1
|
Chatanaka MK, Sohaei D, Diamandis EP, Prassas I. Beyond the amyloid hypothesis: how current research implicates autoimmunity in Alzheimer's disease pathogenesis. Crit Rev Clin Lab Sci 2023; 60:398-426. [PMID: 36941789 DOI: 10.1080/10408363.2023.2187342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
The amyloid hypothesis has so far been at the forefront of explaining the pathogenesis of Alzheimer's Disease (AD), a progressive neurodegenerative disorder that leads to cognitive decline and eventual death. Recent evidence, however, points to additional factors that contribute to the pathogenesis of this disease. These include the neurovascular hypothesis, the mitochondrial cascade hypothesis, the inflammatory hypothesis, the prion hypothesis, the mutational accumulation hypothesis, and the autoimmunity hypothesis. The purpose of this review was to briefly discuss the factors that are associated with autoimmunity in humans, including sex, the gut and lung microbiomes, age, genetics, and environmental factors. Subsequently, it was to examine the rise of autoimmune phenomena in AD, which can be instigated by a blood-brain barrier breakdown, pathogen infections, and dysfunction of the glymphatic system. Lastly, it was to discuss the various ways by which immune system dysregulation leads to AD, immunomodulating therapies, and future directions in the field of autoimmunity and neurodegeneration. A comprehensive account of the recent research done in the field was extracted from PubMed on 31 January 2022, with the keywords "Alzheimer's disease" and "autoantibodies" for the first search input, and "Alzheimer's disease" with "IgG" for the second. From the first search, 19 papers were selected, because they contained recent research on the autoantibodies found in the biofluids of patients with AD. From the second search, four papers were selected. The analysis of the literature has led to support the autoimmune hypothesis in AD. Autoantibodies were found in biofluids (serum/plasma, cerebrospinal fluid) of patients with AD with multiple methods, including ELISA, Mass Spectrometry, and microarray analysis. Through continuous research, the understanding of the synergistic effects of the various components that lead to AD will pave the way for better therapeutic methods and a deeper understanding of the disease.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Dorsa Sohaei
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
- Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| |
Collapse
|
2
|
Giszas B, Reuken PA, Katzer K, Kiehntopf M, Schmerler D, Rummler S, Stallmach A, Klink A. Immunoadsorption to treat patients with severe post-COVID syndrome. Ther Apher Dial 2023; 27:790-801. [PMID: 36719401 DOI: 10.1111/1744-9987.13974] [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: 09/04/2022] [Revised: 11/25/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Following SARS-CoV-2-infection up to 21% of patients will develop post-COVID-syndrome. Autoantibodies (AAbs) targeting neuronal-ß-adrenergic and muscarinic receptors may provide crucial contributions to the pathophysiology of this condition. Immunoadsorption (IA) has been identified as an effective means of removing AAbs and has resulted in clinical improvements of other autoantibody-associated diseases. METHODS We determined AAb-levels (anti-ß1/ß2 and anti-M3/M4 receptor) in 178 patients diagnosed with post-COVID-syndrome and described the clinical courses of two patients with elevated AAb-levels that underwent IA-treatment. RESULTS AAbs were detected in 57% (101/178) of patients diagnosed with post-COVID-syndrome. Substantial reductions in AAb-levels and clinical remission were achieved in one of two patients who was treated with IA. However, this patient relapsed within 6 weeks with a concomitant increase in AAb-levels. CONCLUSION Collectively, AAbs may play a pathophysiologic role in post-COVID and their removal provide transient benefits in some patients. However, these findings should be further investigated in randomized-controlled-trials.
Collapse
Affiliation(s)
- Benjamin Giszas
- Department of Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Philipp A Reuken
- Department of Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Katrin Katzer
- Department of Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Michael Kiehntopf
- Institute of Clinical Chemistry and Laboratory Diagnostics and Integrated Biobank Jena (IBBJ), Jena University Hospital-Friedrich Schiller University, Jena, Germany
| | - Diana Schmerler
- Institute of Clinical Chemistry and Laboratory Diagnostics and Integrated Biobank Jena (IBBJ), Jena University Hospital-Friedrich Schiller University, Jena, Germany
| | - Silke Rummler
- Institute for Transfusion Medicine, University Hospital Jena, Jena, Germany
| | - Andreas Stallmach
- Department of Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Anne Klink
- Institute for Transfusion Medicine, University Hospital Jena, Jena, Germany
| |
Collapse
|
3
|
Cognitive Impairment, Sleep Disturbance, and Depression in Women with Silicone Breast Implants: Association with Autoantibodies against Autonomic Nervous System Receptors. Biomolecules 2022; 12:biom12060776. [PMID: 35740901 PMCID: PMC9221347 DOI: 10.3390/biom12060776] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Silicone breast implants (SBIs) has been shown to be associated with an increased risk of autoimmune diseases. In the current study, we aimed to explore the potential association between circulating autoantibodies against the autonomic nervous system and cognitive impairment, memory deficit, and depressive symptoms reported by women with SBIs. Methods: ELISA assays were used to quantify anti-adrenergic receptors (α1, α2, β1, β2), anti-muscarinic receptors (M1-M5), anti-endothelin receptor type A, and anti-angiotensin II type 1 receptor titers in the sera of 93 symptomatic female subjects with SBIs and 36 age-matched healthy female controls. Results: A significant difference was detected in the level of autoantibodies against the autonomic nervous system receptors in women with SBIs who reported memory impairment, cognitive impairment, and sleep disturbance as compared with both women with SBIs who did not complain of these symptoms or with healthy individuals without SBIs. Conclusions: Clinical symptoms such as depression, cognitive impairment, and sleep disturbances were found to be associated with dysregulation of the levels of circulating autoantibodies targeting the autonomous nervous system receptors in women with SBIs. These autoantibodies may have diagnostic significance in diseases associated with breast implants.
Collapse
|
4
|
Benmelouka AY, Ouerdane Y, Outani O, Alnasser YT, Alghamdi BS, Perveen A, Ashraf GM, Ebada MA. Alzheimer's Disease-Related Psychosis: An Overview of Clinical Manifestations, Pathogenesis, and Current Treatment. Curr Alzheimer Res 2022; 19:285-301. [PMID: 35440308 DOI: 10.2174/1567205019666220418151914] [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: 01/20/2022] [Revised: 02/25/2022] [Accepted: 03/06/2022] [Indexed: 11/22/2022]
Abstract
Behavioral and psychotic manifestations, including aggression, delusions, and hallucinations, are frequent comorbidities in patients with debilitating nervous illnesses such as Alzheimer's disease (AD), Amyotrophic Lateral Sclerosis, Multiple Sclerosis, and Parkinson's disease. AD-related psychosis may be linked to a poor disease prognosis, highlighting that early detection and management are mandatory. The manifestations are variable and may be very heterogeneous, imposing a real diagnostic issue. Some assessment tools such as BEHAVE-AD, CERAD-BRSD, and the Psycho-Sensory Hallucinations Scale have been designed to facilitate the diagnosis. The mechanisms behind neurodegeneration-related psychosis are complex and are not fully understood, imposing a burden on researchers to find appropriate management modalities. Familial history and some genetic disturbances may have a determinant role in these delusions and hallucinations in cases with AD. The loss of neuronal cells, atrophy in some regions of the central nervous, and synaptic dysfunction may also contribute to these comorbidities. Furthermore, inflammatory disturbances triggered by pro-inflammatory agents such as interleukins and tumor necrosis factors are stratified among the potential risk factors of the onset of numerous psychotic symptoms in Alzheimer's patients. Little is known about the possible management tools; therefore, it is urgent to conduct well-designed trials to investigate pharmacological and non-pharmacological interventions that can improve the care process of these patients. This review summarizes the current findings regarding the AD-related psychosis symptoms, pathological features, assessment, and management.
