Cbl-b overexpression in EAE. Finally, flow cytometry and immunofluorescence staining showed that overexpression of CBL or Cbl-b decreased the proportion of Th17 cells in the spinal cord of EAE mice, ultimately reducing neuronal damage. Taken together, these data demonstrate that CBL and Cbl-b can synergistically regulate Fas to inhibit Th17 cell differentiation, thereby alleviating spinal cord nerve injury in the EAE model.
Collapse
Affiliation(s)
- Dongsheng Guan
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yingxia Li
- The College of Basic Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Binyan Chen
- Department of Neurology, Henan Province Hospital of Traditional Chinese Medicine Qiongzhong Branch/Qiongzhong Li and Miao Autonomous County People's Hospital, Qiongzhong, China
| | - Kun Wang
- Department of Pharmacy, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yanke Guo
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ning Dong
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yinglin Cui
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yinghe Gao
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Mengmeng Wang
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Jing Wang
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yihan Ren
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Penghui Shang
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yuxuan Liu
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
2
|
Guan D, Li Y, Zhao X, Wang K, Guo Y, Dong N, Cui Y, Gao Y, Wang M, Wang J, Ren Y, Shang P, Liu Y. Hederagenol improves multiple sclerosis by modulating Th17 cell differentiation. IUBMB Life 2024; 76:845-857. [PMID: 38838376 DOI: 10.1002/iub.2863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024]
Abstract
Multiple sclerosis (MS) is a common autoimmune illness that is difficult to treat. The upregulation of Th17 cells is critical in the pathological process of MS. Hederagenol (Hed) has been shown to lower IL-17 levels, although its role in MS pathophysiology is uncertain. In this study, we explore whether Hed could ameliorate MS by modulating Th17 cell differentiation, with the goal of identifying new treatment targets for MS. The experimental autoimmune encephalomyelitis (EAE) mouse model was conducted and Hed was intraperitoneally injected into mice. The weight was recorded and the clinical symptom grade was assessed. Hematoxylin-eosin staining was carried out to determine the extent of inflammation in the spinal cord and liver. The luxol Fast Blue staining was performed to detect the pathological changes in the myelin sheath. Nerve damage was detected using NeuN immunofluorescence staining and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. Immunohistology approaches were used to study alterations in immune cells in the spinal cord. The proportions of T cell subsets in the spleens were analyzed by flow cytometry. RORγt levels were measured using quantitative real-time PCR or Western blot. The activity of the RORγt promoter was analyzed by Chromatin immunoprecipitation. Hed administration reduced the clinical symptom grade of EAE mice, as well as the inflammatory infiltration, demyelination, and cell disorder of the spinal cord, while having no discernible effect on the mouse weight. In addition, Hed treatment significantly reduced the number of T cells, particularly Th17 cells in the spinal cord and spleen-isolated CD4+ T cells. Hed lowered the RORγt levels in spleens and CD4+ T cells and overexpression of RORγt reversed the inhibitory effect of Hed on Th17 differentiation. Hed decreased nerve injury by modulating Th17 differentiation through the RORγt promoter. Hed regulates Th17 differentiation by reducing RORγt promoter activity, which reduces nerve injury and alleviates EAE.
Collapse
MESH Headings
- Animals
- Th17 Cells/immunology
- Th17 Cells/drug effects
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Cell Differentiation/drug effects
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/pathology
- Multiple Sclerosis/immunology
- Mice
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Female
- Oleanolic Acid/analogs & derivatives
- Oleanolic Acid/pharmacology
- Mice, Inbred C57BL
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Spinal Cord/metabolism
- Spinal Cord/immunology
- Interleukin-17/metabolism
- Interleukin-17/genetics
Collapse
Affiliation(s)
- Dongsheng Guan
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yingxia Li
- The College of Basic Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xu Zhao
- Department of Pharmacy, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Kun Wang
- Department of Pharmacy, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yanke Guo
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ning Dong
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yinglin Cui
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yinghe Gao
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Mengmeng Wang
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Jing Wang
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yihan Ren
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Penghui Shang
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yuxuan Liu
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
3
|
Agarwal N, Fan A, Huang X, Dehkharghani S, van der Kolk A. ISMRM Clinical Focus Meeting 2023: "Imaging the Fire in the Brain". J Magn Reson Imaging 2024. [PMID: 39193867 DOI: 10.1002/jmri.29587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 08/29/2024] Open
Abstract
Set during the Annual Meeting of the International Society for Magnetic Resonance in Medicine (ISMRM), the "Clinical Focus Meeting" (CFM) aims to bridge the gap between innovative magnetic resonance imaging (MRI) scientific research and daily patient care. This initiative is dedicated to maximizing the impact of MRI technology on healthcare outcomes for patients. At the 2023 Annual Meeting, clinicians and scientists from across the globe were invited to discuss neuroinflammation from various angles (entitled "Imaging the Fire in the Brain"). Topics ranged from fundamental mechanisms and biomarkers of neuroinflammation to the role of different contrast mechanisms, including both proton and non-proton techniques, in brain tumors, autoimmune disorders, and pediatric neuroinflammatory diseases. Discussions also delved into how systemic inflammation can trigger neuroinflammation and the role of the gut-brain axis in causing brain inflammation. Neuroinflammation arises from various external and internal factors and serves as a vital mechanism to mitigate tissue damage and provide neuroprotection. Nonetheless, excessive neuroinflammatory responses can lead to significant tissue injury and subsequent neurological impairments. Prolonged neuroinflammation can result in cellular apoptosis and neurodegeneration, posing severe consequences. MRI can be used to visualize these consequences, by detecting blood-brain barrier damage, characterizing brain lesions, quantifying edema, and identifying specific metabolites. It also facilitates monitoring of chronic changes in both the brain and spinal cord over time, potentially leading to better patient outcomes. This paper represents a summary of the 2023 CFM, and is intended to guide the enthusiastic MR user to several key and novel sequences that MRI offers to image pathophysiologic processes underlying acute and chronic neuroinflammation. EVIDENCE LEVEL: 5 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Nivedita Agarwal
- Diagnostic Imaging and Neuroradiology Unit, IRCCS Scientific Institute E. Medea, Bosisio Parini, Lecco, Italy
| | - Audrey Fan
- Department of Neurology, University of California Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Xiaoqi Huang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Seena Dehkharghani
- Department of Radiology, Albert Einstein College of Medicine-Montefiore Health, New York, New York, USA
| | - Anja van der Kolk
- Department of Medical Imaging, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
4
|
Censi ST, Mariani-Costantini R, Granzotto A, Tomassini V, Sensi SL. Endogenous retroviruses in multiple sclerosis: A network-based etiopathogenic model. Ageing Res Rev 2024; 99:102392. [PMID: 38925481 DOI: 10.1016/j.arr.2024.102392] [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: 04/08/2024] [Revised: 06/10/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
The present perspective article proposes an etiopathological model for multiple sclerosis pathogenesis and progression associated with the activation of human endogenous retroviruses. We reviewed preclinical, clinical, epidemiological, and evolutionary evidence indicating how the complex, multi-level interplay of genetic traits and environmental factors contributes to multiple sclerosis. We propose that endogenous retroviruses transactivation acts as a critical node in disease development. We also discuss the rationale for combined anti-retroviral therapy in multiple sclerosis as a disease-modifying therapeutic strategy. Finally, we propose that the immuno-pathogenic process triggered by endogenous retrovirus activation can be extended to aging and aging-related neurodegeneration. In this regard, endogenous retroviruses can be envisioned to act as epigenetic noise, favoring the proliferation of disorganized cellular subpopulations and accelerating system-specific "aging". Since inflammation and aging are two sides of the same coin (plastic dis-adaptation to external stimuli with system-specific degree of freedom), the two conditions may be epiphenomenal products of increased epigenomic entropy. Inflammation accelerates organ-specific aging, disrupting communication throughout critical systems of the body and producing symptoms. Overlapping neurological symptoms and syndromes may emerge from the activity of shared molecular networks that respond to endogenous retroviruses' reactivation.
Collapse
Affiliation(s)
- Stefano T Censi
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara, Italy; Institute for Advanced Biomedical Technologies (ITAB), "G. d'Annunzio" University, Chieti-Pescara, Italy.
| | - Renato Mariani-Costantini
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Alberto Granzotto
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Valentina Tomassini
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara, Italy; Institute for Advanced Biomedical Technologies (ITAB), "G. d'Annunzio" University, Chieti-Pescara, Italy; Multiple Sclerosis Centre, Institute of Neurology, SS Annunziata Hospital, "G. d'Annunzio" University, Chieti, Italy
| | - Stefano L Sensi
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara, Italy; Institute for Advanced Biomedical Technologies (ITAB), "G. d'Annunzio" University, Chieti-Pescara, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti-Pescara, Italy; Multiple Sclerosis Centre, Institute of Neurology, SS Annunziata Hospital, "G. d'Annunzio" University, Chieti, Italy.
| |
Collapse
|
5
|
Ascsillán AA, Kemény LV. The Skin-Brain Axis: From UV and Pigmentation to Behaviour Modulation. Int J Mol Sci 2024; 25:6199. [PMID: 38892387 PMCID: PMC11172643 DOI: 10.3390/ijms25116199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/24/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
The skin-brain axis has been suggested to play a role in several pathophysiological conditions, including opioid addiction, Parkinson's disease and many others. Recent evidence suggests that pathways regulating skin pigmentation may directly and indirectly regulate behaviour. Conversely, CNS-driven neural and hormonal responses have been demonstrated to regulate pigmentation, e.g., under stress. Additionally, due to the shared neuroectodermal origins of the melanocytes and neurons in the CNS, certain CNS diseases may be linked to pigmentation-related changes due to common regulators, e.g., MC1R variations. Furthermore, the HPA analogue of the skin connects skin pigmentation to the endocrine system, thereby allowing the skin to index possible hormonal abnormalities visibly. In this review, insight is provided into skin pigment production and neuromelanin synthesis in the brain and recent findings are summarised on how signalling pathways in the skin, with a particular focus on pigmentation, are interconnected with the central nervous system. Thus, this review may supply a better understanding of the mechanism of several skin-brain associations in health and disease.
Collapse
Affiliation(s)
- Anna A. Ascsillán
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1094 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1094 Budapest, Hungary
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Lajos V. Kemény
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1094 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
6
|
Lu J, Wang J, Ni H, Li B, Yang J, Zhu J, Qian J, Gao R, Xu R. Activation of the melanocortin-1 receptor attenuates neuronal apoptosis after traumatic brain injury by upregulating Merlin expression. Brain Res Bull 2024; 207:110870. [PMID: 38185389 DOI: 10.1016/j.brainresbull.2024.110870] [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: 03/15/2023] [Revised: 12/30/2023] [Accepted: 01/04/2024] [Indexed: 01/09/2024]
Abstract
Traumatic brain injury (TBI) is a common disease worldwide with high mortality and disability rates. Besides the primary mechanical injury, the secondary injury associated with TBI can also induce numerous pathological changes, such as brain edema, nerve apoptosis, and neuroinflammation, which further aggravates neurological dysfunction and even causes the death due to the primary injury. Among them, neuronal apoptosis is a key link in the injury. Melanocortin-1 receptor (MC1R) is a G protein coupled receptor, belonging to the melanocortin receptor family. Studies have shown that activation of MC1R inhibits oxidative stress and apoptosis, and confers neuroprotective effects against various neurological diseases. Merlin is a protein product of the NF2 gene, which is widely expressed in the central nervous system (CNS) of mice, rats, and humans. Studies have indicated that Merlin is associated with MC1R. In this study, we explored the anti-apoptotic effects and potential mechanisms of MC1R. A rat model of TBI was established through controlled cortical impact. The MC1R-specific agonist Nle4-D-Phe7-α-Melanocyte (NDP-MSH) and the inhibitor MSG-606 were employed to explore the effects of MC1R and Merlin following TBI and investigated the associated mechanisms. The results showed that the expression levels of MC1R and Merlin were upregulated after TBI, and activation of MC1R promoted Merlin expression. Further, we found that MC1R activation significantly improved neurological dysfunction and reduced brain edema and neuronal apoptosis induced by TBI in rats. Mechanistically, its neuroprotective function and anti-apoptotic were partly associated with MC1R activation. In conclusion, we demonstrated that MC1R activation after TBI may inhibit apoptosis and confer neuroprotection by upregulating the expression of Merlin.
