1
|
Pérez-González AP, de Anda-Jáuregui G, Hernández-Lemus E. Differential Transcriptional Programs Reveal Modular Network Rearrangements Associated with Late-Onset Alzheimer's Disease. Int J Mol Sci 2025; 26:2361. [PMID: 40076979 PMCID: PMC11900169 DOI: 10.3390/ijms26052361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is a complex, genetically heterogeneous disorder. The diverse phenotypes associated with AD result from interactions between genetic and environmental factors, influencing multiple biological pathways throughout disease progression. Network-based approaches offer a way to assess phenotype-specific states. In this study, we calculated key network metrics to characterize the network transcriptional structure and organization in LOAD, focusing on genes and pathways implicated in AD pathology within the dorsolateral prefrontal cortex (DLPFC). Our findings revealed disease-specific coexpression markers associated with diverse metabolic functions. Additionally, significant differences were observed at both the mesoscopic and local levels between AD and control networks, along with a restructuring of gene coexpression and biological functions into distinct transcriptional modules. These results show the molecular reorganization of the transcriptional program occurring in LOAD, highlighting specific adaptations that may contribute to or result from cellular responses to pathological stressors. Our findings may support the development of a unified model for the causal mechanisms of AD, suggesting that its diverse manifestations arise from multiple pathways working together to produce the disease's complex clinical patho-phenotype.
Collapse
Affiliation(s)
- Alejandra Paulina Pérez-González
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Programa de Doctorado en Ciencias Biomédicas, Unidad de Posgrado Edificio B Primer Piso, Ciudad Universitaria, Mexico City 04510, Mexico
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
| | - Guillermo de Anda-Jáuregui
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Investigadores por M’exico, Conahcyt, Mexico City 03940, Mexico
| | - Enrique Hernández-Lemus
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
2
|
Zhao M, Lin Y, Zeng Y, Lv Z, Liang J, Tang P, Zhen X, Han L. Biomimetic membrane-coated nanoparticles specially permeate the inflammatory blood-brain barrier to deliver plasmin therapy for brain metastases. J Control Release 2025; 378:763-775. [PMID: 39732369 DOI: 10.1016/j.jconrel.2024.12.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/13/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Many brain-targeting drug delivery strategies have been reported to permeate the blood-brain barrier (BBB) via hijacking receptor-mediated transport. However, these receptor-based strategies could mediate whole-brain BBB crossing due to the wide intracranial expression of target receptors and lead to unwanted accumulation and side effects on healthy brain tissues. Inspired by brain metastatic processes and the selectivity of brain metastatic cancer cells for the inflammatory BBB, a biomimetic nanoparticle was developed by coating drug-loaded core with the inflammatory BBB-seeking erythrocyte-brain metastatic hybrid membrane, which can resist homotypic aggregation and specially bind and permeate the inflammatory BBB for specific drug delivery. Dexamethasone and embelin were used as model drugs to be loaded in the biomimetic nanoparticles to restore plasmin-mediated attacks against brain metastases. The drug-loaded nanoparticles were proved to inhibit tumor serpin secretion to restore local plasmin production, which could inactivate tumor cell surface L1CAM to inhibit vessel-spreading-dependent tumor growth and produce lethal soluble factor-related apoptosis ligands (sFasL) to induce tumor cell apoptosis, leading to the suppression of the intracranial metastatic nodule development and prolonged survival of mice with brain metastases. The inflammatory BBB-seeking biomimetic approach represents an effective regimen for potent therapy against brain metastases.
Collapse
Affiliation(s)
- Mei Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuanyuan Lin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuteng Zeng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ziyan Lv
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jiayu Liang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Puxian Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Liang Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| |
Collapse
|
3
|
Zhou M, Lin Y, Chen H, Zhao M, Zeng Y, Hu X, Tang P, Fu Y, Wei L, Han L. Brain-tumor-seeking and serpin-inhibiting outer membrane vesicles restore plasmin-mediated attacks against brain metastases. J Control Release 2024; 375:116-126. [PMID: 39236899 DOI: 10.1016/j.jconrel.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Many chemotherapeutic and molecular targeted drugs have been used to treat brain metastases, e.g., anti-angiogenic vandetanib. However, the blood-brain barrier and brain-specific resistance mechanisms make these systemic therapeutic approaches inefficacious. Brain metastatic cancer cells could mimic neurons to upregulate multiple serpins and secrete them into the extracellular environment to reduce local plasmin production to promote L1CAM-mediated vessel co-option and resist anti-angiogenesis therapy. Here, we developed brain-tumor-seeking and serpin-inhibiting outer membrane vesicles (DE@OMVs) to traverse across the blood-brain barrier, bypass neurons, and specially enter metastatic cancer cells via targeting GRP94 and vimentin. Through specific delivery of dexamethasone and embelin, reduced serpin secretion, restored plasmin production, significant L1CAM inactivation and tumor cell apoptosis were specially found in intracranial metastatic regions, leading to delayed tumor growth and prolonged survival in mice with brain metastases. By combining the brain-tumor-seeking properties with the regulation of the serpin/plasminogen activator/plasmin/L1CAM axis, this study provides a potent and highly-selective systemic therapeutic option for brain metastases.