Collapse
Affiliation(s)
| | | | - Oumaima Outani
- Faculty of Medicine and Pharmacy of Rabat, Mohammed 5 University
| | | | - Badrah S Alghamdi
- Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah.,Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah
| | - Asma Perveen
- Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah.,Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah
| | - Mahmoud Ahmed Ebada
- Faculty of Medicine, Zagazig University, Zagazig, Al-Sharkia.,Internal Medicine Resident, Ministry of Health and Population of Egypt, Cairo
| |
Collapse
|
5
|
Yu ZY, Yi X, Wang YR, Zeng GH, Tan CR, Cheng Y, Sun PY, Liu ZH, Wang YJ, Liu YH. Inhibiting α1-adrenergic receptor signaling pathway ameliorates AD-type pathologies and behavioral deficits in APPswe/PS1 mouse model. J Neurochem 2022; 161:293-307. [PMID: 35244207 DOI: 10.1111/jnc.15603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 01/25/2022] [Accepted: 02/23/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Zhong-Yuan Yu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xu Yi
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ye-Ran Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Cheng-Rong Tan
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Cheng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Pu-Yang Sun
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zhi-Hao Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Hui Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
6
|
Auler N, Tonner H, Pfeiffer N, Grus FH. Antibody and Protein Profiles in Glaucoma: Screening of Biomarkers and Identification of Signaling Pathways. BIOLOGY 2021; 10:biology10121296. [PMID: 34943212 PMCID: PMC8698915 DOI: 10.3390/biology10121296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary Glaucoma is a chronic eye disease that is one of the leading causes of blindness worldwide. Currently, the only therapeutic option is to lower intraocular pressure. The onset of the disease is often delayed because patients do not notice visual impairment until very late, which is why glaucoma is also known as “the silent thief of sight”. Therefore, early detection and definition of specific markers, the so-called biomarkers, are immensely important. For the methodical implementation, high-throughput methods and omic-based methods came more and more into focus. Thus, interesting targets for possible biomarkers were already suggested by clinical research and basic research, respectively. This review article aims to join the findings of the two disciplines by collecting overlaps as well as differences in various clinical studies and to shed light on promising candidates concerning findings from basic research, facilitating conclusions on possible therapy options. Abstract Glaucoma represents a group of chronic neurodegenerative diseases, constituting the second leading cause of blindness worldwide. To date, chronically elevated intraocular pressure has been identified as the main risk factor and the only treatable symptom. However, there is increasing evidence in the recent literature that IOP-independent molecular mechanisms also play an important role in the progression of the disease. In recent years, it has become increasingly clear that glaucoma has an autoimmune component. The main focus nowadays is elucidating glaucoma pathogenesis, finding early diagnostic options and new therapeutic approaches. This review article summarizes the impact of different antibodies and proteins associated with glaucoma that can be detected for example by microarray and mass spectrometric analyzes, which (i) provide information about expression profiles and associated molecular signaling pathways, (ii) can possibly be used as a diagnostic tool in future and, (iii) can identify possible targets for therapeutic approaches.
Collapse
|
7
|
Lao K, Zhang R, Luan J, Zhang Y, Gou X. Therapeutic Strategies Targeting Amyloid-β Receptors and Transporters in Alzheimer's Disease. J Alzheimers Dis 2021; 79:1429-1442. [PMID: 33459712 DOI: 10.3233/jad-200851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease that has been recognized as one of the most intractable medical problems with heavy social and economic costs. Amyloid-β (Aβ) has been identified as a major factor that participates in AD progression through its neurotoxic effects. The major mechanism of Aβ-induced neurotoxicity is by interacting with membrane receptors and subsequent triggering of aberrant cellular signaling. Besides, Aβ transporters also plays an important role by affecting Aβ homeostasis. Thus, these Aβ receptors and transporters are potential targets for the development of AD therapies. Here, we summarize the reported therapeutic strategies targeting Aβ receptors and transporters to provide a molecular basis for future rational design of anti-AD agents.
Collapse
Affiliation(s)
- Kejing Lao
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, PR China
| | - Ruisan Zhang
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, PR China
| | - Jing Luan
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, PR China
| | - Yuelin Zhang
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, PR China
| | - Xingchun Gou
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, PR China
| |
Collapse
|
8
|
Perez DM. Current Developments on the Role of α 1-Adrenergic Receptors in Cognition, Cardioprotection, and Metabolism. Front Cell Dev Biol 2021; 9:652152. [PMID: 34113612 PMCID: PMC8185284 DOI: 10.3389/fcell.2021.652152] [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: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
The α1-adrenergic receptors (ARs) are G-protein coupled receptors that bind the endogenous catecholamines, norepinephrine, and epinephrine. They play a key role in the regulation of the sympathetic nervous system along with β and α2-AR family members. While all of the adrenergic receptors bind with similar affinity to the catecholamines, they can regulate different physiologies and pathophysiologies in the body because they couple to different G-proteins and signal transduction pathways, commonly in opposition to one another. While α1-AR subtypes (α1A, α1B, α1C) have long been known to be primary regulators of vascular smooth muscle contraction, blood pressure, and cardiac hypertrophy, their role in neurotransmission, improving cognition, protecting the heart during ischemia and failure, and regulating whole body and organ metabolism are not well known and are more recent developments. These advancements have been made possible through the development of transgenic and knockout mouse models and more selective ligands to advance their research. Here, we will review the recent literature to provide new insights into these physiological functions and possible use as a therapeutic target.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
9
|
Hohberger B, Hosari S, Wallukat G, Kunze R, Krebs J, Müller M, Hennig T, Lämmer R, Horn F, Muñoz LE, Herrmann M, Mardin C. Agonistic autoantibodies against ß2-adrenergic receptor influence retinal microcirculation in glaucoma suspects and patients. PLoS One 2021; 16:e0249202. [PMID: 33961631 PMCID: PMC8104926 DOI: 10.1371/journal.pone.0249202] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/14/2021] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Agonistic β2-adrenergic receptor autoantibodies (β2-agAAb) have been observed in sera of patients with ocular hypertension and open-angle glaucoma (OAG). They target the β2-receptors on trabecular meshwork, ciliary body and pericytes (Junemann et al. 2018; Hohberger et al. 2019). In addition to their influence on the intraocular pressure, an association to retinal microcirculation is discussed. This study aimed to investigate foveal avascular zone (FAZ) characteristics by en face OCT angiography (OCT-A) in glaucoma suspects and its relationship to β2-agAAb status in patients with OAG. MATERIAL AND METHODS Thirty-four patients (28 OAG, 6 glaucoma suspects) underwent standardized, clinical examination including sensory testing as white-on-white perimetry (Octopus G1, mean defect, MD) and structural measures as retinal nerve fibre layer (RNFL) thickness, neuroretinal rim width (BMO-MRW), retinal ganglion cell layer (RGCL) thickness, and inner nuclear layer (INL) thickness with high-resolution OCT. FAZ characteristics were measured by OCT-A scans of superficial vascular plexus (SVP), intermediate capillary plexus (ICP), and deep capillary plexus (DCP). FAZ-R was calculated (area FAZ (SVP)/area FAZ (ICP)). Using cardiomyocyte bioassays we analysed serum samples for the presence of β2-agAAb. RESULTS (I) Total mean FAZ area [mm2]: 0.34±0.16 (SVP), 0.24±0.12 (ICP), and 0.49±0.24 (DCP); mean FAZ-R 1.58±0.94. No correlation was seen for FAZ-R with MD, RNFL, BMO-MRW, RGCL thickness and INL thickness (p>0.05). (II) ß2-agAAb have been observed in 91% patients and showed no correlation with MD, RNFL, BMO-MRW, RGCL thickness and INL thickness (p>0.05). (III) FAZ-R correlated significantly with the β2-agAAb-induced increase of the beat rate of cardiomyocyte (p = 0.028). CONCLUSION FAZ characteristics did not correlate with any glaucoma associated functional and morphometric follow-up parameter in the present cohort. However, level of β2-agAAb showed a significantly correlation with FAZ-ratio. We conclude that β2-agAAb might be a novel biomarker in glaucoma pathogenesis showing association to FAZ-ratio with OCT-A.
Collapse
Affiliation(s)
- Bettina Hohberger
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Sami Hosari
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gerd Wallukat
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Rudolf Kunze
- Science Office, Berlin-Buch, Campus Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Johann Krebs
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Meike Müller
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Till Hennig
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Lämmer
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Folkert Horn
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Luis E. Muñoz
- Department of Internal Medicine 3—Rheumatology and Immunology, University of Erlangen-Nürnberg, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3—Rheumatology and Immunology, University of Erlangen-Nürnberg, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Germany
| | - Christian Mardin
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
10
|
Tian X, Qin Y, Tian Y, Ge X, Cui J, Han H, Liu L, Yu H. Identification of vascular dementia and Alzheimer's disease hub genes expressed in the frontal lobe and temporal cortex by weighted co-expression network analysis and construction of a protein-protein interaction. Int J Neurosci 2021; 132:1049-1060. [PMID: 33401985 DOI: 10.1080/00207454.2020.1860966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background: It is difficult to distinguish cognitive decline due to AD from that sustained by cerebrovascular disease in view of the great overlap. It is uncertain in the molecular biological pathway behind AD and VaD.Objective: Our study aimed to explore the hub molecules and their associations with each other to identify potential biomarkers and therapeutic targets for the AD and VaD.Methods: We screened the differentially expressed genes of AD and VaD, used weighted gene co-expression network analysis and then constructed a VaD-AD-specific protein-protein interaction network with functional annotation to their related metabolic pathways. Finally, we performed a ROC curve analysis of hub proteins to get an idea about their diagnostic value.Results: In the frontal lobe and temporal cortex, hub genes were identified. With regard to VaD, there were only three hub genes which encoded the neuropeptides, SST, NMU and TAC1. The AUC of these genes were 0.804, 0.768 and 0.779, respectively. One signature was established for these three hub genes with AUC of 0.990. For the identification of AD and VaD, all hub genes were receptors. These genes included SH3GL2, PROK2, TAC3, HTR2A, MET, TF, PTH2R CNR1, CHRM4, PTPN3 and CRH. The AUC of these genes were 0.853, 0.859, 0.796, 0.775, 0.706, 0.677, 0.696, 0.668 and 0.652, respectively. The other signature was built for eleven hub genes with AUC of 0.990.Conclusion: In the frontal lobe and temporal cortex regions, hub genes are used as diagnostic markers, which may provide insight into personalized potential biomarkers and therapeutic targets for patients with VaD and AD.