Collapse
Affiliation(s)
- Jinqi Lu
- Department of Pathology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Jin Wang
- Department of Orthopaedic Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Haibo Ni
- Department of Neurosurgery, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Bing Li
- Department of Neurosurgery, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Jingjing Yang
- Department of Pathology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Jie Zhu
- Department of Pathology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Jie Qian
- Department of Pathology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Rong Gao
- Department of Neurosurgery, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China.
| | - Rong Xu
- Department of Pathology, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
7
|
Singh K, Gupta JK, Kumar S, Soni U. A Review of the Common Neurodegenerative Disorders: Current Therapeutic Approaches and the Potential Role of Bioactive Peptides. Curr Protein Pept Sci 2024; 25:507-526. [PMID: 38561605 DOI: 10.2174/0113892037275221240327042353] [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: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Neurodegenerative disorders, which include Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS), represent a significant and growing global health challenge. Current therapies predominantly focus on symptom management rather than altering disease progression. In this review, we discuss the major therapeutic strategies in practice for these disorders, highlighting their limitations. For AD, the mainstay treatments are cholinesterase inhibitors and N-methyl-D-aspartate (NMDA) receptor antagonists. For PD, dopamine replacement therapies, including levodopa, are commonly used. HD is managed primarily with symptomatic treatments, and reusable extends survival in ALS. However, none of these therapies halts or substantially slows the neurodegenerative process. In contrast, this review highlights emerging research into bioactive peptides as potential therapeutic agents. These naturally occurring or synthetically designed molecules can interact with specific cellular targets, potentially modulating disease processes. Preclinical studies suggest that bioactive peptides may mitigate oxidative stress, inflammation, and protein misfolding, which are common pathological features in neurodegenerative diseases. Clinical trials using bioactive peptides for neurodegeneration are limited but show promising initial results. For instance, hemiacetal, a γ-secretase inhibitor peptide, has shown potential in AD by reducing amyloid-beta production, though its development was discontinued due to side effects. Despite these advancements, many challenges remain, including identifying optimal peptides, confirming their mechanisms of action, and overcoming obstacles related to their delivery to the brain. Future research should prioritize the discovery and development of novel bioactive peptides and improve our understanding of their pharmacokinetics and pharmacodynamics. Ultimately, this approach may lead to more effective therapies for neurodegenerative disorders, moving beyond symptom management to potentially modify the course of these devastating diseases.
Collapse
Affiliation(s)
- Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Jeetendra Kumar Gupta
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Shivendra Kumar
- Department of Pharmacology, Rajiv Academy for Pharmacy, Mathura, Uttar Pradesh, India
| | - Urvashi Soni
- Department of Pharmacology, School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Kothrud, Pune, Maharashtra, India
| |
Collapse
|
8
|
Bisht P, Rathore C, Rathee A, Kabra A. Astrocyte Activation and Drug Target in Pathophysiology of Multiple Sclerosis. Methods Mol Biol 2024; 2761:431-455. [PMID: 38427254 DOI: 10.1007/978-1-0716-3662-6_30] [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: 03/02/2024]
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease, which is also referred to as an autoimmune disorder with chronic inflammatory demyelination affecting the core system that is the central nervous system (CNS). Demyelination is a pathological manifestation of MS. It is the destruction of myelin sheath, which is wrapped around the axons, and it results in the loss of synaptic connections and conduction along the axon is also compromised. Various attempts are made to understand MS and demyelination using various experimental models out of them. The most popular model is experimental autoimmune encephalomyelitis (EAE), in which autoimmunity against CNS components is induced in experimental animals by immunization with self-antigens derived from basic myelin protein. Astrocytes serve as a dual-edged sword both in demyelination and remyelination. Various drug targets have also been discussed that can be further explored for the treatment of MS. An extensive literature research was done from various online scholarly and research articles available on PubMed, Google Scholar, and Elsevier. Keywords used for these articles were astrocyte, demyelination, astrogliosis, and reactive astrocytes. This includes articles being the most relevant information to the area compiled to compose a current review.
Collapse
Affiliation(s)
- Preeti Bisht
- University Institute of Pharma Sciences, Chandigarh University, Ajitgarh, Punjab, India
| | - Charul Rathore
- University Institute of Pharma Sciences, Chandigarh University, Ajitgarh, Punjab, India
| | - Ankit Rathee
- University Institute of Pharma Sciences, Chandigarh University, Ajitgarh, Punjab, India
| | - Atul Kabra
- University Institute of Pharma Sciences, Chandigarh University, Ajitgarh, Punjab, India
| |
Collapse
|
9
|
Srivastava P, Nishiyama S, Zhou F, Lin SH, Srivastava A, Su C, Xu Y, Peng W, Levy M, Schwarzschild M, Chen X. Peripheral MC1R Activation Modulates Immune Responses and is Neuroprotective in a Mouse Model of Parkinson's Disease. J Neuroimmune Pharmacol 2023; 18:704-717. [PMID: 38110615 PMCID: PMC10769915 DOI: 10.1007/s11481-023-10094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 10/17/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Melanocortin 1 receptor (MC1R) is a key pigmentation gene, and loss-of-function of MC1R variants that produce red hair may be associated with Parkinson's disease (PD). We previously reported compromised dopaminergic neuron survival in Mc1r mutant mice and dopaminergic neuroprotective effects of local injection of a MC1R agonist to the brain or a systemically administered MC1R agonist with appreciable central nervous system (CNS) permeability. Beyond melanocytes and dopaminergic neurons, MC1R is expressed in other peripheral tissues and cell types, including immune cells. The present study investigates the impact of NDP-MSH, a synthetic melanocortin receptor (MCR) agonist that does not cross BBB, on the immune system and the nigrostriatal dopaminergic system in mouse model of PD. METHODS C57BL/6 mice were treated systemically with MPTP.HCl (20 mg/kg) and LPS (1 mg/kg) from day 1 to day 4 and NDP-MSH (400 µg/kg) or vehicle from day 1 to day 12 following which the mice were sacrificed. Peripheral and CNS immune cells were phenotyped and inflammatory markers were measured. The nigrostriatal dopaminergic system was assessed behaviorally, chemically, immunologically, and pathologically. To understand the role of regulatory T cells (Tregs) in this model, CD25 monoclonal antibody was used to deplete CD25 + Tregs. RESULTS Systemic NDP-MSH administration significantly attenuated striatal dopamine depletion and nigral dopaminergic neuron loss induced by MPTP + LPS. It improved the behavioral outcomes in the pole test. Mc1r mutant mice injected with NDP-MSH in the MPTP and LPS paradigm showed no changes in striatal dopamine levels suggesting that the NDP-MSH acts through the MC1R pathway. Although no NDP-MSH was detected in the brain, peripheral, NDP-MSH attenuated neuroinflammation as observed by diminished microglial activation in the nigral region, along with reduced TNF-α and IL1β levels in the ventral midbrain. Depletion of Tregs was associated with diminished neuroprotective effects of NDP-MSH. CONCLUSIONS Our study demonstrates that peripherally acting NDP-MSH confers protection on dopaminergic nigrostriatal neurons and reduces hyperactivated microglia. NDP-MSH modulates peripheral immune responses, and Tregs may be involved in the neuroprotective effect of NDP-MSH.
Collapse
Affiliation(s)
- Pranay Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Shuhei Nishiyama
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Fang Zhou
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Sonia H Lin
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Akriti Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Chienwen Su
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Yuehang Xu
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Weiyi Peng
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Michael Levy
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Michael Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
10
|
Chang CL, Cai Z, Hsu SYT. A gel-forming α-MSH analog promotes lasting melanogenesis. Eur J Pharmacol 2023; 958:176008. [PMID: 37673364 DOI: 10.1016/j.ejphar.2023.176008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/08/2023]
Abstract
The α-MSH peptide plays a significant role in the regulation of pigmentation via the melanocortin 1 receptor (MC1R). It increases the DNA repair capacity of melanocytes and reduces the incidence of skin cancers. As such, α-MSH analogs could have the utility for protecting against UV-induced skin DNA damage in susceptible patients. Recently, α-MSH analogs have been approved for the treatment of erythropoietic protoporphyria, hypoactive sexual desire, or pediatric obesity. However, the delivery of these drugs requires inconvenient implants or frequent injections. We recently found that select palmitoylated melanocortin analogs such as afamelanotide and adrenocorticotropin peptides self-assemble to form liquid gels in situ. To explore the utility of these novel analogs, we studied their pharmacological characteristics in vitro and in vivo. Acylated afamelanotide (DDE 313) and ACTH1-24 (DDE314) analogs form liquid gels at 6-20% and have a significantly increased viscosity at >2.5% compared to original analogs. Using the DDE313 analog as a prototype, we showed gel-formation reduces the passage of DDE313 through Centricon filters, and subcutaneous injection of analog gel in rats leads to the sustained presence of the peptide in circulation for >12 days. In addition, DDE313 darkened the skin of frogs for >4 weeks, whereas those injected with an equivalent dose of afamelanotide lost the tanning response within a few days. Because self-assembled gels allow sustained activation of melanocortin receptors, further studies of these analogs may allow the development of effective and convenient tanning therapies to prophylactically protect against UV-induced malignant transformation of skin cells in susceptible patients.
Collapse
Affiliation(s)
- Chia Lin Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Zheqing Cai
- CL Laboratory LLC, Gaithersburg, MD, 20878, United States
| | | |
Collapse
|
11
|
Romanova IV, Mikhailova EV, Mikhrina AL, Shpakov AO. Type 1 melanocortin receptors in pro-opiomelanocortin-, vasopressin-, and oxytocin-immunopositive neurons in different areas of mouse brain. Anat Rec (Hoboken) 2023; 306:2388-2399. [PMID: 35475324 DOI: 10.1002/ar.24934] [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: 02/26/2022] [Revised: 03/24/2022] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
Abstract
Information on the localization of the Type 1 melanocortin receptors (MC1Rs) in different regions of the brain is very scarce. As a result, the role of MC1Rs in the functioning of brain neurons and in the central regulation of physiological functions has not been studied. This work aimed to study the expression and distribution of MС1Rs in different brain areas of female C57Bl/6J mice. Using real-time polymerase chain reaction, we demonstrated the Mс1R gene expression in the cerebral cortex, midbrain, hypothalamus, medulla oblongata, and hippocampus. Using an immunohistochemical approach, we showed the MС1R localization in neurons of the hypothalamic arcuate, paraventricular and supraoptic nuclei, nucleus tractus solitarius (NTS), dorsal hippocampus, substantia nigra, and cerebral cortex. Using double immunolabeling, the MC1Rs were visualized on the surface and in the bodies and outgrowths of pro-opiomelanocortin (POMC)-immunopositive neurons in the hypothalamic arcuate nucleus, NTS, hippocampal CA3 and CA1 regions, and cerebral cortex. Co-localization with POMC indicates that MC1R, like MC3R, is able to function as an autoreceptor. In the paraventricular and supraoptic nuclei, MC1Rs were visualized on the surface and in the cell bodies of vasopressin- and oxytocin-immunopositive neurons, indicating a relationship between hypothalamic MC1R signaling and vasopressin and oxytocin production. The data obtained indicate a wide distribution of MC1Rs in different areas of the mouse brain and their localization in POMC-, vasopressin- and oxytocin-immunopositive neurons, which may indicate the participation of MC1Rs in the control of many physiological processes in the central nervous system.
