1
|
Michalak KP, Michalak AZ, Brenk-Krakowska A. Acute COVID-19 and LongCOVID syndrome - molecular implications for therapeutic strategies - review. Front Immunol 2025; 16:1582783. [PMID: 40313948 PMCID: PMC12043656 DOI: 10.3389/fimmu.2025.1582783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been recognized not only for its acute effects but also for its ability to cause LongCOVID Syndrome (LCS), a condition characterized by persistent symptoms affecting multiple organ systems. This review examines the molecular and immunological mechanisms underlying LCS, with a particular focus on autophagy inhibition, chronic inflammation, oxidative, nitrosative and calcium stress, viral persistence and autoimmunology. Potential pathophysiological mechanisms involved in LCS include (1) autoimmune activation, (2) latent viral persistence, where SARS-CoV-2 continues to influence host metabolism, (3) reactivation of latent pathogens such as Epstein-Barr virus (EBV) or cytomegalovirus (CMV), exacerbating immune and metabolic dysregulation, and (4) possible persistent metabolic and inflammatory dysregulation, where the body fails to restore post-infection homeostasis. The manipulation of cellular pathways by SARS-CoV-2 proteins is a critical aspect of the virus' ability to evade immune clearance and establish long-term dysfunction. Viral proteins such as NSP13, ORF3a and ORF8 have been shown to disrupt autophagy, thereby impairing viral clearance and promoting immune evasion. In addition, mitochondrial dysfunction, dysregulated calcium signaling, oxidative stress, chronic HIF-1α activation and Nrf2 inhibition create a self-sustaining inflammatory feedback loop that contributes to tissue damage and persistent symptoms. Therefore understanding the molecular basis of LCS is critical for the development of effective therapeutic strategies. Targeting autophagy and Nrf2 activation, glycolysis inhibition, and restoration calcium homeostasis may provide novel strategies to mitigate the long-term consequences of SARS-CoV-2 infection. Future research should focus on personalized therapeutic interventions based on the dominant molecular perturbations in individual patients.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | | - Alicja Brenk-Krakowska
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| |
Collapse
|
2
|
Shi M, Chu F, Zhu F, Zhu J. Peripheral blood amyloid-β involved in the pathogenesis of Alzheimer's disease via impacting on peripheral innate immune cells. J Neuroinflammation 2024; 21:5. [PMID: 38178136 PMCID: PMC10765910 DOI: 10.1186/s12974-023-03003-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024] Open
Abstract
A key pathological factor of Alzheimer's disease (AD), the most prevalent form of age-related dementia in the world, is excessive β-amyloid protein (Aβ) in extracellular aggregation in the brain. And in the peripheral blood, a large amount of Aβ is derived from platelets. So far, the causality between the levels of peripheral blood Aβ and its aggregation in the brain, particularly the role of the peripheral blood Aβ in the pathology of AD, is still unclear. And the relation between the peripheral blood Aβ and tau tangles of brain, another crucial pathologic factor contributing to the pathogenesis of AD, is also ambiguous. More recently, the anti-Aβ monoclonal antibodies are approved for treatment of AD patients through declining the peripheral blood Aβ mechanism of action to enhance plasma and central nervous system (CNS) Aβ clearance, leading to a decrease Aβ burden in brain and improving cognitive function, which clearly indicates that the levels of the peripheral blood Aβ impacted on the Aβ burden in brain and involved in the pathogenesis of AD. In addition, the role of peripheral innate immune cells in AD remains mostly unknown and the results obtained were controversial. In the present review, we summarize recent studies on the roles of peripheral blood Aβ and the peripheral innate immune cells in the pathogenesis of AD. Finally, based on the published data and our own work, we believe that peripheral blood Aβ plays an important role in the development and progression of AD by impacting on the peripheral innate immune cells.
Collapse
Affiliation(s)
- Mingchao Shi
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Fengna Chu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Feiqi Zhu
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden.
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China.