Collapse
Affiliation(s)
- Xiaodou Tian
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, P.R. China
| | - Yao Qin
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, P.R. China
| | - Yuling Tian
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, P.R. China
| | - Xiaoyan Ge
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, P.R. China
| | - Jing Cui
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, P.R. China
| | - Hongjuan Han
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, P.R. China
| | - Long Liu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, P.R. China
| | - Hongmei Yu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, P.R. China.,Shanxi Provincial Key Laboratory of Major Diseases Risk Assessment, Shanxi Medical University, Taiyuan, P.R. China
| |
Collapse
|
11
|
Skiba MA, Kruse AC. Autoantibodies as Endogenous Modulators of GPCR Signaling. Trends Pharmacol Sci 2020; 42:135-150. [PMID: 33358695 DOI: 10.1016/j.tips.2020.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/24/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023]
Abstract
Endogenous self-reactive autoantibodies (AAs) recognize a range of G-protein-coupled receptors (GPCRs). They are frequently associated with cardiovascular, neurological, and autoimmune disorders, and in some cases directly impact disease progression. Many GPCR AAs modulate receptor signaling, but molecular details of their modulatory activity are not well understood. Technological advances have provided insight into GPCR biology, which now facilitates deeper understanding of GPCR AA function at the molecular level. Most GPCR AAs are allosteric modulators and exhibit a broad range of pharmacological properties, altering both receptor signaling and trafficking. Understanding GPCR AAs is not only important for defining how these unusual GPCR modulators function in disease, but also provides insight into the potential use and limitations of using therapeutic antibodies to modulate GPCR signaling.
Collapse
Affiliation(s)
- Meredith A Skiba
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Perez DM. α 1-Adrenergic Receptors in Neurotransmission, Synaptic Plasticity, and Cognition. Front Pharmacol 2020; 11:581098. [PMID: 33117176 PMCID: PMC7553051 DOI: 10.3389/fphar.2020.581098] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
α1-adrenergic receptors are G-Protein Coupled Receptors that are involved in neurotransmission and regulate the sympathetic nervous system through binding and activating the neurotransmitter, norepinephrine, and the neurohormone, epinephrine. There are three α1-adrenergic receptor subtypes (α1A, α1B, α1D) that are known to play various roles in neurotransmission and cognition. They are related to two other adrenergic receptor families that also bind norepinephrine and epinephrine, the β- and α2-, each with three subtypes (β1, β2, β3, α2A, α2B, α2C). Previous studies assessing the roles of α1-adrenergic receptors in neurotransmission and cognition have been inconsistent. This was due to the use of poorly-selective ligands and many of these studies were published before the characterization of the cloned receptor subtypes and the subsequent development of animal models. With the availability of more-selective ligands and the development of animal models, a clearer picture of their role in cognition and neurotransmission can be assessed. In this review, we highlight the significant role that the α1-adrenergic receptor plays in regulating synaptic efficacy, both short and long-term synaptic plasticity, and its regulation of different types of memory. We will also present evidence that the α1-adrenergic receptors, and particularly the α1A-adrenergic receptor subtype, are a potentially good target to treat a wide variety of neurological conditions with diminished cognition.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
13
|
Bock C, Vogt B, Mattecka S, Yapici G, Brunner P, Fimpel S, Unger JK, Sheriff A. C-Reactive Protein Causes Blood Pressure Drop in Rabbits and Induces Intracellular Calcium Signaling. Front Immunol 2020; 11:1978. [PMID: 32983135 PMCID: PMC7483553 DOI: 10.3389/fimmu.2020.01978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/22/2020] [Indexed: 01/02/2023] Open
Abstract
Systemic diseases characterized by elevated levels of C-reactive protein (CRP), such as sepsis or systemic inflammatory response syndrome, are usually associated with hardly controllable haemodynamic instability. We therefore investigated whether CRP itself influences blood pressure and heart rate. Immediately after intravenous injection of purified human CRP (3.5 mg CRP/kg body weight) into anesthetized rabbits, blood pressure dropped critically in all animals, while control animals injected with bovine serum albumin showed no response. Heart rate did not change in either group. Approaching this impact on a cellular level, we investigated the effect of CRP in cell lines expressing adrenoceptors (CHO-α1A and DU-145). CRP caused a Ca2+ signaling being dependent on the CRP dose. After complete activation of the adrenoceptors by agonists, CRP caused additional intracellular Ca2+ mobilization. We assume that CRP interacts with hitherto unknown structures on the surface of vital cells and thus interferes with the desensitization of adrenoceptors.
Collapse
Affiliation(s)
- Christopher Bock
- Division of Nephrology and Internal Intensive Care Medicine, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Birgit Vogt
- Division of Nephrology and Internal Intensive Care Medicine, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stephan Mattecka
- Division of Nephrology and Internal Intensive Care Medicine, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gülcan Yapici
- Division of Nephrology and Internal Intensive Care Medicine, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Juliane K Unger
- Department of Experimental Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ahmed Sheriff
- Division of Nephrology and Internal Intensive Care Medicine, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
14
|
Scherbaum I, Heidecke H, Bunte K, Peters U, Beikler T, Boege F. Autoantibodies against M 5-muscarinic and beta 1-adrenergic receptors in periodontitis patients. Aging (Albany NY) 2020; 12:16609-16620. [PMID: 32857064 PMCID: PMC7485715 DOI: 10.18632/aging.103864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/21/2020] [Indexed: 12/12/2022]
Abstract
Autoantibodies against muscarinic and beta1-adrenergic receptors are considered a potential cause and/or risk factor for chronic heart failure. Association of periodontitis with such autoantibodies and with impaired heart function has been observed in patients exposed to endemic Chagas' disease, which triggers by itself cardiomyopathy and receptor immunization.Here we studied the association between periodontitis, markers of cardiac injury and receptor autoimmunization in periodontitis patients (n = 147) not exposed to Chagas' disease. The autoantibodies were determined by IgG binding to native intact muscarinic and beta1-adrenergic receptors or to a cyclic peptide mimicking the disease-relevant conformational autoepitope presented by the active beta1-adrenergic receptor. Possible cardiac injury and inflammatory status were judged by serum levels of proBNP/Troponin I and CRP/IL-6, respectively. These parameters were analysed in healthy and periodontally diseased individuals as well as before and after periodontal therapy.Patients with periodontitis had significantly (p < 0.001) higher levels of autoantibodies against M5-muscarinic and beta1-adrenergic receptors, which further increased following periodontal therapy. Receptor autoantibodies were associated with increased inflammatory status but not with increased markers of cardiac injury. Thus, our data indicate that periodontitis triggers systemic inflammation, which is associated with receptor autoimmunization, and, independently thereof, with cardiac injury.
Collapse
Affiliation(s)
- Isabel Scherbaum
- Central Institute for Clinical Chemistry and Laboratory Diagnostics, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | | | - Kübra Bunte
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrike Peters
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Beikler
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fritz Boege
- Central Institute for Clinical Chemistry and Laboratory Diagnostics, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| |
Collapse
|
15
|
Abstract
PURPOSE To review the recent developments on the effect of chronic high mean arterial blood pressure (MAP) on cerebral blood flow (CBF) autoregulation and supporting the notion that CBF autoregulation impairment has connection with chronic cerebral diseases. Method: A narrative review of all the relevant papers known to the authors was conducted. Results: Our understanding of the connection between cerebral perfusion impairment and chronic high MAP and cerebral disease is rapidly evolving, from cerebral perfusion impairment being the result of cerebral diseases to being the cause of cerebral diseases. We now better understand the intertwined impact of hypertension and Alzheimer's disease (AD) on cerebrovascular sensory elements and recognize cerebrovascular elements that are more vulnerable to these diseases. Conclusion: We conclude with the suggestion that the sensory elements pathology plays important roles in intertwined mechanisms of chronic high MAP and AD that impact cerebral perfusion.