Collapse
Affiliation(s)
- Mengyuan Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Yuanyuan Lin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Haiyan Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mei Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuteng Zeng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xiaoxiao Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Puxian Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuxuan Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lin Wei
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China; School of Life Sciences, Anhui Medical University, Hefei 230032, Anhui, China.
| | - Liang Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| |
Collapse
|
4
|
Brenna S, Glatzel M, Magnus T, Puig B, Galliciotti G. Neuroserpin and Extracellular Vesicles in Ischemic Stroke: Partners in Neuroprotection? Aging Dis 2024; 15:2191-2204. [PMID: 39191396 PMCID: PMC11346402 DOI: 10.14336/ad.2024.0518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/05/2024] [Indexed: 08/29/2024] Open
Abstract
Ischemic stroke represents a significant global health challenge, often resulting in death or long-term disability, particularly among the elderly, where advancing age stands as the most unmodifiable risk factor. Arising from the blockage of a brain-feeding artery, the only therapies available to date aim at removing the blood clot to restore cerebral blood flow and rescue neuronal cells from death. The prevailing treatment approach involves thrombolysis by administration of recombinant tissue plasminogen activator (tPA), albeit with a critical time constraint. Timely intervention is imperative, given that delayed thrombolysis increases tPA leakage into the brain parenchyma, causing harmful effects. Strategies to preserve tPA's vascular benefits while shielding brain cells from its toxicity have been explored. Notably, administering neuroserpin (Ns), a brain-specific tPA inhibitor, represents one such approach. Following ischemic stroke, Ns levels rise and correlate with favorable post-stroke outcomes. Studies in rodent models of focal cerebral ischemia have demonstrated the beneficial effects of Ns administration. Ns treatment maintains blood-brain barrier (BBB) integrity, reducing stroke volume. Conversely, Ns-deficient animals exhibit larger stroke injury, increased BBB permeability and enhanced microglia activation. Furthermore, Ns administration extends the therapeutic window for tPA intervention, underscoring its potential in stroke management. Remarkably, our investigation reveals the presence of Ns within extracellular vesicles (EVs), small membrane-surrounded particles released by all cells and critical for intercellular communication. EVs influence disease outcome following stroke through cargo transfer between cells. Clarifying the role of EVs containing NS could open up urgently needed novel therapeutic approaches to improve post-ischemic stroke outcome.
Collapse
Affiliation(s)
- Santra Brenna
- Experimental Research in Stroke and Inflammation (ERSI) Group, Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tim Magnus
- Experimental Research in Stroke and Inflammation (ERSI) Group, Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Berta Puig
- Experimental Research in Stroke and Inflammation (ERSI) Group, Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Giovanna Galliciotti
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
5
|
Chen H, Lu Z, Ni X, Zhang H, Chen G, Wu X, Ding M. The development of anti-PD-1 antibody-induced spinal cord injury in bone marrow transplant C57BL/6 Rag1-/- mouse model. Immunotherapy 2024; 16:975-985. [PMID: 39115961 PMCID: PMC11486090 DOI: 10.1080/1750743x.2024.2383557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Aims: This paper was to scrutinize the toxicity mechanism of anti-programmed death 1 (anti-PD-1) therapy-caused spinal cord injury (SCI).Methods: Bone marrow transplant Rag1-/- mice were used to establish SCI model.Results: Anti-PD-1 results in SCI via CD8+ T-cells activation, while excessive activation of CD8+ T-cells further aggravated SCI. Both anti-PD-1 and the activation of CD8+ T-cells induced the expression of apoptosis-related perforin, GrB and FasL, but suppressed PI-9 level. The opposite results were observed in the effects of neuroserpin on these factors. CD8+ T-cells activation induced neurotoxicity via upregulation perforin, GrB and FasL and inhibiting PI-9. Additionally, neuroserpin suppressed CD8+ T-cells activation via perforin/GrB/PI-9/FasL pathways.Conclusion: These results may provide theoretical foundation for the clinical treatment of SCI caused by anti-PD-1.