Collapse
Affiliation(s)
- Irina V Romanova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Elena V Mikhailova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Anastasiya L Mikhrina
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
12
|
Bennett JL, Grove NC, Johnson RK, Mizenko C, DuPont JC, Wagner BD, Lynch AM, Frohman TC, Shindler KS, Frohman EM. A Randomized Prospective Trial Comparing Repository Corticotropin Injection and Intravenous Methylprednisolone for Neuroprotection in Acute Optic Neuritis. J Neuroophthalmol 2023; 43:323-329. [PMID: 37261907 PMCID: PMC10414149 DOI: 10.1097/wno.0000000000001878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
BACKGROUND Repository corticotrophin injection (RCI, Acthar Gel) and intravenous methylprednisolone (IVMP) improve the rate but not the extent of visual recovery following acute optic neuritis. RCI has adrenal-stimulating and melanocortin receptor-stimulating properties that may endow it with unique anti-inflammatory properties relative to IVMP. METHODS Individuals with acute optic neuritis of less than 2 weeks duration were prospectively enrolled and randomized 1:1 to receive either RCI or IVMP. Peripapillary retinal nerve fiber layer (pRNFL) and ganglion cell plus inner plexiform layer thickness (GC + IPL) were serially evaluated by OCT. In addition, patient-reported outcomes (PROs) for changes in fatigue, mood, visual function, depression, and quality of life (QOL) were measured, and high and low contrast visual acuity were recorded. RESULTS Thirty-seven subjects were enrolled (19 RCI; 18 IVMP); the average time from symptom to treatment was 8.8 days. At 6 months, there was no difference in the primary outcome: loss of average pRNFL thickness in the affected eye (RCI vs IVMP: -13.1 vs -11.7 µm, P = 0.88) 6 months after randomization. Additional outcomes also showed no difference between treatment groups: 6-month attenuation of GC + IPL thickness (RCI vs IVMP: -13.8 vs -12.0 µm, P = 0.58) and frequency of pRNFL swelling at 1 month (RCI vs IVMP: 63% vs 72%, P = 0.73) and 3 months (RCI vs IVMP: 26% vs 31%, P = 0.99). Both treatments resulted in improvement in visual function and PROs. CONCLUSIONS Treatment of acute optic neuritis with RCI or IVMP produced no clinically meaningful differences in optic nerve structure or visual function.
Collapse
|
13
|
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
|
14
|
Gravina AG, Pellegrino R, Durante T, Palladino G, Imperio G, D'Amico G, Trotta MC, Dallio M, Romeo M, D'Amico M, Federico A. The Melanocortin System in Inflammatory Bowel Diseases: Insights into Its Mechanisms and Therapeutic Potentials. Cells 2023; 12:1889. [PMID: 37508552 PMCID: PMC10378568 DOI: 10.3390/cells12141889] [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: 06/24/2023] [Revised: 07/08/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
The melanocortin system is a complex set of molecular mediators and receptors involved in many physiological and homeostatic processes. These include the regulation of melanogenesis, steroidogenesis, neuromodulation and the modulation of inflammatory processes. In the latter context, the system has assumed importance in conditions of chronic digestive inflammation, such as inflammatory bowel diseases (IBD), in which numerous experiences have been accumulated in mouse models of colitis. Indeed, information on how such a system can counteract colitis inflammation and intervene in the complex cytokine imbalance in the intestinal microenvironment affected by chronic inflammatory damage has emerged. This review summarises the evidence acquired so far and highlights that molecules interfering with the melanocortin system could represent new drugs for treating IBD.
Collapse
Affiliation(s)
- Antonietta Gerarda Gravina
- Hepatogastroenterology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Raffaele Pellegrino
- Hepatogastroenterology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Tommaso Durante
- Mental Health Department, S. Pio Hospital, Via dell'Angelo, 82100 Benevento, Italy
| | - Giovanna Palladino
- Hepatogastroenterology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Giuseppe Imperio
- Hepatogastroenterology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | | | - Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Marcello Dallio
- Hepatogastroenterology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Mario Romeo
- Hepatogastroenterology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Michele D'Amico
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Alessandro Federico
- Hepatogastroenterology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
15
|
Srivastava P, Nishiyama S, Lin SH, Srivastava A, Su C, Peng W, Levy M, Schwarzschild M, Xu Y, Chen X. Peripheral MC1R activation modulates immune responses and is neuroprotective in a mouse model of Parkinson's disease. RESEARCH SQUARE 2023:rs.3.rs-3042571. [PMID: 37398302 PMCID: PMC10312952 DOI: 10.21203/rs.3.rs-3042571/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background Melanocortin 1 receptor (MC1R) is a key pigmentation gene, and loss-of-function of MC1R variants that produce red hair may be associated with Parkinson's disease (PD). We previously reported compromised dopaminergic neuron survival in Mc1r mutant mice and dopaminergic neuroprotective effects of local injection of a MC1R agonist to the brain or a systemically administered MC1R agonist with appreciable CNS permeability. Beyond melanocytes and dopaminergic neurons, MC1R is expressed in other peripheral tissues and cell types, including immune cells. The present study investigates the impact of NDP-MSH, a synthetic melanocortin receptor (MCR) agonist that does not cross BBB, on the immune system and the nigrostriatal dopaminergic system in mouse model of PD. Methods C57BL/6 mice were treated systemically with MPTP.HCl (20 mg/kg) and LPS (1 mg/kg) from day 1 to day 4 and NDP-MSH (400 μg/kg) or vehicle from day 1 to day 12 following which the mice were sacrificed. Peripheral and CNS immune cells were phenotyped and inflammatory markers were measured. The nigrostriatal dopaminergic system was assessed behaviorally, chemically, immunologically, and pathologically. To understand the role of regulatory T cells (Tregs) in this model, CD25 monoclonal antibody was used to deplete CD25+ Tregs. Results Systemic NDP-MSH administration significantly attenuated striatal dopamine depletion and nigral dopaminergic neuron loss induced by MPTP+LPS. It improved the behavioral outcomes in the pole test. Mc1r mutant mice injected with NDP-MSH in the MPTP and LPS paradigm showed no changes in striatal dopamine levels suggesting that the NDP-MSH acts through the MC1R pathway. Although no NDP-MSH was detected in the brain, peripheral, NDP-MSH attenuated neuroinflammation as observed by diminished microglial activation in the nigral region, along with reduced TNF-α and IL1β levels in the ventral midbrain. Depletion of Tregs limited the neuroprotective effects of NDP-MSH. Conclusions Our study demonstrates that peripherally acting NDP-MSH confers protection on dopaminergic nigrostriatal neurons and reduces hyperactivated microglia. NDP-MSH modulates peripheral immune responses, and Tregs may be involved in the neuroprotective effect of NDP-MSH.
Collapse
Affiliation(s)
- Pranay Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Shuhei Nishiyama
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Sonia H Lin
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Akriti Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Chienwen Su
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston
| | - Michael Levy
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Michael Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Yuehang Xu
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| |
Collapse
|
16
|
Zhou Y, Ju H, Hu Y, Li T, Chen Z, Si Y, Sun X, Shi Y, Fang H. Tregs dysfunction aggravates postoperative cognitive impairment in aged mice. J Neuroinflammation 2023; 20:75. [PMID: 36932450 PMCID: PMC10022212 DOI: 10.1186/s12974-023-02760-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/09/2023] [Indexed: 03/19/2023] Open
Abstract
OBJECTIVES Enhanced neuroinflammation is an important mechanism underlying perioperative neurocognitive disorders. Regulatory T cells (Tregs) play a crucial role in regulating systemic immune responses. The present study was aimed to investigate the participation of Tregs in the development of postoperative cognitive dysfunction (POCD). METHODS Surgery-associated neurocognitive disorder was induced in 18-month-old mice subjected to internal fixation of tibial fracture. Morris water maze was used to examine mice cognitive function. Splenic Tregs were collected for RNA sequencing and flow cytometry. Levels of inflammatory factors in the circulation and hippocampus were measured by enzyme-linked immunosorbent assay. Protein presences of tight junction proteins were detected by immunofluorescence. RESULTS Surgery of internal fixation of tibial fracture induced cognitive impairment in aged mice, accompanied by elevated plasma levels of inflammatory factors and increased circulating Tregs. Transfusion of Tregs from young mice partially restored the structure of the blood-brain barrier and alleviated POCD in aged mice. Compared with young Tregs, differentially expressed genes in aged Tregs were enriched in tumor necrosis factor (TNF) signaling pathway and cytokine-cytokine receptor interaction. Flow cytometry revealed that aged Tregs had blunted functions under basal and stimulated conditions. Blockade of the CD25 epitope protected the blood-brain barrier structure, reduced TNF-α levels in the hippocampus, and improved surgery-associated cognition in aged mice. CONCLUSIONS Blocking peripheral regulatory T cells improves surgery-induced cognitive function in aged mice. Therefore, aged Tregs play an essential role in the occurrence of POCD.
Collapse
Affiliation(s)
- Yile Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huihui Ju
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tingting Li
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhouyi Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Si
- Department of Anesthesiology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xia Sun
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, China.
| | - Yi Shi
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Anesthesiology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Chen B, Guan X, Gunning WT, Ge Y, Gohara AF, Dworkin LD, Gong R. Negative Modulation of B Cell Activation by Melanocortin 1 Receptor Signaling Protects against Membranous Nephropathy. J Am Soc Nephrol 2023; 34:467-481. [PMID: 36446431 PMCID: PMC10103281 DOI: 10.1681/asn.2022050605] [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: 05/25/2022] [Revised: 10/09/2022] [Accepted: 11/06/2022] [Indexed: 12/05/2022] Open
Abstract
SIGNIFICANCE STATEMENT Emerging evidence suggests that melanocortin neuropeptides-specifically adrenocorticotropic hormone-offer a novel, steroidogenic-independent therapeutic modality for membranous nephropathy (MN). The molecular mechanism underlying this beneficial effect, however, remains largely elusive. To investigate whether melanocortins modulate humoral immunity, the authors induced passive Heymann nephritis, a model of human MN, in wild-type and melanocortin 1 receptor (MC1R) knockout rats and treated them with melanocortin agents. Additional rats received adoptive transfer of bone marrow-derived cells beforehand from wild-type or MC1R knockout rats. The findings indicate that MC1R signaling plays a key role in negative modulation of B-cell activation and thereby suppresses humoral immune responses in passive Heymann nephritis, and suggest that MC1R signaling might offer a novel B cell-targeted therapeutic strategy for MN. BACKGROUND Emerging evidence suggests that the pituitary neuropeptide melanocortins-specifically, adrenocorticotropic hormone-offer a novel nonsteroidogenic therapeutic modality for membranous nephropathy (MN). However, the mechanism(s) of action remains elusive. METHODS To investigate whether melanocortins modulate humoral immunity, we induced passive Heymann nephritis (PHN), a model of MN, in wild-type (WT) and melanocortin 1 receptor (MC1R) knockout (KO) rats. We treated the animals with melanocortin agents-repository corticotropin injection, the nonsteroidogenic pan-melanocortin receptor agonist [Nle 4 , DPhe 7 ]-α-melanocyte stimulating hormone, the selective MC1R agonist MS05, vehicle gel, or phosphate-buffered saline-and evaluated kidney function, histology, and molecular changes. Additional rats received adoptive transfer of syngeneic bone marrow-derived cells beforehand from WT or MC1R KO rats. RESULTS KO of MC1R worsened PHN and this was associated with increased deposition of autologous immunoglobulin G (IgG) and complement C5b-9 in glomeruli and higher circulating levels of autologous IgG-evidence of a sensitized humoral immune response. Melanocortin therapy ameliorated PHN in WT rats, coinciding with reduced glomerular deposition of autologous IgG and C5b -9. The beneficial efficacy of melanocortins was blunted in KO rats but restored by adoptive transfer of syngeneic bone marrow-derived cells derived from WT rats. Mechanistically, MC1R was expressed in B lymphocytes and was negatively associated with B cell activation. MC1R agonism triggered the expression of microphthalmia-associated transcription factor in activated B cells in a cAMP-dependent mode and also repressed the expression of interferon regulatory factor 4 (a lymphoid transcription factor essential for B-cell development and maturation), resulting in suppressed plasma cell differentiation and IgG production. CONCLUSIONS MC1R signaling negatively modulates B cell activation and suppresses humoral immune responses in PHN, suggesting that MC1R signaling might offer a novel therapeutic target for MN.