Collapse
Affiliation(s)
- Noushin Yazdani
- College of Public Health, University of South Florida , Tampa, FL, USA
| | - Mark S Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida , Tampa, FL, USA.,Biomedical Research, James A. Haley VA Medical Center , Tampa, FL, USA
| | - Saeid Taheri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida , Tampa, FL, USA.,Byrd Neuroscience Institute, University of South Florida , Tampa, FL, USA
| |
Collapse
|
16
|
Giannoni P, Claeysen S, Noe F, Marchi N. Peripheral Routes to Neurodegeneration: Passing Through the Blood-Brain Barrier. Front Aging Neurosci 2020; 12:3. [PMID: 32116645 PMCID: PMC7010934 DOI: 10.3389/fnagi.2020.00003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/08/2020] [Indexed: 12/21/2022] Open
Abstract
A bidirectional crosstalk between peripheral players of immunity and the central nervous system (CNS) exists. Hence, blood-brain barrier (BBB) breakdown is emerging as a participant mechanism of dysregulated peripheral-CNS interplay, promoting diseases. Here, we examine the implication of BBB damage in neurodegeneration, linking it to peripheral brain-directed autoantibodies and gut-brain axis mechanisms. As BBB breakdown is a factor contributing to, or even anticipating, neuronal dysfunction(s), we here identify contemporary pharmacological strategies that could be exploited to repair the BBB in disease conditions. Developing neurovascular, add on, therapeutic strategies may lead to a more efficacious pre-clinical to clinical transition with the goal of curbing the progression of neurodegeneration.
Collapse
Affiliation(s)
| | - Sylvie Claeysen
- CNRS, INSERM U1191, Institut de Génomique Fonctionnelle, University of Montpellier, Montpellier, France
| | - Francesco Noe
- HiLIFE – Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Nicola Marchi
- CNRS, INSERM U1191, Institut de Génomique Fonctionnelle, University of Montpellier, Montpellier, France
| |
Collapse
|
17
|
Li C, Yan X, Wu D, Zhang K, Liang X, Pan Y, Zhou Y, Chen F, Chen X, Yang S, Zhou Z, Wei Y, Liao Y, Qiu Z. Vaccine Targeted Alpha 1D-Adrenergic Receptor for Hypertension. Hypertension 2019; 74:1551-1562. [PMID: 31607175 DOI: 10.1161/hypertensionaha.119.13700] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The α1-AR (α1 adrenergic receptor) blockers currently on the market cannot meet clinical needs because of low-selectivity for subtypes of α1-ARs, short half-life, and uncertain role in cardiovascular end point events. The study sought to find a vaccine specifically against α1D-AR (α1D-adrenergic receptor) for treating hypertension. A short peptide ADR-004 (cgiteeagy) belonging to α1D-AR was screened, and the ADRQβ-004 vaccine was produced and injected into spontaneously hypertensive rats model (including a short-term study, 10 weeks, and a long-term observation study, 39 weeks) and NG-nitro-l-arginine methyl ester + spontaneously hypertensive rats model (15 weeks). The antihypertensive effect and target organ protection of the ADRQβ-004 vaccine were carefully evaluated. The possible immune-mediated damage was detected in normal vaccinated Sprague Dawley rats. The ADR-004 peptide has perfect immunogenicity, and the ADRQβ-004 vaccine could induce strong antibody production. In the short-term study, the ADRQβ-004 vaccine averagely decreased the systolic blood pressure of spontaneously hypertensive rats up to 15 mm Hg and that of NG-nitro-l-arginine methyl ester+spontaneously hypertensive rats up to 29 mm Hg. In the long-term observation model, the antihypertensive effect of the ADRQβ-004 vaccine was quite stable, and the average decline of systolic blood pressure was 22 mm Hg. The ADRQβ-004 vaccine effectively prevented vascular structural remodeling, cardiac hypertrophy and fibrosis, and renal injury of hypertensive animals, superior to prazosin at renal level. Moreover, the ADRQβ-004 vaccine obviously downregulated the expression of α1D-AR, but not α1A-AR. Additionally, no significant immune-mediated damage was detected in immunized animals. The present results demonstrate that the ADRQβ-004 vaccine may provide a novel and promising method for the treatment of hypertension.
Collapse
Affiliation(s)
- Chang Li
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaole Yan
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danyu Wu
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhang
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Liang
- Medical Statistics Office, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (X.L.)
| | - Yajie Pan
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanzhao Zhou
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Chen
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Chen
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shijun Yang
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihua Zhou
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yumiao Wei
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhua Liao
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihua Qiu
- From the Department of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Cardiology (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education (C.L., X.Y., D.W., K.Z., Y.P., Y.Z., F.C., X.C., S.Y., Z.Z., Y.W., Y.L., Z.Q.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Hohberger B, Kunze R, Wallukat G, Kara K, Mardin CY, Lämmer R, Schlötzer-Schrehardt U, Hosari S, Horn F, Munoz L, Herrmann M. Autoantibodies Activating the β2-Adrenergic Receptor Characterize Patients With Primary and Secondary Glaucoma. Front Immunol 2019; 10:2112. [PMID: 31632387 PMCID: PMC6779694 DOI: 10.3389/fimmu.2019.02112] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/21/2019] [Indexed: 01/27/2023] Open
Abstract
Recently, agonistic autoantibodies (agAAb) activating the β2-adrenergic receptor were detected in primary open-angle glaucoma (POAG) or ocular hypertension (OHT) patients and were linked to intraocular pressure (IOP) (1). The aim of the present study was to quantify β2-agAAb in the sera of glaucoma suspects and patients with primary and secondary glaucoma. Patients with OHT (n = 33), pre-perimetric POAG (pre-POAG; n = 11), POAG (n = 28), and 11 secondary OAG (SOAG) underwent ophthalmological examinations including examinations with Octopus G1 perimetry and morphometry. Twenty-five healthy individuals served as controls. Serum-derived IgG samples were analyzed for β2-agAAb using a functional bioassay. The beat-rate-increase of spontaneously beating cultured neonatal rat cardiomyocytes was monitored with 1.6 beats/15 s as cut-off. None of the sera of normal subjects showed β2-agAAb. In POAG or OHT patients increased beating rates of 4.1 ± 2.2 beats/15 s, and 3.7 ± 2.8 beats/15 s were detected (p > 0.05). Glaucoma patients with (POAG) and without perimetric (pre-POAG) defects did not differ (pre-POAG 4.4 ± 2.6 beats/15 s, POAG 4.1 ± 2.0 beats/15 s, p > 0.05). Patients with SOAG yielded mean beating rates of 4.7 ± 1.7 beats/15 s (p > 0.05). β2-agAAb were seen in 73% of OHT, 82% of pre-POAG, 82% of POAG, and 91% SOAG patients (p < 0.001). Clinical data did not correlate with beating rate (p > 0.05). The robust β2-agAAb seropositivity in patients with OHT, pre-POAG, POAG, and SOAG suggest a primary common role for β2-agAAb starting early in glaucoma pathophysiology and turned out to be a novel marker identifying all patients with increased IOP independent of glaucoma stage and entity.
Collapse
Affiliation(s)
- Bettina Hohberger
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Rudolf Kunze
- Science Office, Berlin-Buch, Campus Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Gerd Wallukat
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Katja Kara
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Y Mardin
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Lämmer
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | | | - Sami Hosari
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Folkert Horn
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Luis Munoz
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
19
|
Thyrian JR, Hertel J, Schulze LN, Dörr M, Prüss H, Hempel P, Bimmler M, Kunze R, Grabe HJ, Teipel S, Hoffmann W. Prevalence and Determinants of Agonistic Autoantibodies Against α1-Adrenergic Receptors in Patients Screened Positive for Dementia: Results from the Population-Based DelpHi-Study. J Alzheimers Dis 2019; 64:1091-1097. [PMID: 30010118 DOI: 10.3233/jad-171096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is a need to assess promising biomarkers for diagnosis and treatment response in real-life settings. Despite the important role of vascular risk factors, cardiovascular biomarkers have played a minor role in dementia research. Agonistic autoantibodies (agAAB) directed against G-protein-coupled receptors (GPCR) are discussed as modulators of pathology and clinical manifestation. OBJECTIVE 1) Describe prevalence of agAAB directed against GPCR, especially agABB against α1-adrenergic receptors (α1-AR-agAAB) and agABB directed against β2-adrenergic receptors (β2-AR-agAAB) and 2) identify factors associated with agAAB in people with dementia during routine care. METHODS Blood samples and data from 95 subjects who screened positive for dementia from a primary care cohort, analyzed using an enzyme-linked immunosorbent assay (ELISA) for detecting agAAB. Sociodemographic and clinical data were assessed, and medical records checked. RESULTS In 40 (42%) samples, agAAB was detected, with n = 29 (31%) representing α1-AR-agAAB and n = 21 (22%) β2-AR-agAAB. There was no association between the presence of any antibody and a formal diagnosis of dementia. However, patients with coronary heart disease were more likely (OR = 4.23) to have α1-AR-agAAB than those without coronary heart disease. There were no associations between agAAB and age, sex, education, or cognitive impairment. DISCUSSION For the first time, we show that autoantibodies have a significant prevalence in people with dementia in a routine care setting. Previous findings were restricted to highly selective samples. We replicated the association between α1-AR-agAAB in patients with coronary heart diseases but were not able to find other factors associated with the presence of agAAB.