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden.
| |
Collapse
|
3
|
Barin A, Das RK, Bastani NE, Iversen PO, Duttaroy AK. Extracts of tamarillo, horned melon, and raspberry, but not extract of pear, inhibit human blood platelet aggregation: Investigating the underlying factors for their differential mechanisms. J Funct Foods 2023; 110:105847. [DOI: 10.1016/j.jff.2023.105847] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
4
|
Wolska N, Celikag M, Failla AV, Tarafdar A, Renné T, Torti M, Canobbio I, Pula G. Human platelets release amyloid peptides β 1-40 and β 1-42 in response to haemostatic, immune, and hypoxic stimuli. Res Pract Thromb Haemost 2023; 7:100154. [PMID: 37222974 PMCID: PMC7614566 DOI: 10.1016/j.rpth.2023.100154] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023] Open
Abstract
Background Platelets contain high levels of amyloid β (Aβ) peptides and have been suggested to participate in the deposition of amyloid plaques in Alzheimer's Disease (AD). Objectives This study aimed to determine whether human platelets release pathogenic Aβ peptides Aβ1-42 and Aβ1-40 and to characterise the mechanisms regulating this phenomenon. Methods and Results Enzyme-linked immunosorbent assays (ELISAs) revealed that the haemostatic stimulus thrombin and the pro-inflammatory molecule lipopolysaccharide (LPS) induce platelet release of both Aβ1-42 and Aβ1-40. Notably, LPS preferentially induced the release of Aβ1-42, which was potentiated by the reduction of oxygen from atmospheric levels to physiological hypoxia. The selective β secretase (BACE) inhibitor LY2886721 showed no effect on the release of either Aβ1-40 or Aβ1-42 in our ELISA experiments. This suggested a store-and-release mechanism that was confirmed in immunostaining experiments showing co-localisation of cleaved Aβ peptides with platelet alpha granules. Conclusions Taken together, our data suggest that human platelets release pathogenic Aβ peptides as a result of a store-and-release mechanism rather than a de novo proteolytic event. Although further studies are required to fully characterise this phenomenon, we suggest the possibility of a role for platelets in the deposition of Aβ peptides and the formation of amyloid plaques. Interestingly, the combination of hypoxia and inflammation that we simulated in vitro with reduced oxygen tension and LPS may increase the release of fibrillogenic Aβ1-42 and, consequently, exacerbate amyloid plaque deposition in the brain of AD patients.
Collapse
Affiliation(s)
- Nina Wolska
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Eppendorf (UKE), Hamburg (Germany)
| | - Meral Celikag
- UK Dementia Research Institute at University College London, London (UK)
| | | | - Anuradha Tarafdar
- Cancer Research Horizons, Babraham Research Campus, Cambridge (UK)
- Institute of Biomedical & Clinical Science, University of Exeter Medical School (UEMS), Exeter (UK)
| | - Thomas Renné
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Eppendorf (UKE), Hamburg (Germany)
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (Ireland)
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz (Germany)
| | - Mauro Torti
- Department of Biology and Biotechnology, University of Pavia (Italy)
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia (Italy)
| | - Giordano Pula
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Eppendorf (UKE), Hamburg (Germany)
- Institute of Biomedical & Clinical Science, University of Exeter Medical School (UEMS), Exeter (UK)
- Biomedical Institute for Multimorbidity, Hull and York Medical School (HYMS), Hull (UK)
| |
Collapse
|
5
|
Ormazabal P, Rodriguez L, Paredes A, Morales G, Fuentes E, Palomo I. Antiplatelet activity of Lampaya medicinalis Phil. in human platelets. NFS JOURNAL 2022. [DOI: 10.1016/j.nfs.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
6
|
Page MJ, Pretorius E. Platelet Behavior Contributes to Neuropathologies: A Focus on Alzheimer's and Parkinson's Disease. Semin Thromb Hemost 2021; 48:382-404. [PMID: 34624913 DOI: 10.1055/s-0041-1733960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The functions of platelets are broad. Platelets function in hemostasis and thrombosis, inflammation and immune responses, vascular regulation, and host defense against invading pathogens, among others. These actions are achieved through the release of a wide set of coagulative, vascular, inflammatory, and other factors as well as diverse cell surface receptors involved in the same activities. As active participants in these physiological processes, platelets become involved in signaling pathways and pathological reactions that contribute to diseases that are defined by inflammation (including by pathogen-derived stimuli), vascular dysfunction, and coagulation. These diseases include Alzheimer's and Parkinson's disease, the two most common neurodegenerative diseases. Despite their unique pathological and clinical features, significant shared pathological processes exist between these two conditions, particularly relating to a central inflammatory mechanism involving both neuroinflammation and inflammation in the systemic environment, but also neurovascular dysfunction and coagulopathy, processes which also share initiation factors and receptors. This triad of dysfunction-(neuro)inflammation, neurovascular dysfunction, and hypercoagulation-illustrates the important roles platelets play in neuropathology. Although some mechanisms are understudied in Alzheimer's and Parkinson's disease, a strong case can be made for the relevance of platelets in neurodegeneration-related processes.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| |
Collapse
|
7
|
Sanzana S, Rodríguez L, Barraza Barrionuevo H, Albornoz Poblete C, Maróstica Junior MR, Fuentes E, Palomo I. Antiplatelet Activity of Cucurbita maxima. J Med Food 2021; 24:1197-1205. [PMID: 34463138 DOI: 10.1089/jmf.2021.0006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Natural extracts constitute an important source in the prevention of noncommunicable diseases, such as cardiovascular diseases. The pumpkin, Cucurbita maxima, is widely consumed in Chile. Pumpkin seeds, despite having crude protein, lipids, and carbohydrates, are regarded as agro-industrial waste. In this work, we correlated the antiplatelet activity of aqueous, ethanolic, and methanolic extracts from pumpkin seeds with their bioactive compounds. In vitro platelet aggregation and activation studies were performed by turbidimetry and flow cytometry, respectively. Results reveal that the extracts inhibited, in a dose-dependent manner, platelet aggregation induced by adenosine diphosphate, thrombin receptor activator peptide 6 (TRAP-6), and collagen. Pumpkin seed extracts inhibited P-selectin secretion and glycoprotein IIb/IIIa activation on TRAP-6-activated platelets. They were found to be rich in fatty acids and a powerful source of plant-based protein, which could be related to the high antiplatelet potential identified in extracts. This research demonstrated that pumpkin seed extracts could be a candidate in the prevention of thrombotic events.