Collapse
Affiliation(s)
- Bohan Chen
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio
- Current address: Blood Purification Center, Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuejing Guan
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio
| | - William T. Gunning
- Department of Pathology, University of Toledo Medical Center, Toledo, Ohio
| | - Yan Ge
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio
| | - Amira F. Gohara
- Department of Pathology, University of Toledo Medical Center, Toledo, Ohio
| | - Lance D. Dworkin
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio
- Division of Kidney Disease and Hypertension, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Rujun Gong
- Division of Nephrology, University of Toledo College of Medicine, Toledo, Ohio
- Division of Kidney Disease and Hypertension, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio
- Center for Diabetes and Endocrine Research, University of Toledo Medical Center, Toledo, Ohio
| |
Collapse
|
18
|
Guan X, Chen B, Malhotra DK, Gohara AF, Dworkin LD, Gong R. Hematopoietic-specific melanocortin 1 receptor signaling protects against nephrotoxic serum nephritis and mediates the beneficial effect of melanocortin therapy. Kidney Int 2023; 103:331-342. [PMID: 36374665 PMCID: PMC10431720 DOI: 10.1016/j.kint.2022.09.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/06/2022]
Abstract
The melanocortin hormone system has emerged as a novel therapeutic target for treating refractory glomerular diseases. However, the role of hematopoietic melanocortin 1 receptor (MC1R) signaling remains unknown. Upon insult by rabbit nephrotoxic serum, MC1R null-mutant mice developed more severe crescentic glomerulonephritis than wild-type mice, marked by aggravated proteinuria, kidney dysfunction and histologic lesions. Melanocortin therapy, using Repository Corticotropin Injection (Acthar Gel), the pan-melanocortin receptor agonist NDP-MSH, or the MC1R agonist MS05, ameliorated experimental nephritis in wild-type mice but this effect was blunted in null mice. Exacerbated experimental nephritis in null mice was associated with increased glomerular deposition of autologous IgG and C5b-9, in parallel with higher circulating levels of autologous IgG2c and IgG3. Additionally, the Th1 immune response was potentiated in null mice with experimental nephritis, accompanied by diminished kidney FoxP3+ regulatory T cells. Kidney infiltration of macrophages was also augmented by MC1R deficiency with an enhanced M1 polarization. Moreover, adoptive transfer of syngeneic bone marrow-derived cells from wild-type mice mitigated experimental nephritis in null mice and restored the beneficial efficacy of melanocortins. Mechanistically, MC1R was expressed by diverse subsets of kidney leukocytes, including macrophages, T and B lymphocytes, and was inversely associated with the NFκB pathway, a key player in immune responses. MS05 attenuated the production of rabbit IgG-specific IgG2c and IgG3 in cultured wild-type splenocytes, and promoted M2 polarization in M1-primed wild-type macrophages, associated with NFκB inhibition. In contrast, in null splenocytes or macrophages, this effect of MS05 was barely detectable, but was mimicked by an NFκB inhibitor. Thus, hematopoietic MC1R signaling attenuates experimental nephritis and mediates the beneficial effect of melanocortin therapy via, in part, regulating the immune response.
Collapse
Affiliation(s)
- Xuejing Guan
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA; The Center for Diabetes and Endocrine Research, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Bohan Chen
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA; The Center for Diabetes and Endocrine Research, University of Toledo Medical Center, Toledo, Ohio, USA; Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island, USA
| | - Deepak K Malhotra
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Amira F Gohara
- Department of Pathology, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Lance D Dworkin
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Rujun Gong
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA; The Center for Diabetes and Endocrine Research, University of Toledo Medical Center, Toledo, Ohio, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio, USA.
| |
Collapse
|
19
|
Shahraki AH, Tian R, Zhang C, Fregien NL, Bejarano P, Mirsaeidi M. Anti-inflammatory Properties of the Alpha-Melanocyte-Stimulating Hormone in Models of Granulomatous Inflammation. Lung 2022; 200:463-472. [PMID: 35717488 PMCID: PMC9360058 DOI: 10.1007/s00408-022-00546-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/29/2022] [Indexed: 11/08/2022]
Abstract
Purpose Alpha-melanocyte stimulating hormone (α-MSH) is known to have anti-inflammatory effects. However, the anti-inflammatory properties of α-MSH on normal bronchial epithelial cells are largely unknown, especially in the context of in vitro sarcoidosis models. Methods We evaluated the anti-inflammatory effects of α-MSH on two different in vitro sarcoidosis models (lung-on-membrane model; LOMM and three-dimensional biochip pulmonary sarcoidosis model; 3D-BSGM) generated from NBECs and an in vivo sarcoidosis mouse model. Results Treatment with α-MSH decreased inflammatory cytokine levels and downregulated type I interferon pathway genes and related proteins in LOMM and 3D-BSGM models. Treatment with α-MSH also significantly decreased macrophages and cytotoxic T-cells counts in a sarcoidosis mice model. Conclusion Our results confirm the direct role of type I IFNs in the pathogenesis of sarcoid lung granulomas and highlight α-MSH as a potential novel therapeutic agent for treating pulmonary sarcoidosis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s00408-022-00546-x.
Collapse
Affiliation(s)
- Abdolrazagh Hashemi Shahraki
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Jacksonville, University of Florida, 655 West 11th Street, Jacksonville, FL, 32209, USA
| | - Runxia Tian
- Department of Cell Biology, University of Miami, Miami, FL, USA
| | - Chongxu Zhang
- Department of Cell Biology, University of Miami, Miami, FL, USA
| | - Nevis L Fregien
- Department of Cell Biology, University of Miami, Miami, FL, USA
| | - Pablo Bejarano
- Department of Pathology, Cleveland Clinic, Weston, FL, USA
| | - Mehdi Mirsaeidi
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Jacksonville, University of Florida, 655 West 11th Street, Jacksonville, FL, 32209, USA.
| |
Collapse
|
20
|
Lužnik Marzidovšek Z, Blanco T, Sun Z, Alemi H, Ortiz G, Nakagawa H, Chauhan SK, Taylor AW, Jurkunas UV, Yin J, Dana R. The Neuropeptide Alpha-Melanocyte-Stimulating Hormone Is Critical for Corneal Endothelial Cell Protection and Graft Survival after Transplantation. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:270-280. [PMID: 34774519 PMCID: PMC8908049 DOI: 10.1016/j.ajpath.2021.10.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 02/03/2023]
Abstract
Corneal transplantation is the most common form of tissue transplantation. The success of corneal transplantation mainly relies on the integrity of corneal endothelial cells (CEnCs), which maintain tissue transparency by pumping out excess water from the cornea. After transplantation, the rate of CEnC loss far exceeds that seen with normal aging, which can threaten sight. The underlying mechanisms are poorly understood. Alpha-melanocyte-stimulating hormone (α-MSH) is a neuropeptide that is constitutively found in the aqueous humor with both cytoprotective and immunomodulatory effects. The curent study found high expression of melanocortin 1 receptor (MC1R), the receptor for α-MSH, on CEnCs. The effect of α-MSH/MC1R signaling on endothelial function and allograft survival in vitro and in vivo was investigated using MC1R signaling-deficient mice (Mc1re/e mice with a nonfunctional MC1R). Herein, the results indicate that in addition to its well-known immunomodulatory effect, α-MSH has cytoprotective effects on CEnCs after corneal transplantation, and the loss of MC1R signaling significantly decreases long-term graft survival in vivo. In conclusion, α-MSH/MC1R signaling is critical for CEnC function and graft survival after corneal transplantation.
Collapse
Affiliation(s)
- Zala Lužnik Marzidovšek
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts,Eye Hospital, University Medical Centre, Ljubljana, Slovenia
| | - Tomas Blanco
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Zhongmou Sun
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Hamid Alemi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Gustavo Ortiz
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Hayate Nakagawa
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Sunil K. Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Andrew W. Taylor
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts
| | - Ula V. Jurkunas
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Jia Yin
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts,Address correspondence to Reza Dana, M.D., M.P.H., M.Sc., or Jia Yin, M.D., Ph.D., M.P.H., Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA 02114.
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts,Address correspondence to Reza Dana, M.D., M.P.H., M.Sc., or Jia Yin, M.D., Ph.D., M.P.H., Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA 02114.
| |
Collapse
|
21
|
Schroeter CB, Huntemann N, Bock S, Nelke C, Kremer D, Pfeffer K, Meuth SG, Ruck T. Crosstalk of Microorganisms and Immune Responses in Autoimmune Neuroinflammation: A Focus on Regulatory T Cells. Front Immunol 2021; 12:747143. [PMID: 34691057 PMCID: PMC8529161 DOI: 10.3389/fimmu.2021.747143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells (Tregs) are the major determinant of peripheral immune tolerance. Many Treg subsets have been described, however thymus-derived and peripherally induced Tregs remain the most important subpopulations. In multiple sclerosis, a prototypical autoimmune disorder of the central nervous system, Treg dysfunction is a pathogenic hallmark. In contrast, induction of Treg proliferation and enhancement of their function are central immune evasion mechanisms of infectious pathogens. In accordance, Treg expansion is compartmentalized to tissues with high viral replication and prolonged in chronic infections. In friend retrovirus infection, Treg expansion is mainly based on excessive interleukin-2 production by infected effector T cells. Moreover, pathogens seem also to enhance Treg functions as shown in human immunodeficiency virus infection, where Tregs express higher levels of effector molecules such as cytotoxic T-lymphocyte-associated protein 4, CD39 and cAMP and show increased suppressive capacity. Thus, insights into the molecular mechanisms by which intracellular pathogens alter Treg functions might aid to find new therapeutic approaches to target central nervous system autoimmunity. In this review, we summarize the current knowledge of the role of pathogens for Treg function in the context of autoimmune neuroinflammation. We discuss the mechanistic implications for future therapies and provide an outlook for new research directions.
Collapse
Affiliation(s)
- Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefanie Bock
- Department of Neurology With Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
22
|
Zhou Y, Chawla MK, Rios-Monterrosa JL, Wang L, Zempare MA, Hruby VJ, Barnes CA, Cai M. Aged Brains Express Less Melanocortin Receptors, Which Correlates with Age-Related Decline of Cognitive Functions. Molecules 2021; 26:6266. [PMID: 34684847 PMCID: PMC8541441 DOI: 10.3390/molecules26206266] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/03/2021] [Accepted: 10/11/2021] [Indexed: 12/20/2022] Open
Abstract
Brain G-protein coupled receptors have been hypothesized to be potential targets for maintaining or restoring cognitive function in normal aged individuals or in patients with neurodegenerative disease. A number of recent reports suggest that activation of melanocortin receptors (MCRs) in the brain can significantly improve cognitive functions of normal rodents and of different rodent models of the Alzheimer's disease. However, the potential impact of normative aging on the expression of MCRs and their potential roles for modulating cognitive function remains to be elucidated. In the present study, we first investigated the expression of these receptors in six different brain regions of young (6 months) and aged (23 months) rats following assessment of their cognitive status. Correlation analysis was further performed to reveal potential contributions of MCR subtypes to spatial learning and memory. Our results revealed statistically significant correlations between the expression of several MCR subtypes in the frontal cortex/hypothalamus and the hippocampus regions and the rats' performance in spatial learning and memory only in the aged rats. These findings support the hypothesis that aging has a direct impact on the expression and function of MCRs, establishing MCRs as potential drug targets to alleviate aging-induced decline of cognitive function.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA; (Y.Z.); (J.L.R.-M.); (L.W.); (V.J.H.)
| | - Monica K. Chawla
- Evelyn F. McKnight Brain Institute, The University of Arizona, Tucson, AZ 85721, USA; (M.K.C.); (M.A.Z.); (C.A.B.)
- Division of Neural Systems, Memory & Aging, The University of Arizona, Tucson, AZ 85721, USA
| | - Jose L. Rios-Monterrosa
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA; (Y.Z.); (J.L.R.-M.); (L.W.); (V.J.H.)
| | - Lingzhi Wang
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA; (Y.Z.); (J.L.R.-M.); (L.W.); (V.J.H.)
| | - Marc A. Zempare
- Evelyn F. McKnight Brain Institute, The University of Arizona, Tucson, AZ 85721, USA; (M.K.C.); (M.A.Z.); (C.A.B.)