Collapse
|
20
|
Werner C, Müller N, Müller UA. Agonistic autoantibodies against B2-adrenergic receptors correlating with macrovascular disease in longstanding diabetes type 2. Acta Diabetol 2019; 56:659-665. [PMID: 30770998 DOI: 10.1007/s00592-019-01296-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/02/2019] [Indexed: 10/27/2022]
Abstract
AIMS Agonistic autoantibodies directed against adrenergic, endothelin, and angiotensin receptors are known as pathogenic factors in disease-causing vascular impairments such as Buergers' disease, dilatative cardiomyopathy, dementia, and preeclampsia. Diabetes mellitus also causes micro- and macrovascular damages, but pathogenesis is still not fully understood. Following indications for a pathogenic role of the mentioned antibodies from our preliminary investigations, we investigated the prevalence in a bigger cohort of patients with longstanding diabetes with or without diabetic complications. METHODS We included 200 patients in four groups (grouping due to duration of diabetes and presence of complications) from our university polyclinic with longstanding diabetes mellitus type 2 and evaluated the prevalence of the agonistic autoantibodies using ELISA technique. RESULTS Antibodies directed against the alpha1-(39%), the first extracellular loop of the beta2-(34,5%), and the first extracellular loop of the beta1-adrenergic receptor (29,0%) were the most often detectable. With progression of diabetes and its complications, we found a decrease in the prevalence of the antibodies. Regression analyses revealed a positive association of antibodies against the first loop of the beta2-receptor and the presence of macrovascular complications. CONCLUSIONS This investigation found mid frequent prevalence of agonistic autoantibodies in patients with longstanding diabetes mellitus type 2. The association between an antibody against one epitope and the presence of macrovascular complications may indicates a pathogenic linkage. This finding is inconsistent with our preliminary data and needs further evaluation, maybe by follow-up.
Collapse
Affiliation(s)
- Christoph Werner
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany
| | - Nicolle Müller
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany.
| | | |
Collapse
|
21
|
Giil LM, Aarsland D, Hellton K, Lund A, Heidecke H, Schulze-Forster K, Riemekasten G, Vik-Mo AO, Kristoffersen EK, Vedeler CA, Nordrehaug JE. Antibodies to Multiple Receptors are Associated with Neuropsychiatric Symptoms and Mortality in Alzheimer’s Disease: A Longitudinal Study. J Alzheimers Dis 2018; 64:761-774. [DOI: 10.3233/jad-170882] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Lasse M. Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Norway
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, Kings College, UK
- Centre for Age-Related Diseases (SESAM), Stavanger University Hospital, Norway
| | | | - Anders Lund
- Department of Clinical Science, University of Bergen, Norway
| | | | | | - Gabriela Riemekasten
- Department of Rheumatology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Audun Osland Vik-Mo
- Department of Clinical Science, University of Bergen, Norway
- Centre for Age-Related Diseases (SESAM), Stavanger University Hospital, Norway
| | - Einar K. Kristoffersen
- Department of Clinical Science, University of Bergen, Norway
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | - Christian A. Vedeler
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Jan Erik Nordrehaug
- Department of Clinical Science, University of Bergen, Norway
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
22
|
Giil LM, Vedeler CA, Kristoffersen EK, Nordrehaug JE, Heidecke H, Dechend R, Schulze-Forster K, Muller DN, von Goetze VS, Cabral-Marques O, Riemekasten G, Vogelsang P, Nygaard S, Lund A, Aarsland D. Antibodies to Signaling Molecules and Receptors in Alzheimer's Disease are Associated with Psychomotor Slowing, Depression, and Poor Visuospatial Function. J Alzheimers Dis 2018; 59:929-939. [PMID: 28697567 DOI: 10.3233/jad-170245] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is associated with several antibodies as well as signaling molecules and receptors. These may be detrimental in the presence of a disrupted blood-brain barrier (BBB). OBJECTIVE To investigate whether the levels of antibodies toward 33 signaling molecules involved in neurotransmitter, vascular, and immune functions were associated with AD and, within the AD group; cognitive function and mood. METHODS Antibodies in sera from patients with mild AD [(n = 91) defined as a Mini-Mental State Examination ≥ 20 or a Clinical Dementia Rating Scale≤1] and healthy controls (n = 102) were measured with enzyme-linked immunosorbent assays. Levels in AD and controls were compared by Mann-Whitney test. In the AD group, associations between antibodies and psychometric test scores were analyzed by robust regression. The false discovery threshold was set to 0.05. RESULTS Antibodies to serotonin receptors [5-HT2AR (effect size (r) = 0.21, p = 0.004), 5-HT2CR (r = 0.25, p = 0.0005) and 5-HT7R (r = 0.21, p = 0.003)], vascular endothelial growth factor receptor 1 [VEGFR1 (r = 0.29, p < 0.001)] and immune-receptors (Stabilin-1 (r = 0.23, p = 0.001) and C5aR1 (r = 0.21, p = 0.004) were higher in AD. Psychomotor speed was associated with D1R-abs (β 0.49, p < 0.001), depression with ETAR-abs (β 0.31, p < 0.001), and visuospatial function with 5-HT1AR-abs (β 0.27, p = 0.004) despite similar antibody levels compared to controls. CONCLUSIONS Antibody levels to VEGFR1, serotonergic receptors, and receptors in the immune system were increased in AD. Antibodies at similar levels as in controls were associated cognitive dysfunction and depression in AD.
Collapse
Affiliation(s)
- Lasse M Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Christian A Vedeler
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Einar K Kristoffersen
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | - Jan Erik Nordrehaug
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | | | - Ralf Dechend
- Experimental and Clinical Research Center, Charité Medical Faculty and the Max-Delbruck Center for Molecular Medicine, Berlin, Germany.,HELIOS-Klinikum Berlin, Berlin, Germany
| | | | - Dominik N Muller
- Experimental and Clinical Research Center, Charité Medical Faculty and the Max-Delbruck Center for Molecular Medicine, Berlin, Germany.,Max-Delbruck Center for Molecular Medicine, Berlin, Germany
| | | | | | - Gabriela Riemekasten
- Department of Rheumatology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Petra Vogelsang
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway.,Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | - Staale Nygaard
- Research Group for Biomedical Informatics, University of Oslo, Oslo, Norway
| | - Anders Lund
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, Kings College, UK.,Centre for Age-Related Diseases (SESAM), Stavanger University Hospital, Norway
| |
Collapse
|
23
|
Abstract
Dementia in general and Alzheimer’s disease in particular is increasingly seen in association with autoimmunity being causatively or supportively involved in the pathogenesis. Besides classic autoantibodies (AABs) present in dementia patients, there is the new autoantibody class called functional autoantibodies, which is directed against G-protein coupled receptors (GPCRs; GPCR-AABs) and are seen as pathogenic players. However, less is known about dementia patients’ burden with functional autoantibodies. We present here for the first time a study analyzing the prevalence of GPCR-AABs in patients with different dementia forms such as unclassified, Lewy body, vascular and Alzheimer’s dementia. We identified the GPCR-AABs’ specific targets on the receptors and introduced a neutralization strategy for GPCR-AABs. Patients with Alzheimer’s and vascular dementia carried GPCR-AABs targeting the first loop of the alpha1- and the second loop of the beta2-adrenergic receptors (α1-AABs; β2-AABs). Nearly all vascular dementia patients also carry autoantibodies targeting the endothelin A receptor (ETA-AABs). The majority of patients with Lewy body dementia lacked any of the GPCR-AABs. In vitro, the function of the dementia-associated GPCR-AABs could be neutralized by the aptamer BC007. Due to the presence of GPCR-AABs in dementia patients mainly in those suffering from Alzheimer’s and vascular dementia, the orchestra of immune players in these dementia forms, so far preferentially represented by the classic autoantibodies, should be supplemented by functional autoantibodies. As dementia-associated functional autoantibodies could be neutralized by the aptamer BC007, the first step was taken for a new in vivo treatment option in dementia patients who were positive for GPCR-AABs.