Collapse
Affiliation(s)
- Sigrid Sanzana
- Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Lyanne Rodríguez
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Hayleen Barraza Barrionuevo
- Department of Food and Nutrition Sciences, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - César Albornoz Poblete
- Department of Food and Nutrition Sciences, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Mário Roberto Maróstica Junior
- Ibero-American Network for the Integrated Use of Underutilized Indigenous Foods (ALSUB-CYTED).,School of Food Engineering, University of Campinas, Campinas, São Paulo, Brazil.,University of Campinas (UNICAMP), Faculty of School Engineering (FEA), Campinas, São Paulo, Brazil
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| |
Collapse
|
8
|
Antiplatelet Activity of Isorhamnetin via Mitochondrial Regulation. Antioxidants (Basel) 2021; 10:antiox10050666. [PMID: 33922903 PMCID: PMC8146847 DOI: 10.3390/antiox10050666] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 01/02/2023] Open
Abstract
With the diet, we ingest nutrients capable of modulating platelet function, which plays a crucial role in developing cardiovascular events, one of the leading causes of mortality worldwide. Studies that demonstrate the antiplatelet and antithrombotic potential of bioactive compounds are vital to maintaining good cardiovascular health. In this work, we evaluate the flavonol isorhamnetin’s antiplatelet effect on human platelets, using collagen, thrombin receptor activator peptide 6 (TRAP-6), and phorbol myristate acetate (PMA) as agonists. Isorhamnetin induced a significant inhibition on collagen- and TRAP-6-induced platelet aggregation, with half-maximum inhibitory concentration (IC50) values of 8.1 ± 2.6 and 16.1 ± 11.1 µM, respectively; while it did not show cytotoxic effect. Isorhamnetin reduced adenosine triphosphate levels (ATP) in platelets stimulated by collagen and TRAP-6. We also evidenced that isorhamnetin’s antiplatelet activity was related to the inhibition of mitochondrial function without effect on reactive oxygen species (ROS) levels. Additionally, we investigated isorhamnetin’s effect on thrombus formation in vitro under flow conditions on the damaged vessel wall. In this context, we demonstrate that isorhamnetin at 20 µM induced a significant inhibition on platelet deposition, confirming its antithrombotic effect. Our findings corroborate the antiplatelet and antithrombotic potential of isorhamnetin present in many foods of daily consumption.
Collapse
|
9
|
Wu T, Chen L, Zhou L, Xu J, Guo K. Platelets transport β-amyloid from the peripheral blood into the brain by destroying the blood-brain barrier to accelerate the process of Alzheimer's disease in mouse models. Aging (Albany NY) 2021; 13:7644-7659. [PMID: 33668038 PMCID: PMC7993748 DOI: 10.18632/aging.202662] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/25/2021] [Indexed: 12/17/2022]
Abstract
Extracellular aggregation of the β-amyloid (Aβ) peptide into toxic multimers in the brain is a prominent event occurring in the pathogenesis of Alzheimer's disease (AD), and a large amount of Aβ in the blood is derived from platelets. Thus, we speculated that platelets may play an important role in the process of AD. We first investigated the changes in platelet Aβ secretion with age. Then, we injected platelets from aged amyloid precursor protein APP/PS1 mice into young C57 mice and assessed their memory capacity along with their brain and peripheral blood Aβ expression levels. The Aβ content in mouse platelets increased with age. Exogenously aged APP/PS1 platelets changed the permeability of the blood-brain barrier in vitro, accelerating Aβ deposition in the brain and increasing the Aβ content in peripheral blood, leading to learning and memory deficits in the recipient mice. Subsequently, aspirin was administered to mice as an inhibitor of platelet activation, which effectively alleviated these toxic processes. Finally, we chose an in vitro blood-brain barrier model to explore the possible cytotoxicity of these platelets.