- Division of Neural Systems, Memory & Aging, The University of Arizona, Tucson, AZ 85721, USA
| | - Victor J. Hruby
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA; (Y.Z.); (J.L.R.-M.); (L.W.); (V.J.H.)
| | - Carol A. Barnes
- Evelyn F. McKnight Brain Institute, The University of Arizona, Tucson, AZ 85721, USA; (M.K.C.); (M.A.Z.); (C.A.B.)
- Division of Neural Systems, Memory & Aging, The University of Arizona, Tucson, AZ 85721, USA
- Department of Psychology, Neurology and Neuroscience, The University of Arizona, Tucson, AZ 85721, USA
| | - Minying Cai
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA; (Y.Z.); (J.L.R.-M.); (L.W.); (V.J.H.)
| |
Collapse
|
23
|
Brain Protein Expression Profile Confirms the Protective Effect of the ACTH (4-7)PGP Peptide (Semax) in a Rat Model of Cerebral Ischemia-Reperfusion. Int J Mol Sci 2021; 22:ijms22126179. [PMID: 34201112 PMCID: PMC8226508 DOI: 10.3390/ijms22126179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 12/02/2022] Open
Abstract
The Semax (Met-Glu-His-Phe-Pro-Gly-Pro) peptide is a synthetic melanocortin derivative that is used in the treatment of ischemic stroke. Previously, studies of the molecular mechanisms underlying the actions of Semax using models of cerebral ischemia in rats showed that the peptide enhanced the transcription of neurotrophins and their receptors and modulated the expression of genes involved in the immune response. A genome-wide RNA-Seq analysis revealed that, in the rat transient middle cerebral artery occlusion (tMCAO) model, Semax suppressed the expression of inflammatory genes and activated the expression of neurotransmitter genes. Here, we aimed to evaluate the effect of Semax in this model via the brain expression profiling of key proteins involved in inflammation and cell death processes (MMP-9, c-Fos, and JNK), as well as neuroprotection and recovery (CREB) in stroke. At 24 h after tMCAO, we observed the upregulation of active CREB in subcortical structures, including the focus of the ischemic damage; downregulation of MMP-9 and c-Fos in the adjacent frontoparietal cortex; and downregulation of active JNK in both tissues under the action of Semax. Moreover, a regulatory network was constructed. In conclusion, the suppression of inflammatory and cell death processes and the activation of recovery may contribute to the neuroprotective action of Semax at both the transcriptome and protein levels.
Collapse
|
24
|
Ten Bosch GJA, Bolk J, 't Hart BA, Laman JD. Multiple sclerosis is linked to MAPK ERK overactivity in microglia. J Mol Med (Berl) 2021; 99:1033-1042. [PMID: 33948692 PMCID: PMC8313465 DOI: 10.1007/s00109-021-02080-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/31/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022]
Abstract
Reassessment of published observations in patients with multiple sclerosis (MS) suggests a microglial malfunction due to inappropriate (over)activity of the mitogen-activated protein kinase pathway ERK (MAPKERK). These observations regard biochemistry as well as epigenetics, and all indicate involvement of this pathway. Recent preclinical research on neurodegeneration already pointed towards a role of MAPK pathways, in particular MAPKERK. This is important as microglia with overactive MAPK have been identified to disturb local oligodendrocytes which can lead to locoregional demyelination, hallmark of MS. This constitutes a new concept on pathophysiology of MS, besides the prevailing view, i.e., autoimmunity. Acknowledged risk factors for MS, such as EBV infection, hypovitaminosis D, and smoking, all downregulate MAPKERK negative feedback phosphatases that normally regulate MAPKERK activity. Consequently, these factors may contribute to inappropriate MAPKERK overactivity, and thereby to neurodegeneration. Also, MAPKERK overactivity in microglia, as a factor in the pathophysiology of MS, could explain ongoing neurodegeneration in MS patients despite optimized immunosuppressive or immunomodulatory treatment. Currently, for these patients with progressive disease, no effective treatment exists. In such refractory MS, targeting the cause of overactive MAPKERK in microglia merits further investigation as this phenomenon may imply a novel treatment approach.
Collapse
Affiliation(s)
- George J A Ten Bosch
- Department of Medical Oncology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Jolande Bolk
- Department of Anesthesiology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Bert A 't Hart
- Department Anatomy and Neuroscience, Amsterdam University Medical Center (VUmc), Amsterdam, The Netherlands.,Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, Groningen, The Netherlands
| | - Jon D Laman
- Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
25
|
Sunlight exposure exerts immunomodulatory effects to reduce multiple sclerosis severity. Proc Natl Acad Sci U S A 2021; 118:2018457118. [PMID: 33376202 PMCID: PMC7817192 DOI: 10.1073/pnas.2018457118] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) disease risk is associated with reduced sun-exposure. This study assessed the relationship between measures of sun exposure (vitamin D [vitD], latitude) and MS severity in the setting of two multicenter cohort studies (n NationMS = 946, n BIONAT = 990). Additionally, effect-modification by medication and photosensitivity-associated MC1R variants was assessed. High serum vitD was associated with a reduced MS severity score (MSSS), reduced risk for relapses, and lower disability accumulation over time. Low latitude was associated with higher vitD, lower MSSS, fewer gadolinium-enhancing lesions, and lower disability accumulation. The association of latitude with disability was lacking in IFN-β-treated patients. In carriers of MC1R:rs1805008(T), who reported increased sensitivity toward sunlight, lower latitude was associated with higher MRI activity, whereas for noncarriers there was less MRI activity at lower latitudes. In a further exploratory approach, the effect of ultraviolet (UV)-phototherapy on the transcriptome of immune cells of MS patients was assessed using samples from an earlier study. Phototherapy induced a vitD and type I IFN signature that was most apparent in monocytes but that could also be detected in B and T cells. In summary, our study suggests beneficial effects of sun exposure on established MS, as demonstrated by a correlative network between the three factors: Latitude, vitD, and disease severity. However, sun exposure might be detrimental for photosensitive patients. Furthermore, a direct induction of type I IFNs through sun exposure could be another mechanism of UV-mediated immune-modulation in MS.
Collapse
|
26
|
Filippenkov IB, Dergunova LV, Limborska SA, Myasoedov NF. Neuroprotective Effects of Peptides in the Brain: Transcriptome Approach. BIOCHEMISTRY (MOSCOW) 2021; 85:279-287. [PMID: 32564732 DOI: 10.1134/s0006297920030037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The importance of studying the action mechanisms of drugs based on natural regulatory peptides is commonly recognized. Particular attention is paid to the peptide drugs that contribute to the restoration of brain functions after acute cerebrovascular accidents (stroke), which for many years continues to be one of the main problems and threats to human health. However, molecular genetic changes in the brain in response to ischemia, as well as the mechanisms of protective effects of peptides, have not been sufficiently studied. This limits the use of neuroprotective peptides and makes it difficult to develop new, more efficient drugs with targeted action on brain functions. Transcriptome analysis is a promising approach for studying the mechanisms of the damaging effects of cerebral ischemia and neuroprotective action of peptide drugs. Beside investigating the role of mRNAs in protein synthesis, the development of new neuroprotection strategies requires studying the involvement of regulatory RNAs in ischemia. Of greatest interest are microRNAs (miRNAs) and circular RNAs (circRNAs), which are expressed predominantly in the brain. CircRNAs can interact with miRNAs and diminish their activity, thereby inhibiting miRNA-mediated repression of mRNAs. It has become apparent that analysis of the circRNA/miRNA/mRNA system is essential for deciphering the mechanisms of brain damage and repair. Here, we present the results of studies on the ischemia-induced changes in the activity of genes and peptide-mediated alterations in the transcriptome profiles in experimental ischemia and formulate the basic principles of peptide regulation in the ischemia-induced damage.
Collapse
Affiliation(s)
- I B Filippenkov
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia.
| | - L V Dergunova
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - S A Limborska
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - N F Myasoedov
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| |
Collapse
|
27
|
Activation of MC1R with BMS-470539 attenuates neuroinflammation via cAMP/PKA/Nurr1 pathway after neonatal hypoxic-ischemic brain injury in rats. J Neuroinflammation 2021; 18:26. [PMID: 33468172 PMCID: PMC7814630 DOI: 10.1186/s12974-021-02078-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022] Open
Abstract
Background Microglia-mediated neuroinflammation plays a crucial role in the pathogenesis of hypoxic-ischemic (HI)-induced brain injury. Activation of melanocortin-1 receptor (MC1R) has been shown to exert anti-inflammatory and neuroprotective effects in several neurological diseases. In the present study, we have explored the role of MC1R activation on neuroinflammation and the potential underlying mechanisms after neonatal hypoxic-ischemic brain injury in rats. Methods A total of 169 post-natal day 10 unsexed rat pups were used. HI was induced by right common carotid artery ligation followed by 2.5 h of hypoxia. BMS-470539, a specific selective MC1R agonist, was administered intranasally at 1 h after HI induction. To elucidate the potential underlying mechanism, MC1R CRISPR KO plasmid or Nurr1 CRISPR KO plasmid was administered via intracerebroventricular injection at 48 h before HI induction. Percent brain infarct area, short- and long-term neurobehavioral tests, Nissl staining, immunofluorescence staining, and Western blot were conducted. Results The expression levels of MC1R and Nurr1 increased over time post-HI. MC1R and Nurr1 were expressed on microglia at 48 h post-HI. Activation of MC1R with BMS-470539 significantly reduced the percent infarct area, brain atrophy, and inflammation, and improved short- and long-term neurological deficits at 48 h and 28 days post-HI. MC1R activation increased the expression of CD206 (a microglial M2 marker) and reduced the expression of MPO. Moreover, activation of MC1R with BMS-470539 significantly increased the expression levels of MC1R, cAMP, p-PKA, and Nurr1, while downregulating the expression of pro-inflammatory cytokines (TNFα, IL-6, and IL-1β) at 48 h post-HI. However, knockout of MC1R or Nurr1 by specific CRISPR reversed the neuroprotective effects of MC1R activation post-HI. Conclusions Our study demonstrated that activation of MC1R with BMS-470539 attenuated neuroinflammation, and improved neurological deficits after neonatal hypoxic-ischemic brain injury in rats. Such anti-inflammatory and neuroprotective effects were mediated, at least in part, via the cAMP/PKA/Nurr1 signaling pathway. Therefore, MC1R activation might be a promising therapeutic target for infants with hypoxic-ischemic encephalopathy (HIE). Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02078-2.
Collapse
|
28
|
Dinparastisaleh R, Mirsaeidi M. Antifibrotic and Anti-Inflammatory Actions of α-Melanocytic Hormone: New Roles for an Old Player. Pharmaceuticals (Basel) 2021; 14:ph14010045. [PMID: 33430064 PMCID: PMC7827684 DOI: 10.3390/ph14010045] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/16/2022] Open
Abstract
The melanocortin system encompasses melanocortin peptides, five receptors, and two endogenous antagonists. Besides pigmentary effects generated by α-Melanocytic Hormone (α-MSH), new physiologic roles in sexual activity, exocrine secretion, energy homeostasis, as well as immunomodulatory actions, exerted by melanocortins, have been described recently. Among the most common and burdensome consequences of chronic inflammation is the development of fibrosis. Depending on the regenerative capacity of the affected tissue and the quality of the inflammatory response, the outcome is not always perfect, with the development of some fibrosis. Despite the heterogeneous etiology and clinical presentations, fibrosis in many pathological states follows the same path of activation or migration of fibroblasts, and the differentiation of fibroblasts to myofibroblasts, which produce collagen and α-SMA in fibrosing tissue. The melanocortin agonists might have favorable effects on the trajectories leading from tissue injury to inflammation, from inflammation to fibrosis, and from fibrosis to organ dysfunction. In this review we briefly summarized the data on structure, receptor signaling, and anti-inflammatory and anti-fibrotic properties of α-MSH and proposed that α-MSH analogues might be promising future therapeutic candidates for inflammatory and fibrotic diseases, regarding their favorable safety profile.