Collapse
Affiliation(s)
| | - Harald Prüss
- Klinik für Neurologie, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Berlin, Germany
| | | | | |
Collapse
|
24
|
Jünemann A, Hohberger B, Rech J, Sheriff A, Fu Q, Schlötzer-Schrehardt U, Voll RE, Bartel S, Kalbacher H, Hoebeke J, Rejdak R, Horn F, Wallukat G, Kunze R, Herrmann M. Agonistic Autoantibodies to the β2-Adrenergic Receptor Involved in the Pathogenesis of Open-Angle Glaucoma. Front Immunol 2018; 9:145. [PMID: 29483909 PMCID: PMC5816038 DOI: 10.3389/fimmu.2018.00145] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/17/2018] [Indexed: 11/21/2022] Open
Abstract
Glaucoma is a frequent ocular disease that may lead to blindness. Primary open-angle glaucoma (POAG) and ocular hypertension (OHT) are common diseases with increased intraocular pressure (IOP), which are mainly responsible for these disorders. Their pathogenesis is widely unknown. We screened the sera of patients with POAG and OHT for the prevalence of autoantibodies (AAb) against G protein-coupled receptors (GPCRs) in comparison to controls. Employing frequency modulation of spontaneously contracting neonatal rat cardiomyocytes in vitro, agonistic GPCR AAb were to be detected in roughly 75% of the patients with POAG and OHT, however, not in controls. Using inhibitory peptides the AAb’ target was identified as β2 adrenergic receptor (β2AR). The AAb interact with the second extracellular loop of β2AR. The peptides 181–187 and 186–192 were identified as binding sites of the AAb within the extracellular loop II. The binding of the AAb to β2ARs was verified by surface-plasmon-resonance analysis. The isotype of the AAb was (immunoglobulin) IgG3. In an additional pilot principal-of-proof study, including four patients with POAG, the removal of the AAb against the β2AR and other immunoglobulins G by immunoadsorption resulted in a transient reduction of IOP. These findings might indicate a possible role of agonistic AAb directed against β2ARs in the dynamics of aqueous humor and might support a contribution of adaptive autoimmunity in the etiopathogenesis of POAG and OHT.
Collapse
Affiliation(s)
- Anselm Jünemann
- Department of Ophthalmology, University of Rostock, Rostock, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Rech
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ahmed Sheriff
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Qin Fu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Reinhard Edmund Voll
- IZKF Research Group 2, Nikolaus-Fiebiger-Center of Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Sabine Bartel
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Hubert Kalbacher
- IFIB - Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Johan Hoebeke
- C.N.R.S. UPR 9021 «Chimie et Immunologie Thérapeutiques», Strasbourg, France
| | - Robert Rejdak
- Department of General Ophthalmology, Medical University of Lublin, Lublin, Poland
| | - Folkert Horn
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gerd Wallukat
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Rudolf Kunze
- Science Office, Berlin-Buch, Campus Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Martin Herrmann
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
25
|
Hu JY, Liu BB, Du YP, Zhang Y, Zhang YW, Zhang YY, Xu M, He B. Increased circulating β 2-adrenergic receptor autoantibodies are associated with smoking-related emphysema. Sci Rep 2017; 7:43962. [PMID: 28262783 PMCID: PMC5338268 DOI: 10.1038/srep43962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/31/2017] [Indexed: 01/21/2023] Open
Abstract
Smoking is a dominant risk factor for chronic obstructive pulmonary disease (COPD) and emphysema, but not every smoker develops emphysema. Immune responses in smokers vary. Some autoantibodies have been shown to contribute to the development of emphysema in smokers. β2-adrenergic receptors (β2-ARs) are important targets in COPD therapy. β2-adrenergic receptor autoantibodies (β2-AAbs), which may directly affect β2-ARs, were shown to be increased in rats with passive-smoking-induced emphysema in our current preliminary studies. Using cigarette-smoke exposure (CS-exposure) and active-immune (via injections of β2-AR second extracellular loop peptides) rat models, we found that CS-exposed rats showed higher serum β2-AAb levels than control rats before alveolar airspaces became enlarged. Active-immune rats showed increased serum β2-AAb levels, and exhibited alveolar airspace destruction. CS-exposed-active-immune treated rats showed more extensive alveolar airspace destruction than rats undergoing CS-exposure alone. In our current clinical studies, we showed that plasma β2-AAb levels were positively correlated with the RV/TLC (residual volume/total lung capacity) ratio (r = 0.455, p < 0.001) and RV%pred (residual volume/residual volume predicted percentage, r = 0.454, p < 0.001) in 50 smokers; smokers with higher plasma β2-AAb levels exhibited worse alveolar airspace destruction. We suggest that increased circulating β2-AAbs are associated with smoking-related emphysema.
Collapse
Affiliation(s)
- Jia-Yi Hu
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China
| | - Bei-Bei Liu
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China
| | - Yi-Peng Du
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China
| | - Yuan Zhang
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China
| | - Yi-Wei Zhang
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China
| | - You-Yi Zhang
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Beijing Key Laboratory of cardiovascular Receptors Research, Beijing, China
| | - Ming Xu
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Beijing Key Laboratory of cardiovascular Receptors Research, Beijing, China
| | - Bei He
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
26
|
Shpakov AO, Zharova OA, Derkach KV. Antibodies to extracellular regions of G protein-coupled receptors and receptor tyrosine kinases as one of the causes of autoimmune diseases. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s1234567817020021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
27
|
Cardiomyopathy - An approach to the autoimmune background. Autoimmun Rev 2017; 16:269-286. [PMID: 28163240 DOI: 10.1016/j.autrev.2017.01.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/20/2016] [Indexed: 12/15/2022]
Abstract
Autoimmunity is increasingly accepted as the origin or amplifier of various diseases. In contrast to classic autoantibodies (AABs), which induce immune responses resulting in the destruction of the affected tissue, an additional class of AABs is directed against G-protein-coupled receptors (GPCRs; GPCR-AABs). GPCR-AABs functionally affect their related GPCRs for activation of receptor mediated signal cascades. Diseases which are characterized by the presence of GPCR-AABs with evidence for disease-specific pathogenic activity could be named "functional autoantibody disease". We briefly summarize here the historical view on autoimmunity in cardiomyopathy, followed by an approach to the mechanistic autoimmunity background. Furthermore, autoantibodies with outstanding importance for cardiomyopathies as a functional autoantibody disease, such as GPCR-AABs, and mainly those directed against the beta1-adrenergic and muscarinic 2 receptor autoantibodies, are introduced. Anti-cardiac myosin and anti-cardiac troponin autoantibodies, as further potential players in autoimmune cardiomyopathy, are additionally taken into account. The basic view on the autoantibodies, their related receptor interactions and pathogenic consequences are presented. Focused specifically on GPCR-AABs, "pros and cons" of assays such as indirect assays (functional changes of cell preparations are monitored after GPCR-AAB receptor binding) and direct assays based on the ELISA technologies (GPCR epitope mimics for GPCR-AAB binding) are critically discussed. Last but not least, treatment strategies for "functional autoantibody disease", such as for GPCR-AAB removal (therapeutic plasma exchange, immunoadsorption) and in vivo GPCR-AAB attack such as intravenous IgG treatment (IVIG), B-cell depletion and GPCR-AAB binding and neutralization, are critically reflected with respect to their patient benefits.
Collapse
|
28
|
Counts SE, Ikonomovic MD, Mercado N, Vega IE, Mufson EJ. Biomarkers for the Early Detection and Progression of Alzheimer's Disease. Neurotherapeutics 2017; 14:35-53. [PMID: 27738903 PMCID: PMC5233625 DOI: 10.1007/s13311-016-0481-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The recent failures of potential disease-modifying drugs for Alzheimer's disease (AD) may reflect the fact that the enrolled participants in clinical trials are already too advanced to derive a clinical benefit. Thus, well-validated biomarkers for the early detection and accurate diagnosis of the preclinical stages of AD will be crucial for therapeutic advancement. The combinatorial use of biomarkers derived from biological fluids, such as cerebrospinal fluid (CSF), with advanced molecular imaging and neuropsychological testing may eventually achieve the diagnostic sensitivity and specificity necessary to identify people in the earliest stages of the disease when drug modification is most likely possible. In this regard, positive amyloid or tau tracer retention on positron emission tomography imaging, low CSF concentrations of the amyloid-β 1-42 peptide, high CSF concentrations in total tau and phospho-tau, mesial temporal lobe atrophy on magnetic resonance imaging, and temporoparietal/precuneus hypometabolism or hypoperfusion on 18F-fluorodeoxyglucose positron emission tomography have all emerged as biomarkers for the progression to AD. However, the ultimate AD biomarker panel will likely involve the inclusion of novel CSF and blood biomarkers more precisely associated with confirmed pathophysiologic mechanisms to improve its reliability for detecting preclinical AD. This review highlights advancements in biological fluid and imaging biomarkers that are moving the field towards achieving the goal of a preclinical detection of AD.