Collapse
Affiliation(s)
- Tong Wu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Lizhi Chen
- Department of Science and Education, Guangdong Second Provincial General Hospital, Guangzhou, P.R. China
| | - Lingqi Zhou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Jie Xu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Kaihua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, P.R. China
| |
Collapse
|
10
|
Rojas-Garbanzo C, Rodríguez L, Pérez AM, Mayorga-Gross AL, Vásquez-Chaves V, Fuentes E, Palomo I. Anti-platelet activity and chemical characterization by UPLC-DAD-ESI-QTOF-MS of the main polyphenols in extracts from Psidium leaves and fruits. Food Res Int 2021; 141:110070. [PMID: 33641960 DOI: 10.1016/j.foodres.2020.110070] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 11/30/2022]
Abstract
In Costa Rica, two species of Psidium fruits, P. guajava and P. friedrichsthalianum, are widely consumed as food and used in folk medicine. Although studies have revealed the health effects of these fruits, there has been little research showing the antiplatelet activity of these species. This work evaluated the antiplatelet potential of aqueous extracts made from leaves and fruits of pink guava and Costa Rican guava. Platelet aggregation was induced by the platelet agonists ADP, TRAP-6, collagen and PMA. Platelet activation and secretion were studied using flow cytometry. The chemical profiles of the four extracts were characterized using UPLC-DAD-ESI-QTOF-MS. The studies revealed that the aqueous extracts of leaves and fruits of P. guajava and P. friedrichsthalianum inhibited platelet aggregation induced by ADP (4 µM), TRAP-6 (5 µM), collagen (1 µg mL-1) and PMA (100 nM), and the effect was dependent on the extract concentration. Extracts of leaves and fruits of pink guava and Costa Rican guava reduced secretion of P-selectin and activation of GP IIb/IIIa. The extracts of leaves and fruits of pink guava and Costa Rican guava proved to be a rich source of phenolic compounds, mainly quercetin aglycones and proanthocyanidins derived from (epi) catechin units. Other compounds such as ellagitannins, and benzophenones were also putatively identified. This research showed that P. guajava and P. friedrichsthalianum could potentially be used for the prevention of thrombotic events.
Collapse
Affiliation(s)
- Carolina Rojas-Garbanzo
- National Center of Food Science and Technology (CITA), University of Costa Rica, 11501-2060 San José, Costa Rica
| | - Lyanne Rodríguez
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, University of Talca, 3460000 Talca, Chile.
| | - Ana M Pérez
- National Center of Food Science and Technology (CITA), University of Costa Rica, 11501-2060 San José, Costa Rica.
| | - Ana Lucía Mayorga-Gross
- National Center of Food Science and Technology (CITA), University of Costa Rica, 11501-2060 San José, Costa Rica.
| | - Víctor Vásquez-Chaves
- Research Center in Natural Products (CIPRONA), University of Costa Rica, 11501-2060 San José, Costa Rica.
| | - Eduardo Fuentes
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, University of Talca, 3460000 Talca, Chile.
| | - Iván Palomo
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, University of Talca, 3460000 Talca, Chile.
| |
Collapse
|
11
|
Isas AS, Mariotti Celis MS, Pérez Correa JR, Fuentes E, Rodríguez L, Palomo I, Mozzi F, Van Nieuwenhove C. Functional fermented cherimoya (Annona cherimola Mill.) juice using autochthonous lactic acid bacteria. Food Res Int 2020; 138:109729. [DOI: 10.1016/j.foodres.2020.109729] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022]
|
12
|
Effects of the age/rage axis in the platelet activation. Int J Biol Macromol 2020; 166:1149-1161. [PMID: 33161078 DOI: 10.1016/j.ijbiomac.2020.10.270] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/28/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
Abstract
Platelet activity is essential in cardiovascular diseases. Therefore our objective was to evaluate the main effects of activating RAGE in platelets which are still unknown. A search for RAGE expression in different databases showed poor or a nonexistent presence in platelets. We confirmed the expression in platelets and secreted variable of RAGE (sRAGE). Platelets from elderly adults expressed in resting showed 3.2 fold more RAGE from young individuals (p < 0.01) and 3.3 fold with TRAP-6 (p < 0.001). These results could indicate that the expression of RAGE is more inducible in older adults. Then we found that activating RAGE with AGE-BSA-derived from methylglyoxal and subthreshold TRAP-6, showed a considerable increase with respect to the control in platelet aggregation and expression of P-selectin (respectively, p < 0.01). This effect was almost completely blocked by using a specific RAGE inhibitor (FSP-ZM1), confirming that RAGE is important for the function and activation platelet. Finally, we predict the region stimulated by AGE-BSA is located in region V of RAGE and 13 amino acids are critical for its binding. In conclusion, the activation of RAGE affects platelet activation and 13 amino acids are critical for its stimulation, this information is crucial for future possible treatments for CVD.