Collapse
Affiliation(s)
- Roshan Dinparastisaleh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Mehdi Mirsaeidi
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL 33146, USA
- Correspondence: ; Tel.: +1-305-243-1377
| |
Collapse
|
29
|
Lee EY, Chan LC, Wang H, Lieng J, Hung M, Srinivasan Y, Wang J, Waschek JA, Ferguson AL, Lee KF, Yount NY, Yeaman MR, Wong GCL. PACAP is a pathogen-inducible resident antimicrobial neuropeptide affording rapid and contextual molecular host defense of the brain. Proc Natl Acad Sci U S A 2021; 118:e1917623117. [PMID: 33372152 PMCID: PMC7817161 DOI: 10.1073/pnas.1917623117] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Defense of the central nervous system (CNS) against infection must be accomplished without generation of potentially injurious immune cell-mediated or off-target inflammation which could impair key functions. As the CNS is an immune-privileged compartment, inducible innate defense mechanisms endogenous to the CNS likely play an essential role in this regard. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide known to regulate neurodevelopment, emotion, and certain stress responses. While PACAP is known to interact with the immune system, its significance in direct defense of brain or other tissues is not established. Here, we show that our machine-learning classifier can screen for immune activity in neuropeptides, and correctly identified PACAP as an antimicrobial neuropeptide in agreement with previous experimental work. Furthermore, synchrotron X-ray scattering, antimicrobial assays, and mechanistic fingerprinting provided precise insights into how PACAP exerts antimicrobial activities vs. pathogens via multiple and synergistic mechanisms, including dysregulation of membrane integrity and energetics and activation of cell death pathways. Importantly, resident PACAP is selectively induced up to 50-fold in the brain in mouse models of Staphylococcus aureus or Candida albicans infection in vivo, without inducing immune cell infiltration. We show differential PACAP induction even in various tissues outside the CNS, and how these observed patterns of induction are consistent with the antimicrobial efficacy of PACAP measured in conditions simulating specific physiologic contexts of those tissues. Phylogenetic analysis of PACAP revealed close conservation of predicted antimicrobial properties spanning primitive invertebrates to modern mammals. Together, these findings substantiate our hypothesis that PACAP is an ancient neuro-endocrine-immune effector that defends the CNS against infection while minimizing potentially injurious neuroinflammation.
Collapse
Affiliation(s)
- Ernest Y Lee
- Department of Bioengineering, University of California, Los Angeles, CA 90095
- UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| | - Liana C Chan
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA 90509
- Division of Molecular Medicine, Los Angeles County, Harbor-UCLA Medical Center, Torrance, CA 90509
- Division of Infectious Diseases, Los Angeles County, Harbor-UCLA Medical Center, Torrance, CA 90509
| | - Huiyuan Wang
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA 90509
- Division of Molecular Medicine, Los Angeles County, Harbor-UCLA Medical Center, Torrance, CA 90509
| | - Juelline Lieng
- Department of Bioengineering, University of California, Los Angeles, CA 90095
| | - Mandy Hung
- Department of Bioengineering, University of California, Los Angeles, CA 90095
| | - Yashes Srinivasan
- Department of Bioengineering, University of California, Los Angeles, CA 90095
| | - Jennifer Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095
| | - James A Waschek
- Semel Institute for Neuroscience and Human Behavior, Intellectual Development and Disabilities Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Andrew L Ferguson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
| | - Kuo-Fen Lee
- Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Nannette Y Yount
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA 90509
- Division of Molecular Medicine, Los Angeles County, Harbor-UCLA Medical Center, Torrance, CA 90509
| | - Michael R Yeaman
- Division of Molecular Medicine, Los Angeles County, Harbor-UCLA Medical Center, Torrance, CA 90509;
- Division of Infectious Diseases, Los Angeles County, Harbor-UCLA Medical Center, Torrance, CA 90509
- Semel Institute for Neuroscience and Human Behavior, Intellectual Development and Disabilities Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Gerard C L Wong
- Department of Bioengineering, University of California, Los Angeles, CA 90095;
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
| |
Collapse
|
30
|
Huang YJ, Galen K, Zweifel B, Brooks LR, Wright AD. Distinct binding and signaling activity of Acthar Gel compared to other melanocortin receptor agonists. J Recept Signal Transduct Res 2020; 41:425-433. [PMID: 32938265 DOI: 10.1080/10799893.2020.1818094] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE To compare the binding and agonistic activity of Acthar® Gel and synthetic melanocortin receptor (MCR) agonists and examine how the activity of select agonists affects the in vivo production of corticosterone. MATERIALS AND METHODS In vitro binding was determined using concentration-dependent displacement of the ligand [125I]Nle4, D-Phe7-α-melanocyte-stimulating hormone (α-MSH) on cells expressing MC1R, MC3R, MC4R, or MC5R. Functional activity was determined using a time-resolved fluorescence cyclic adenosine monophosphate (cAMP) assay in cells expressing MC1R, MC2R, MC3R, MC4R, or MC5R. In vivo corticosterone analyses were performed by measuring plasma corticosterone levels in Sprague Dawley rats. RESULTS Acthar Gel and synthetic MCR agonists exhibited the highest binding at MC1R, lowest binding at MC5R, and moderate binding at MC3R and MC4R. Acthar Gel stimulated the production of cAMP in all 5 MCR-expressing cell lines, with MC2R displaying the lowest level of full agonist activity, 3-, 6.6-, and 10-fold lower than MC1R, MC3R, and MC4R, respectively. Acthar Gel was a partial agonist at MC5R. The synthetic MCR agonists induced full activity at all 5 MCRs, with the exception of α-MSH having no activity at MC2R. Acthar Gel treatment had less of an impact on in vivo production of corticosterone compared with synthetic ACTH1-24 depot. CONCLUSIONS Acthar Gel bound to and activated each MCR tested in this study, with partial agonist activity at MC5R and the lowest level of full agonist activity at MC2R, which distinguished it from synthetic MCR agonists. The minimal activity of Acthar Gel at MC2R corresponded to lower endogenous corticosteroid production.
Collapse
Affiliation(s)
- Y Joyce Huang
- Cellular and Molecular Biology, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | - Karen Galen
- Immunology and Pharmacology, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | - Ben Zweifel
- Immunology and Pharmacology, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | - Leah R Brooks
- Medical Affairs, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | - A Dale Wright
- Immunology and Pharmacology, Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| |
Collapse
|
31
|
Advani D, Gupta R, Tripathi R, Sharma S, Ambasta RK, Kumar P. Protective role of anticancer drugs in neurodegenerative disorders: A drug repurposing approach. Neurochem Int 2020; 140:104841. [PMID: 32853752 DOI: 10.1016/j.neuint.2020.104841] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/24/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
The disease heterogeneity and little therapeutic progress in neurodegenerative diseases justify the need for novel and effective drug discovery approaches. Drug repurposing is an emerging approach that reinvigorates the classical drug discovery method by divulging new therapeutic uses of existing drugs. The common biological background and inverse tuning between cancer and neurodegeneration give weight to the conceptualization of repurposing of anticancer drugs as novel therapeutics. Many studies are available in the literature, which highlights the success story of anticancer drugs as repurposed therapeutics. Among them, kinase inhibitors, developed for various oncology indications evinced notable neuroprotective effects in neurodegenerative diseases. In this review, we shed light on the salient role of multiple protein kinases in neurodegenerative disorders. We also proposed a feasible explanation of the action of kinase inhibitors in neurodegenerative disorders with more attention towards neurodegenerative disorders. The problem of neurotoxicity associated with some anticancer drugs is also highlighted. Our review encourages further research to better encode the hidden potential of anticancer drugs with the aim of developing prospective repurposed drugs with no toxicity for neurodegenerative disorders.
Collapse
Affiliation(s)
- Dia Advani
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rohan Gupta
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rahul Tripathi
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Sudhanshu Sharma
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
32
|
Dergunova LV, Filippenkov IB, Limborska SA, Myasoedov NF. Pharmacotranscriptomics of peptide drugs with neuroprotective properties. Med Res Rev 2020; 41:754-769. [PMID: 32638434 DOI: 10.1002/med.21704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 01/05/2023]
Abstract
Here we present a review of studies on the effects of peptides with neuroprotective properties on gene transcription in nerve cells. The few published works in this area clearly demonstrate massive changes in cell transcriptomes induced by peptides under normal conditions and under conditions of experimental brain ischemia. These changes significantly affect signaling and metabolic pathways, affecting various body systems and confirming the multiple target actions of peptides. The importance of noncoding RNAs in the regulation of these processes is shown, and we discuss the prospects of research for determining the main mechanisms of peptide regulation, which is necessary for the further development of drugs with targeted neuroprotective effects.
Collapse
Affiliation(s)
- Lyudmila V Dergunova
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Ivan B Filippenkov
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Svetlana A Limborska
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Nikolai F Myasoedov
- Department of Chemistry of Physiologically Active Compounds, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
33
|
Novel Insights into the Protective Properties of ACTH (4-7)PGP (Semax) Peptide at the Transcriptome Level Following Cerebral Ischaemia-Reperfusion in Rats. Genes (Basel) 2020; 11:genes11060681. [PMID: 32580520 PMCID: PMC7350263 DOI: 10.3390/genes11060681] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022] Open
Abstract
Cerebral ischaemia is the most common cause of impaired brain function. Biologically active peptides represent potential drugs for reducing the damage that occurs after ischaemia. The synthetic melanocortin derivative, ACTH(4-7)PGP (Semax), has been used successfully in the treatment of patients with severe impairment of cerebral blood circulation. However, its molecular mechanisms of action within the brain are not yet fully understood. Previously, we used the transient middle cerebral artery occlusion (tMCAO) model to study the damaging effects of ischaemia-reperfusion on the brain transcriptome in rats. Here, using RNA-Seq analysis, we investigated the protective properties of the Semax peptide at the transcriptome level under tMCAO conditions. We have identified 394 differentially expressed genes (DEGs) (>1.5-fold change) in the brains of rats at 24 h after tMCAO treated with Semax relative to saline. Following tMCAO, we found that Semax suppressed the expression of genes related to inflammatory processes and activated the expression of genes related to neurotransmission. In contrast, ischaemia-reperfusion alone activated the expression of inflammation-related genes and suppressed the expression of neurotransmission-related genes. Therefore, the neuroprotective action of Semax may be associated with a compensation of mRNA expression patterns that are disrupted during ischaemia-reperfusion conditions.
Collapse
|
34
|
Montero-Melendez T, Nagano A, Chelala C, Filer A, Buckley CD, Perretti M. Therapeutic senescence via GPCR activation in synovial fibroblasts facilitates resolution of arthritis. Nat Commun 2020; 11:745. [PMID: 32029712 PMCID: PMC7005314 DOI: 10.1038/s41467-020-14421-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 12/16/2019] [Indexed: 02/08/2023] Open
Abstract
Rheumatoid arthritis affects individuals commonly during the most productive years of adulthood. Poor response rates and high costs associated with treatment mandate the search for new therapies. Here we show that targeting a specific G-protein coupled receptor promotes senescence in synovial fibroblasts, enabling amelioration of joint inflammation. Following activation of the melanocortin type 1 receptor (MC1), synovial fibroblasts acquire a senescence phenotype characterized by arrested proliferation, metabolic re-programming and marked gene alteration resembling the remodeling phase of wound healing, with increased matrix metalloproteinase expression and reduced collagen production. This biological response is attained by selective agonism of MC1, not shared by non-selective ligands, and dependent on downstream ERK1/2 phosphorylation. In vivo, activation of MC1 leads to anti-arthritic effects associated with induction of senescence in the synovial tissue and cartilage protection. Altogether, selective activation of MC1 is a viable strategy to induce cellular senescence, affording a distinct way to control joint inflammation and arthritis.