Collapse
Affiliation(s)
- Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Family Medicine, Michigan State University, Grand Rapids, MI, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Natosha Mercado
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Irving E Vega
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA.
| |
Collapse
|
29
|
Hempel P, Heinig B, Jerosch C, Decius I, Karczewski P, Kassner U, Kunze R, Steinhagen-Thiessen E, Bimmler M. Immunoadsorption of Agonistic Autoantibodies Against α1-Adrenergic Receptors in Patients With Mild to Moderate Dementia. Ther Apher Dial 2016; 20:523-529. [PMID: 27096216 DOI: 10.1111/1744-9987.12415] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 12/21/2022]
Abstract
Dementia has been shown to be associated with agonistic autoantibodies. The deleterious action of autoantibodies on the α1-adrenergic receptor for brain vasculature has been demonstrated in animal studies. In the current study, 169 patients with dementia were screened for the presence of agonistic autoantibodies. 47% of patients suffering from mild to moderate Alzheimer's disease and/or vascular dementia carried these autoantibodies. Eight patients positive for autoantibodies underwent immunoadsorption. Patients treated on four consecutive days were subsequently negative for autoantibodies and displayed stabilization of cognitive and mental condition during 12-18 months' follow-up. In patients treated for 2-3 days, autoantibodies were reduced by only 78%. They suffered a rebound of autoantibodies during follow-up, benefited from immunoadsorption too, but their mental parameters worsened. We provide first data on the clinical relevance of agonistic autoantibodies in dementia and show that immunoadsorption is safe and efficient in removing autoantibodies with overall benefits for patients.
Collapse
Affiliation(s)
| | - Bente Heinig
- Charité - University Medicine Berlin, Berlin, Germany
| | | | - Imke Decius
- Charité - University Medicine Berlin, Berlin, Germany
| | | | | | | | | | - Marion Bimmler
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
30
|
Wallukat G, Müller J, Haberland A, Berg S, Schulz A, Freyse EJ, Vetter R, Salzsieder E, Kreutz R, Schimke I. Aptamer BC007 for neutralization of pathogenic autoantibodies directed against G-protein coupled receptors: A vision of future treatment of patients with cardiomyopathies and positivity for those autoantibodies. Atherosclerosis 2015; 244:44-7. [PMID: 26584137 DOI: 10.1016/j.atherosclerosis.2015.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/20/2015] [Accepted: 11/01/2015] [Indexed: 10/22/2022]
Abstract
Cardiomyopathies such as idiopathic dilated cardiomyopathy (DCM), Chagas' cardiomyopathy and Peripartum cardiomyopathy present with autoantibodies against G-protein coupled receptors (GPCR-AABs) that agonistically activate their receptors. For the treatment of "agonistic autoantibody diseases" and in particular DCM, the removal of the GPCR-AABs by immunoadsorption (IA) has been studied with convincing patient benefit. To overcome cost and logistics problems of IA, the application of the aptamer BC007 for in vivo neutralization of GPCR-AABs could help. We demonstrate here, that the aptamer neutralized, in vitro, the presently known cardiovascular-pathogenic GPCR-AABs. In spontaneously hypertensive rats, the aptamer demonstrated its GPCR-AAB neutralizing potency in vivo. In the serum of DCM patients, the same GPCR-AAB reduction was achieved when patients were either immunoadsorbed or patient's serum was ex vivo treated with the aptamer. In our view, aptamer BC007 treatment in GPCR-AAB-positive patients would have a comparable benefit as that seen after IA. Not knowing all that interfering with our idea of aptamer-dependent neutralization of GPCR-AABs, the first preliminary steps have been taken for bringing the idea closer to patients.
Collapse
Affiliation(s)
| | | | | | - Sabine Berg
- Institut für Diabetes 'Gerhardt Katsch', Karlsburg, Germany
| | - Angela Schulz
- Institut für Klinische Pharmakologie und Toxikologie, Charité - Universitätsmedizin Berlin, Germany
| | | | - Roland Vetter
- Institut für Klinische Pharmakologie und Toxikologie, Charité - Universitätsmedizin Berlin, Germany
| | | | - Reinhold Kreutz
- Institut für Klinische Pharmakologie und Toxikologie, Charité - Universitätsmedizin Berlin, Germany
| | | |
Collapse
|
31
|
Giil LM, Kristoffersen EK, Vedeler CA, Aarsland D, Nordrehaug JE, Winblad B, Cedazo-Minguez A, Lund A, Reksten TR. Autoantibodies Toward the Angiotensin 2 Type 1 Receptor: A Novel Autoantibody in Alzheimer’s Disease. J Alzheimers Dis 2015; 47:523-9. [DOI: 10.3233/jad-150053] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Lasse M. Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Einar K. Kristoffersen
- Institute of Clinical Science, University of Bergen, Bergen, Norway
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | | | - Dag Aarsland
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
- Centre for Age-Related Diseases (SESAM), Stavanger University Hospital, Stavanger, Norway
| | - Jan Erik Nordrehaug
- Institute of Clinical Science, University of Bergen, Bergen, Norway
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Angel Cedazo-Minguez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Anders Lund
- Institute of Clinical Science, University of Bergen, Bergen, Norway
| | - Tove Ragna Reksten
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
- Centre for Age-Related Diseases (SESAM), Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
32
|
Pohlmann A, Karczewski P, Ku MC, Dieringer B, Waiczies H, Wisbrun N, Kox S, Palatnik I, Reimann HM, Eichhorn C, Waiczies S, Hempel P, Lemke B, Niendorf T, Bimmler M. Cerebral blood volume estimation by ferumoxytol-enhanced steady-state MRI at 9.4 T reveals microvascular impact of α1 -adrenergic receptor antibodies. NMR IN BIOMEDICINE 2014; 27:1085-1093. [PMID: 25060359 DOI: 10.1002/nbm.3160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/11/2014] [Accepted: 06/05/2014] [Indexed: 06/03/2023]
Abstract
Cerebrovascular abnormality is frequently accompanied by cognitive dysfunctions, such as dementia. Antibodies against the α1 -adrenoceptor (α1 -AR) can be found in patients with Alzheimer's disease with cerebrovascular disease, and have been shown to affect the larger vessels of the brain in rodents. However, the impact of α1 -AR antibodies on the cerebral vasculature remains unclear. In the present study, we established a neuroimaging method to measure the relative cerebral blood volume (rCBV) in small rodents with the ultimate goal to detect changes in blood vessel density and/or vessel size induced by α1 -AR antibodies. For this purpose, mapping of R2 * and R2 was performed using MRI at 9.4 T, before and after the injection of intravascular iron oxide particles (ferumoxytol). The change in the transverse relaxation rates (ΔR2 *, ΔR2 ) showed a significant rCBV decrease in the cerebrum, cortex and hippocampus of rats (except hippocampal ΔR2 ), which was more pronounced for ΔR2 * than for ΔR2 . Immunohistological analyses confirmed that the α1 -AR antibody induced blood vessel deficiencies. Our findings support the hypothesis that α1 -AR antibodies lead to cerebral vessel damage throughout the brain, which can be monitored by MRI-derived rCBV, a non-invasive neuroimaging method. This demonstrates the value of rCBV estimation by ferumoxytol-enhanced MRI at 9.4 T, and further underlines the significance of this antibody in brain diseases involving vasculature impairments, such as dementia.
Collapse
Affiliation(s)
- Andreas Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
The patent situation concerning the treatment of diseases associated with autoantibodies directed against G-protein-coupled receptors. Pharm Pat Anal 2014; 2:231-48. [PMID: 24237028 DOI: 10.4155/ppa.12.88] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Agonist-like autoantibodies against receptors of the G-protein-coupled signal cascade have been identified as the pathogenic principle for a variety of diseases, especially those of the heart and vascular system. Consequently, the elimination or neutralization of such autoantibodies is an advised goal for causal therapeutic intervention. This article provides a short, noncomplete overview about remarkable developmental strategies and technical solutions for the therapy of diseases, associated with G-protein-coupled receptor autoantibodies. According to the immunoglobulin nature of the therapeutic target, several strategies are possible, such as the use of the autoantibody epitope sequences as competitors or binding molecules for specific autoantibody-elimination by apheresis. Complete immunoglobulin elimination, as is currently being tested in autoantibody-positive cardiomyopathy patients, would be a nonspecific solution as would the use of immunosuppressant agents. The use of autoantibody-binding molecules on an aptamer basis for neutralization or elimination is a newly developed specific therapeutic option.