Collapse
|
13
|
Perényi H, Szegeczki V, Horváth G, Hinnah B, Tamás A, Radák Z, Ábrahám D, Zákány R, Reglodi D, Juhász T. Physical Activity Protects the Pathological Alterations of Alzheimer's Disease Kidneys via the Activation of PACAP and BMP Signaling Pathways. Front Cell Neurosci 2020; 14:243. [PMID: 32922265 PMCID: PMC7457084 DOI: 10.3389/fncel.2020.00243] [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: 05/19/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with typical amyloid beta (Aβ) aggregations. Elimination of the Aβ precursors via the kidneys makes the organ a potential factor in the systemic degeneration leading to AD. Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts neuroprotective effects in AD and plays a protective role in kidney pathologies. Increased physical activity is preventive of the formation of AD, but its detailed mechanism and possible connections with PACAP have not been clarified. In the kidneys of AD mice, the effects of physical activity were investigated by comparing wild-type and AD organs. Aβ plaque formation was reduced in AD kidneys after increased training (TAD). Mechanotransduction elevated PACAP receptor expression in TAD mice and normalized the protein kinase A (PKA)-mediated pathways. BMP4/BMPR1 elevation activated Smad1 expression and normalized collagen type IV in TAD animals. In conclusion, our data suggest that elevated physical activity can prevent the AD-induced pathological changes in the kidneys via, at least in part, the activation of PACAP-BMP signaling crosstalk.
Collapse
Affiliation(s)
- Helga Perényi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Vince Szegeczki
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabriella Horváth
- Department of Anatomy, PTE-MTA PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Barbara Hinnah
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andrea Tamás
- Department of Anatomy, PTE-MTA PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Zsolt Radák
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Dóra Ábrahám
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Róza Zákány
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dora Reglodi
- Department of Anatomy, PTE-MTA PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Tamás Juhász
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
14
|
Liraglutide Protects Against Brain Amyloid-β 1-42 Accumulation in Female Mice with Early Alzheimer's Disease-Like Pathology by Partially Rescuing Oxidative/Nitrosative Stress and Inflammation. Int J Mol Sci 2020; 21:ijms21051746. [PMID: 32143329 PMCID: PMC7084254 DOI: 10.3390/ijms21051746] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/23/2020] [Accepted: 02/28/2020] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia worldwide, being characterized by the deposition of senile plaques, neurofibrillary tangles (enriched in the amyloid beta (Aβ) peptide and hyperphosphorylated tau (p-tau), respectively) and memory loss. Aging, type 2 diabetes (T2D) and female sex (especially after menopause) are risk factors for AD, but their crosslinking mechanisms remain unclear. Most clinical trials targeting AD neuropathology failed and it remains incurable. However, evidence suggests that effective anti-T2D drugs, such as the GLP-1 mimetic and neuroprotector liraglutide, can be also efficient against AD. Thus, we aimed to study the benefits of a peripheral liraglutide treatment in AD female mice. We used blood and brain cortical lysates from 10-month-old 3xTg-AD female mice, treated for 28 days with liraglutide (0.2 mg/kg, once/day) to evaluate parameters affected in AD (e.g., Aβ and p-tau, motor and cognitive function, glucose metabolism, inflammation and oxidative/nitrosative stress). Despite the limited signs of cognitive changes in mature female mice, liraglutide only reduced their cortical Aβ1–42 levels. Liraglutide partially attenuated brain estradiol and GLP-1 and activated PKA levels, oxidative/nitrosative stress and inflammation in these AD female mice. Our results support the earlier use of liraglutide as a potential preventive/therapeutic agent against the accumulation of the first neuropathological features of AD in females.
Collapse
|