Collapse
Affiliation(s)
- Trinidad Montero-Melendez
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK. .,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| | - Ai Nagano
- Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Claude Chelala
- Barts Cancer Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.,Life Sciences Initiative, Queen Mary University of London, London, UK
| | - Andrew Filer
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Institute of Inflammation and Ageing, Birmingham, UK
| | - Christopher D Buckley
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Institute of Inflammation and Ageing, Birmingham, UK.,Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK. .,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| |
Collapse
|
35
|
Xu W, Mo J, Ocak U, Travis ZD, Enkhjargal B, Zhang T, Wu P, Peng J, Li T, Zuo Y, Shao A, Tang J, Zhang J, Zhang JH. Activation of Melanocortin 1 Receptor Attenuates Early Brain Injury in a Rat Model of Subarachnoid Hemorrhage viathe Suppression of Neuroinflammation through AMPK/TBK1/NF-κB Pathway in Rats. Neurotherapeutics 2020; 17:294-308. [PMID: 31486022 PMCID: PMC7007470 DOI: 10.1007/s13311-019-00772-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation plays a vital role in early brain injury (EBI) following subarachnoid hemorrhage (SAH). The hypothesis of this study was that activation of melanocortin 1 receptor (MC1R) with BMS-470539 attenuates EBI by suppression of neuroinflammation after SAH. We utilized BMS-470539, MSG-606, and MRT-68601 to verify the neuroprotective effects of MC1R. We evaluated brain water content, short-term and long-term neurobehavior after SAH. Western blotting and immunofluorescence staining were utilized to assess the changes of protein levels. The results of western blotting suggested that the expressions of MC1R, phosphorylated-adenosine monophosphate-activated protein kinase (p-AMPK), and phosphorylated-TANK binding kinase 1 (p-TBK1) were increased and reached their peak points at 24 h following SAH. Moreover, BMS-470539 treatment notably attenuated neurological deficits caused by SAH, and also notably improved long-term spatial learning and memory abilities after SAH. The underlying mechanisms of the neuroprotection of BMS-470539 involved the suppression of microglia activation, promotion of CD206+ microglia transformation and reduction of neutrophil infiltration by increasing the levels of p-AMPK and p-TBK1 while decreasing the levels of NF-κB, IL-1β, and TNFα. The neuroprotective effects of BMS-470539 were significantly abolished by MSG-606 and MRT-68601. The activation of MC1R with BMS-470539 notably attenuates EBI after SAH by suppression of microglial activation and neutrophil infiltration via the AMPK/TBK1/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Weilin Xu
- Department of Neurosurgery, The second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, Zhejiang, China
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jun Mo
- Department of Neurosurgery, The second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, Zhejiang, China
- Department of Neurosurgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, Zhejiang, China
| | - Umut Ocak
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Zachary D Travis
- Department of Earth and Biological Sciences, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Budbazar Enkhjargal
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Tongyu Zhang
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Pei Wu
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jianhua Peng
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Tao Li
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Yuchun Zuo
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Anwen Shao
- Department of Neurosurgery, The second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, Zhejiang, China
| | - Jiping Tang
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jianmin Zhang
- Department of Neurosurgery, The second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, Zhejiang, China.
- Brain Research Institute, Zhejiang University, Hangzhou, 310009, Zhejiang, China.
| | - John H Zhang
- Department of Physiology & Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA.
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA.
- Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA.
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus St, Loma Linda, CA, 92354, USA.
| |
Collapse
|
36
|
NDP-MSH binding melanocortin-1 receptor ameliorates neuroinflammation and BBB disruption through CREB/Nr4a1/NF-κB pathway after intracerebral hemorrhage in mice. J Neuroinflammation 2019; 16:192. [PMID: 31660977 PMCID: PMC6816206 DOI: 10.1186/s12974-019-1591-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 09/20/2019] [Indexed: 12/22/2022] Open
Abstract
Background Neuroinflammation and blood-brain barrier (BBB) disruption are two vital mechanisms of secondary brain injury following intracerebral hemorrhage (ICH). Recently, melanocortin-1 receptor (Mc1r) activation by Nle4-D-Phe7-α-MSH (NDP-MSH) was shown to play a neuroprotective role in an experimental autoimmune encephalomyelitis (EAE) mouse model. This study aimed to investigate whether NDP-MSH could alleviate neuroinflammation and BBB disruption after experimental ICH, as well as the potential mechanisms of its neuroprotective roles. Methods Two hundred and eighteen male C57BL/6 mice were subjected to autologous blood-injection ICH model. NDP-MSH, an agonist of Mc1r, was administered intraperitoneally injected at 1 h after ICH insult. To further explore the related protective mechanisms, Mc1r small interfering RNA (Mc1r siRNA) and nuclear receptor subfamily 4 group A member 1 (Nr4a1) siRNA were administered via intracerebroventricular (i.c.v) injection before ICH induction. Neurological test, BBB permeability, brain water content, immunofluorescence staining, and Western blot analysis were implemented. Results The Expression of Mc1r was significantly increased after ICH. Mc1r was mainly expressed in microglia, astrocytes, and endothelial cells following ICH. Treatment with NDP-MSH remarkably improved neurological function and reduced BBB disruption, brain water content, and the number of microglia in the peri-hematoma tissue after ICH. Meanwhile, the administration of NDP-MSH significantly reduced the expression of p-NF-κB p65, IL-1β, TNF-α, and MMP-9 and increased the expression of p-CREB, Nr4a1, ZO-1, occludin, and Lama5. Inversely, the knockdown of Mc1r or Nr4a1 abolished the neuroprotective effects of NDP-MSH. Conclusions Taken together, NDP-MSH binding Mc1r attenuated neuroinflammation and BBB disruption and improved neurological deficits, at least in part through CREB/Nr4a1/NF-κB pathway after ICH.
Collapse
|
37
|
Kamermans A, Verhoeven T, van Het Hof B, Koning JJ, Borghuis L, Witte M, van Horssen J, de Vries HE, Rijnsburger M. Setmelanotide, a Novel, Selective Melanocortin Receptor-4 Agonist Exerts Anti-inflammatory Actions in Astrocytes and Promotes an Anti-inflammatory Macrophage Phenotype. Front Immunol 2019; 10:2312. [PMID: 31636637 PMCID: PMC6788433 DOI: 10.3389/fimmu.2019.02312] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/12/2019] [Indexed: 11/13/2022] Open
Abstract
To date, available treatment strategies for multiple sclerosis (MS) are ineffective in preventing or reversing progressive neurologic deterioration, creating a high, and unmet medical need. One potential way to fight MS may be by limiting the detrimental effects of reactive astrocytes, a key pathological hallmark for disease progression. One class of compounds that may exert beneficial effects via astrocytes are melanocortin receptor (MCR) agonists. Among the MCR, MC4R is most abundantly expressed in the CNS and several rodent studies have described that MC4R is—besides neurons—expressed by astrocytes. Activation of MC4R in astrocytes has shown to have potent anti-inflammatory as well as neuroprotective effects in vitro, suggesting that this could be a potential target to ameliorate ongoing inflammation, and neurodegeneration in MS. In this study, we set out to investigate human MC4R expression and analyze its downstream effects. We identified MC4R mRNA and protein to be expressed on astrocytes and observed increased astrocytic MC4R expression in active MS lesions. Furthermore, we show that the novel, highly selective MC4R agonist setmelanotide ameliorates the reactive phenotype in astrocytes in vitro and markedly induced interleukin−6 and −11 production, possibly through enhanced cAMP response element-binding protein (CREB) phosphorylation. Notably, stimulation of human macrophages with medium from astrocytes that were exposed to setmelanotide, skewed macrophages toward an anti-inflammatory phenotype. Taken together, these findings suggest that targeting MC4R on astrocytes might be a novel therapeutic strategy to halt inflammation-associated neurodegeneration in MS.
Collapse
Affiliation(s)
- Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Tom Verhoeven
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Bert van Het Hof
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jasper J Koning
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Lauri Borghuis
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Maarten Witte
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Merel Rijnsburger
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
38
|
Imaging in mice and men: Pathophysiological insights into multiple sclerosis from conventional and advanced MRI techniques. Prog Neurobiol 2019; 182:101663. [PMID: 31374243 DOI: 10.1016/j.pneurobio.2019.101663] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/17/2019] [Accepted: 07/17/2019] [Indexed: 01/16/2023]
Abstract
Magnetic resonance imaging (MRI) is the most important tool for diagnosing multiple sclerosis (MS). However, MRI is still unable to precisely quantify the specific pathophysiological processes that underlie imaging findings in MS. Because autopsy and biopsy samples of MS patients are rare and biased towards a chronic burnt-out end or fulminant acute early stage, the only available methods to identify human disease pathology are to apply MRI techniques in combination with subsequent histopathological examination to small animal models of MS and to transfer these insights to MS patients. This review summarizes the existing combined imaging and histopathological studies performed in MS mouse models and humans with MS (in vivo and ex vivo), to promote a better understanding of the pathophysiology that underlies conventional MRI, diffusion tensor and magnetization transfer imaging findings in MS patients. Moreover, it provides a critical view on imaging capabilities and results in MS patients and mouse models and for future studies recommends how to combine those particular MR sequences and parameters whose underlying pathophysiological basis could be partly clarified. Further combined longitudinal in vivo imaging and histopathological studies on rationally selected, appropriate mouse models are required.
Collapse
|
39
|
Jang EA, Kim JY, Tin TD, Song JA, Lee SH, Kwak SH. The effects of BMS-470539 on lipopolysaccharide-induced acute lung injury. Acute Crit Care 2019; 34:133-140. [PMID: 31723918 PMCID: PMC6786663 DOI: 10.4266/acc.2019.00507] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 12/12/2022] Open
Abstract
Background Overactivation of inflammatory cells, including macrophages and neutrophils, is associated with acute lung injury. BMS-470539 is a selective agonist of melanocortin 1 receptor, which triggers the inhibition of proinflammatory responses, suppressing neutrophil infiltration and protecting tissue. This study evaluated the effects of BMS-470539 on lipopolysaccharide-induced acute lung injury in a mouse model. Methods Mice received a subcutaneous injection of saline or BMS-470539 (18.47 mg/kg) 1 hour before an intratracheal injection of saline or lipopolysaccharide (20 μg). Mice were sacrificed to analyze the severity of pulmonary edema (lung wet-to-dry weight [W/D] ratio) and inflammatory responses (level of leukocytes, polymorphonuclear neutrophils [PMNs] and tumor necrosis factor alpha [TNF-α] in bronchoalveolar lavage fluid [BALF]), and neutrophil infiltration (myeloperoxidase activity). TNF-α activation was also measured in neutrophils from bone marrow. Survival was investigated in a second-hit sepsis mouse model. Results BMS-470539 improved sepsis-induced pulmonary edema, as demonstrated by a decreased W/D ratio (5.76%±0.83% to 3.81%±0.86%, P<0.05). The inflammatory response also improved, as shown by decreased levels of leukocytes (551±116 to 357±86×10²/mm³, P<0.05), PMNs (51.52%±16.23% to 18.41%±7.25%, P<0.01), and TNF-α (550±338 to 128±52 pg/ml, P<0.01) in the BALF. BMS-470539 also improved the inflammatory response, as shown by TNF-α levels (850±158 to 423±59 pg/ml, P<0.01) in neutrophils. BMS-470539 downregulated neutrophil infiltration in the lung (myeloperoxidase: 654±98 to 218±89 U/g, P<0.001). Lastly, BMS improved the survival rate (0% to 70%, P<0.01) in a mice multiple organ failure model. Conclusions BMS-470539 improved lipopolysaccharide-induced acute lung injury and mortality in mice by affecting the inflammatory response.
Collapse
Affiliation(s)
- Eun-A Jang
- Department of Anesthesiology and Pain Medicine, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Jin-Young Kim
- Department of Anesthesiology and Pain Medicine, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Tran Duc Tin
- Brain Korea 21 Project, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
| | - Ji-A Song
- Department of Anesthesiology and Pain Medicine, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Seong-Heon Lee
- Department of Anesthesiology and Pain Medicine, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Sang-Hyun Kwak
- Department of Anesthesiology and Pain Medicine, Chonnam National University Medical School and Hospital, Gwangju, Korea.,Brain Korea 21 Project, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
| |
Collapse
|
40
|
Mehta D, Granstein RD. Immunoregulatory Effects of Neuropeptides on Endothelial Cells: Relevance to Dermatological Disorders. Dermatology 2019; 235:175-186. [PMID: 30808842 DOI: 10.1159/000496538] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/31/2018] [Indexed: 11/19/2022] Open
Abstract
Many skin diseases, including psoriasis and atopic dermatitis, have a neurogenic component. In this regard, bidirectional interactions between components of the nervous system and multiple target cells in the skin and elsewhere have been receiving increasing attention. Neuropeptides released by sensory nerves that innervate the skin can directly modulate functions of keratinocytes, Langerhans cells, dermal dendritic cells, mast cells, dermal microvascular endothelial cells and infiltrating immune cells. As a result, neuropeptides and neuropeptide receptors participate in a complex, interdependent network of mediators that modulate the skin immune system, skin inflammation, and wound healing. In this review, we will focus on recent studies demonstrating the roles of α-melanocyte-stimulating hormone, calcitonin gene-related peptide, substance P, somatostatin, vasoactive intestinal peptide, pituitary adenylate cyclase-activating peptide, and nerve growth factor in modulating inflammation and immunity in the skin through their effects on dermal microvascular endothelial cells.