Collapse
|
34
|
Agonistic autoantibodies directed against G-protein-coupled receptors and their relationship to cardiovascular diseases. Semin Immunopathol 2014; 36:351-63. [PMID: 24777744 DOI: 10.1007/s00281-014-0425-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/01/2014] [Indexed: 02/06/2023]
Abstract
Agonistic autoantibodies (AABs) against G-protein-coupled receptor (GPCR) are present mainly in diseases of the cardiovascular system or in diseases associated with cardiovascular disturbances. The increasing knowledge about the role of autoantibodies against G-protein-coupled receptor (GPCR-AABs) as pathogenic drivers, the resulting development of strategies aimed at their removal or neutralization, and the evidenced patient benefit associated with such therapies have created the need for a summary of GPCR-AAB-associated diseases. Here, we summarize the present knowledge about GPCR-AABs in cardiovascular diseases. The identity of the GPCR-AABs and their prevalence in each of several specific cardiovascular diseases are documented. The structure of GPCR is also briefly discussed. Using this information, differences between classic agonists and GPCR-AABs in their GPCR binding and activation are presented and the resulting pathogenic consequences are discussed. Furthermore, treatment strategies that are currently under study, most of which are aimed at the removal and in vivo neutralization of GPCR-AABs, are indicated and their patient benefits discussed. In this context, immunoadsorption using peptides/proteins or aptamers as binders are introduced. The use of peptides or aptamers for in vivo neutralization of GPCR-AABs is also described. Particular attention is given to the GPCR-AABs directed against the adrenergic beta1-, beta2-, and α1-receptor as well as the muscarinic receptor M2, angiotensin II-angiotensin receptor type I, endothelin1 receptor type A, angiotensin (1-7) Mas-receptor, and 5-hydroxytryptamine receptor 4. Among the diseases associated with GPCR-AABs, special focus is given to idiopathic dilated cardiomyopathy, Chagas' cardiomyopathy, malignant and pulmonary hypertension, and kidney diseases. Relationships of GPCR-AABs are indicated to glaucoma, peripartum cardiomyopathy, myocarditis, pericarditis, preeclampsia, Alzheimer's disease, Sjörgren's syndrome, and metabolic syndrome after cancer chemotherapy.
Collapse
|
35
|
Majláth Z, Toldi J, Vécsei L. The potential role of kynurenines in Alzheimer's disease: pathomechanism and therapeutic possibilities by influencing the glutamate receptors. J Neural Transm (Vienna) 2013; 121:881-9. [PMID: 24346138 DOI: 10.1007/s00702-013-1135-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/03/2013] [Indexed: 12/14/2022]
Abstract
The pathomechanism of neurodegenerative disorders still poses a challenge to neuroscientists, and continuous research is under way with the aim of attaining an understanding of the exact background of these devastating diseases. The pathomechanism of Alzheimer's disease (AD) is associated with characteristic neuropathological features such as extracellular amyloid-β and intracellular tau deposition. Glutamate excitotoxicity and neuroinflammation are also factors that are known to contribute to the neurodegenerative process, but a cerebrovascular dysfunction has recently also been implicated in AD. Current therapeutic tools offer moderate symptomatic treatment, but fail to reduce disease progression. The kynurenine pathway (KP) has been implicated in the development of neurodegenerative processes, and alterations in the KP have been demonstrated in both acute and chronic neurological disorders. Kynurenines have been suggested to be involved in the regulation of neurotransmission and in immunological processes. Targeting the KP, therefore, offers a valuable strategic option for the attenuation of glutamatergic excitotoxicity, and for neuroprotection.
Collapse
Affiliation(s)
- Zsófia Majláth
- Department of Neurology, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary
| | | | | |
Collapse
|
36
|
Immunity and Alzheimer's disease: immunological perspectives on the development of novel therapies. Drug Discov Today 2013; 18:1212-20. [DOI: 10.1016/j.drudis.2013.07.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 07/19/2013] [Accepted: 07/30/2013] [Indexed: 02/07/2023]
|
37
|
Lương KVQ, Nguyen LTH. The role of Beta-adrenergic receptor blockers in Alzheimer's disease: potential genetic and cellular signaling mechanisms. Am J Alzheimers Dis Other Demen 2013; 28:427-39. [PMID: 23689075 PMCID: PMC10852699 DOI: 10.1177/1533317513488924] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
According to genetic studies, Alzheimer's disease (AD) is linked to beta-adrenergic receptor blockade through numerous factors, including human leukocyte antigen genes, the renin-angiotensin system, poly(adenosine diphosphate-ribose) polymerase 1, nerve growth factor, vascular endothelial growth factor, and the reduced form of nicotinamide adenine dinucleotide phosphate. Beta-adrenergic receptor blockade is also implicated in AD due to its effects on matrix metalloproteinases, mitogen-activated protein kinase pathways, prostaglandins, cyclooxygenase-2, and nitric oxide synthase. Beta-adrenergic receptor blockade may also have a significant role in AD, although the role is controversial. Behavioral symptoms, sex, or genetic factors, including Beta 2-adrenergic receptor variants, apolipoprotein E, and cytochrome P450 CYP2D6, may contribute to beta-adrenergic receptor blockade modulation in AD. Thus, the characterization of beta-adrenergic receptor blockade in patients with AD is needed.
Collapse
Affiliation(s)
- Khanh vinh quoc Lương
- Vietnamese American Medical Research Foundation, Westminster, California, CA 92683, USA.
| | | |
Collapse
|
38
|
Karczewski P, Pohlmann A, Wagenhaus B, Wisbrun N, Hempel P, Lemke B, Kunze R, Niendorf T, Bimmler M. Antibodies to the α1-adrenergic receptor cause vascular impairments in rat brain as demonstrated by magnetic resonance angiography. PLoS One 2012; 7:e41602. [PMID: 22860001 PMCID: PMC3408502 DOI: 10.1371/journal.pone.0041602] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/24/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Circulating agonistic autoantibodies acting at G protein-coupled receptors have been associated with numerous sever pathologies in humans. Antibodies directed predominantly against the α(1)-adrenergig receptor were detected in patients suffering from widespread diseases such as hypertension and type 2 diabetes. Their deleterious action has been demonstrated for peripheral organs. We postulate that antibodies to the α(1)-adrenergig receptor are relevant pathomolecules in diseases of the central nervous system associated with vascular impairments. METHODOLOGY/PRINCIPAL FINDINGS Using a rat model we studied the long-term action of antibodies against the α(1)-adrenergig receptor either induced by immunization with a receptor peptide or applied by intravenous injection. The vasculature in the rat brains was investigated by time-of-flight magnetic resonance angiography using a 9.4 Tesla small animal MR imaging system. Visual examination of maximum-intensity-projections (MIPs) of brain angiographs revealed the development of vascular defects in antibody- exposed animals between three and eight months of treatment. Relative vascular areas were derived from representative MIP image sections by grayscale analysis and used to form an index of vascular circulation. Animals exposed to the action of α(1)-adrenergig receptor antibodies showed significantly reduced vascular areas (p<0.05). Calculated index values indicated attenuated blood flow in both antibody-treated cohorts compared to their respective controls reaching with (relative units ± standard error, n = 10) 0.839 ± 0.026 versus 0.919 ± 0.026 statistical significance (p<0.05) for peptide-immunized rats. CONCLUSION/SIGNIFICANCE We present evidence that antibodies to the α(1)-adrenergig receptor cause cerebrovascular impairments in the rat. Our findings suggest the pathological significance of these antibodies in pathologies of the human central nervous system linked to impairments of brain vasculature such as stroke and dementia.
Collapse
|
39
|
Autoimmune responses to brain following stroke. Transl Stroke Res 2012; 3:310-7. [PMID: 24323806 DOI: 10.1007/s12975-012-0154-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 03/09/2012] [Accepted: 03/13/2012] [Indexed: 12/25/2022]
Abstract
This review provides a synthesis of the work done by our laboratory that demonstrates the presence of cellular immune responses directed towards brain antigens in animals following experimental stroke as well as in patients following ischemic stroke. These responses include both antigen-specific TH1(+) responses, which are associated with worse stroke outcome, and antigen-specific TREG responses, which are associated with better stroke outcome. The likelihood of developing a detrimental TH1(+) response to brain antigens is increased by administration of a systemic inflammatory stimulus in experimental stroke and by systemic infection in patients with stroke. We propose that the microenvironment within the lymph nodes and brain is altered by systemic inflammation and allows for bystander activation of lymphocytes and the development of autoimmune responses to brain antigens following cerebral ischemic injury.
Collapse
|