Collapse
Affiliation(s)
- Devina Mehta
- Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| | | |
Collapse
|
41
|
Böhm M, Luger T. Are melanocortin peptides future therapeutics for cutaneous wound healing? Exp Dermatol 2019; 28:219-224. [PMID: 30661264 DOI: 10.1111/exd.13887] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 10/29/2018] [Accepted: 11/20/2018] [Indexed: 12/11/2022]
Abstract
Cutaneous wound healing is a complex process divided into different phases, that is an inflammatory, proliferative and remodelling phase. During these phases, a variety of resident skin cell types but also cells of the immune system orchestrate the healing process. In the last year, it has been shown that the majority of cutaneous cell types express the melanocortin 1 receptor (MC1R) that binds α-melanocyte-stimulating hormone (α-MSH) with high affinity and elicits pleiotropic biological effects, for example modulation of inflammation and immune responses, cytoprotection, antioxidative defense and collagen turnover. Truncated α-MSH peptides such as Lys-Pro-Val (KPV) as well as derivatives like Lys-d-Pro-Thr (KdPT), the latter containing the amino acid sequence 193-195 of interleukin-1β, have been found to possess anti-inflammatory effects but to lack the pigment-inducing activity of α-MSH. We propose here that such peptides are promising future candidates for the treatment of cutaneous wounds and skin ulcers. Experimental approaches in silico, in vitro, ex vivo and in animal models are outlined. This is followed by an unbiased discussion of the pro and contra arguments of such peptides as future candidates for the therapeutic management of cutaneous wounds and a review of the so-far available data on melanocortin peptides and derivatives in wound healing.
Collapse
Affiliation(s)
- Markus Böhm
- Department of Dermatology, Laboratory for Neuroendocrinology of the Skin and Interdisciplinary Endocrinology, University of Münster, Münster, Germany
| | - Thomas Luger
- Department of Dermatology, Laboratory for Neuroendocrinology of the Skin and Interdisciplinary Endocrinology, University of Münster, Münster, Germany
| |
Collapse
|
42
|
Tangherlini G, Kalinin DV, Schepmann D, Che T, Mykicki N, Ständer S, Loser K, Wünsch B. Development of Novel Quinoxaline-Based κ-Opioid Receptor Agonists for the Treatment of Neuroinflammation. J Med Chem 2018; 62:893-907. [PMID: 30543421 DOI: 10.1021/acs.jmedchem.8b01609] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Giovanni Tangherlini
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Dmitrii V. Kalinin
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Tao Che
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Nadine Mykicki
- Department of Dermatology, University of Münster, von-Esmarch-Street 58, D-48149 Münster, Germany
- CRC1009 Breaking Barriers and CRC-TR 128 Multiple Sclerosis, University of Münster, D-48149 Münster, Germany
| | - Sonja Ständer
- Department of Dermatology, University of Münster, von-Esmarch-Street 58, D-48149 Münster, Germany
| | - Karin Loser
- Department of Dermatology, University of Münster, von-Esmarch-Street 58, D-48149 Münster, Germany
- CRC1009 Breaking Barriers and CRC-TR 128 Multiple Sclerosis, University of Münster, D-48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| |
Collapse
|
43
|
Lee MW, Lee EY, Ferguson AL, Wong GCL. Machine learning antimicrobial peptide sequences: Some surprising variations on the theme of amphiphilic assembly. Curr Opin Colloid Interface Sci 2018; 38:204-213. [PMID: 31093008 DOI: 10.1016/j.cocis.2018.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Antimicrobial peptides (AMPs) collectively constitute a key component of the host innate immune system. They span a diverse space of sequences and can be α-helical, β-sheet, or unfolded in structure. Despite a wealth of knowledge about them from decades of experiments, it remains difficult to articulate general principles governing such peptides. How are they different from other molecules that are also cationic and amphiphilic? What other functions, in immunity and otherwise, are enabled by these simple sequences? In this short review, we present some recent work that engages these questions using methods not usually applied to AMP studies, such as machine learning. We find that not only do AMP-like sequences confer membrane remodeling activity to an unexpectedly broad range of protein classes, their cationic and amphiphilic signature also allows them to act as meta-antigens and self-assemble with immune ligands into nanocrystalline complexes for multivalent presentation to Toll-like receptors.
Collapse
Affiliation(s)
- Michelle W Lee
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, CA 90095, United States
| | - Ernest Y Lee
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, CA 90095, United States
| | - Andrew L Ferguson
- Institute for Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, United States
| | - Gerard C L Wong
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, CA 90095, United States
| |
Collapse
|
44
|
|
45
|
Chen Z, Chen S, Liu J. The role of T cells in the pathogenesis of Parkinson's disease. Prog Neurobiol 2018; 169:1-23. [PMID: 30114440 DOI: 10.1016/j.pneurobio.2018.08.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/24/2018] [Accepted: 08/12/2018] [Indexed: 02/06/2023]
Abstract
Recent evidence has shown that neuroinflammation plays a key role in the pathogenesis of Parkinson's disease (PD). However, different components of the brain's immune system may exert diverse effects on neuroinflammatory events in PD. The adaptive immune response, especially the T cell response, can trigger type 1 pro-inflammatory activities and suppress type 2 anti-inflammatory activities, eventually resulting in deregulated neuroinflammation and subsequent dopaminergic neurodegeneration. Additionally, studies have increasingly shown that therapies targeting T cells can alleviate neurodegeneration and motor behavior impairment in animal models of PD. Therefore, we conclude that abnormal T cell-mediated immunity is a fundamental pathological process that may be a promising translational therapeutic target for Parkinson's disease.
Collapse
Affiliation(s)
- Zhichun Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated with the Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated with the Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated with the Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
46
|
Breuer J, Loser K, Mykicki N, Wiendl H, Schwab N. Does the environment influence multiple sclerosis pathogenesis via UVB light and/or induction of vitamin D? J Neuroimmunol 2018; 329:1-8. [PMID: 29793727 DOI: 10.1016/j.jneuroim.2018.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 05/17/2018] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis (MS) is a disease of presumed auto-immune origin. Long-standing observations such as the correlation between MS incidence and geographical latitude or the levels of Vitamin D (Vit D) in the serum have implicated the environmental factors UVB radiation and diet in the etiology of the disease. Clinical trials have been conducted and are currently underway to elucidate whether a Vit D enriched diet or treatment with UVB can influence MS incidence, -severity, and -progression, as well as the ideal time point for treatment. This review summarizes the current scientific knowledge to the environmental factors UVB-light and Vit D concerning the clinical aspects of MS in epidemiological studies and clinical trials.
Collapse
Affiliation(s)
- Johanna Breuer
- Department of Neurology, University of Münster, Münster, Germany
| | - Karin Loser
- Department of Dermatology, University of Münster, Münster, Germany
| | - Nadine Mykicki
- Department of Dermatology, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | - Nicholas Schwab
- Department of Neurology, University of Münster, Münster, Germany.
| |
Collapse
|
47
|
Melanocortin receptor subtypes are expressed on cells in the oligodendroglial lineage and signal ACTH protection. J Neurosci Res 2017; 96:427-435. [PMID: 28877366 DOI: 10.1002/jnr.24141] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 07/15/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023]
|
48
|
Melanocortins, Melanocortin Receptors and Multiple Sclerosis. Brain Sci 2017; 7:brainsci7080104. [PMID: 28805746 PMCID: PMC5575624 DOI: 10.3390/brainsci7080104] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 02/07/2023] Open
Abstract
The melanocortins and their receptors have been extensively investigated for their roles in the hypothalamo-pituitary-adrenal axis, but to a lesser extent in immune cells and in the nervous system outside the hypothalamic axis. This review discusses corticosteroid dependent and independent effects of melanocortins on the peripheral immune system, central nervous system (CNS) effects mediated through neuronal regulation of immune system function, and direct effects on endogenous cells in the CNS. We have focused on the expression and function of melanocortin receptors in oligodendroglia (OL), the myelin producing cells of the CNS, with the goal of identifying new therapeutic approaches to decrease CNS damage in multiple sclerosis as well as to promote repair. It is clear that melanocortin signaling through their receptors in the CNS has potential for neuroprotection and repair in diseases like MS. Effects of melanocortins on the immune system by direct effects on the circulating cells (lymphocytes and monocytes) and by signaling through CNS cells in regions lacking a mature blood brain barrier are clear. However, additional studies are needed to develop highly effective MCR targeted therapies that directly affect endogenous cells of the CNS, particularly OL, their progenitors and neurons.
Collapse
|
49
|
Luger TA, Loser K. Novel insights into the pathogenesis of psoriasis. Clin Immunol 2017; 186:43-45. [PMID: 28736273 DOI: 10.1016/j.clim.2017.07.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 07/19/2017] [Indexed: 12/21/2022]
Affiliation(s)
- T A Luger
- Dept. of Dermatology, University of Münster, Münster, Germany.
| | - K Loser
- Dept. of Dermatology, University of Münster, Münster, Germany
| |
Collapse
|
50
|
Orellana JA, Cerpa W, Carvajal MF, Lerma-Cabrera JM, Karahanian E, Osorio-Fuentealba C, Quintanilla RA. New Implications for the Melanocortin System in Alcohol Drinking Behavior in Adolescents: The Glial Dysfunction Hypothesis. Front Cell Neurosci 2017; 11:90. [PMID: 28424592 PMCID: PMC5380733 DOI: 10.3389/fncel.2017.00090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/15/2017] [Indexed: 12/12/2022] Open
Abstract
Alcohol dependence causes physical, social, and moral harms and currently represents an important public health concern. According to the World Health Organization (WHO), alcoholism is the third leading cause of death worldwide, after tobacco consumption and hypertension. Recent epidemiologic studies have shown a growing trend in alcohol abuse among adolescents, characterized by the consumption of large doses of alcohol over a short time period. Since brain development is an ongoing process during adolescence, short- and long-term brain damage associated with drinking behavior could lead to serious consequences for health and wellbeing. Accumulating evidence indicates that alcohol impairs the function of different components of the melanocortin system, a major player involved in the consolidation of addictive behaviors during adolescence and adulthood. Here, we hypothesize the possible implications of melanocortins and glial cells in the onset and progression of alcohol addiction. In particular, we propose that alcohol-induced decrease in α-MSH levels may trigger a cascade of glial inflammatory pathways that culminate in altered gliotransmission in the ventral tegmental area and nucleus accumbens (NAc). The latter might potentiate dopaminergic drive in the NAc, contributing to increase the vulnerability to alcohol dependence and addiction in the adolescence and adulthood.
Collapse
Affiliation(s)
- Juan A Orellana
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Laboratorio de Neurociencias, Departamento de Neurología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de ChileSantiago, Chile
| | - Waldo Cerpa
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiago, Chile
| | - Maria F Carvajal
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Unidad de Neurociencia, Centro de Investigación Biomédica, Universidad Autónoma de ChileSantiago, Chile
| | - José M Lerma-Cabrera
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Unidad de Neurociencia, Centro de Investigación Biomédica, Universidad Autónoma de ChileSantiago, Chile
| | - Eduardo Karahanian
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Unidad de Neurociencia, Centro de Investigación Biomédica, Universidad Autónoma de ChileSantiago, Chile
| | - Cesar Osorio-Fuentealba
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Facultad de Kinesiología, Artes y Educación Física, Universidad Metropolitana de Ciencias de la EducaciónSantiago, Chile
| | - Rodrigo A Quintanilla
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Laboratory of Neurodegenerative Diseases, Universidad Autónoma de ChileSantiago, Chile
| |
Collapse
|