Collapse
Affiliation(s)
- Huachun Chen
- Department of Respiratory & Critical Care Medicine, Jinhua Guangfu Cancer Hospital, Jinhua, 321001, Zhejiang, China
| | - Zhouxiao Lu
- Department of Respiratory & Critical Care Medicine, Jinhua Guangfu Cancer Hospital, Jinhua, 321001, Zhejiang, China
| | - Xiaowei Ni
- Department of Respiratory & Critical Care Medicine, Jinhua Guangfu Cancer Hospital, Jinhua, 321001, Zhejiang, China
| | - Hui Zhang
- Institute of Pharmacology, Jinhua Food & Drug Inspection & Testing Research Institute, Jinhua, 321002, Zhejiang, China
| | - Guiyuan Chen
- School of Medicine, Jinhua Polytechnic, Jinhua, 321007, Zhejiang, China
| | - Xiaoyu Wu
- Department of Respiratory & Critical Care Medicine, Jinhua Guangfu Cancer Hospital, Jinhua, 321001, Zhejiang, China
| | - Mingxing Ding
- School of Medicine, Jinhua Polytechnic, Jinhua, 321007, Zhejiang, China
| |
Collapse
|
6
|
Kilicdag H, Akillioglu K, Kilic Bagır E, Kose S, Erdogan S. Neuroserpin As an Adjuvant Therapy for Hypothermia on Brain Injury in Neonatal Hypoxic-Ischemic Rats. Am J Perinatol 2024; 41:1538-1543. [PMID: 37611639 DOI: 10.1055/a-2159-0488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
OBJECTIVE We aimed to assess the effects of neuroserpin and its combination with hypothermia on hypoxic-ischemic (HI) brain injury in neonatal rats. Neuroserpin is an axon-secreted serine protease inhibitor and is important for brain development, neuronal survival, and synaptic plasticity. STUDY DESIGN Male Wistar-Albino rats on postnatal day 7 (P7) were randomly divided into five groups: sham group (n = 10), (HI; n = 10), hypoxic-ischemic hypothermia (HIH; n = 10), hypoxic-ischemic neuroserpin (HIN; n = 10), and hypoxic-ischemic neuroserpin-hypothermia (HINH; n = 10). The P7 rat brain's maturation is similar to a late preterm human brain at 34 to 36 weeks of gestation. HI was induced in rats on P7 as previously described. A single dose of 0.2 µM neuroserpin (HINH and HIN) or an equivalent volume of phosphate-buffered saline (sham, HIH, and HI) was administered intraventricularly by a Hamilton syringe immediately after hypoxia. In the follow-up, pups were subjected to systemic hypothermia or normothermia for 2 hours. Euthanasia was performed for histopathological evaluation on P10. Apoptosis was detected by caspase-3 activity and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining and was counted in the hippocampus. RESULTS In comparison to the HI group, the TUNEL-positive and caspase-3-positive neurons in the sham, HIN, HIH, and HINH groups were considerably lower (13.4 ± 1.0 vs. 1.9 ± 0.9, 6.0 ± 0.9, 5.3 ± 1.6, and 4.0 ± 1.1; p < 0.001) and (13.5 ± 1.7 vs. 1.2 ± 0.7, 9.1 ± 2.7, 4.8 ± 1.0, and 3.9 ± 1.6; p < 0.001). HIN, HIH, and HINH, compared to the sham group, showed more TUNEL-positive and caspase-3-positive neurons (6.0 ± 0.9, 5.3 ± 1.6, 4.0 ± 1.1 vs. 1.9 ± 0.9 and 9.1 ± 2.7, 4.8 ± 1.0, 3.9 ± 1.6 vs. 1.2 ± 0.7; p < 0.001). The HINH group (synergistic effect) had significantly fewer TUNEL-positive neurons and caspase-3-positive neurons than the HIN group (4.0 ± 1.1 vs. 6.0 ± 0.9 and 3.9 ± 1.6 vs. 9.1 ± 2.7; p < 0.001). CONCLUSION Our study showed that both neuroserpin alone and as an adjuvant treatment for hypothermia may have a neuroprotective effect on brain injury. KEY POINTS · Neuroserpin decreased brain injury.. · Neuroserpin showed a synergistic effect when used as an adjuvant treatment for hypothermia.. · The neuroprotective effect of neuroserpine was related to its antiapoptotic properties..
Collapse
Affiliation(s)
- Hasan Kilicdag
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Kubra Akillioglu
- Division of Neurophysiology, Department of Physiology, Medical Faculty, University of Cukurova, Turkey
| | - Emine Kilic Bagır
- Department of Pathology, Cukurova University, Medical Faculty, Adana, Turkey
| | - Seda Kose
- Division of Neurophysiology, Department of Physiology, Medical Faculty, University of Cukurova, Turkey
| | - Seyda Erdogan
- Department of Pathology, Cukurova University, Medical Faculty, Adana, Turkey
| |
Collapse
|
7
|
Yepes M. Reprint of: Fibrinolytic and Non-fibrinolytic Roles of Tissue-type Plasminogen Activator in the Ischemic Brain. Neuroscience 2024; 550:21-29. [PMID: 38964373 DOI: 10.1016/j.neuroscience.2024.05.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/06/2023] [Indexed: 07/06/2024]
Abstract
The neurovascular unit (NVU) is assembled by endothelial cells (ECs) and pericytes, and encased by a basement membrane (BM) surveilled by microglia and surrounded by perivascular astrocytes (PVA), which in turn are in contact with synapses. Cerebral ischemia induces the rapid release of the serine proteinase tissue-type plasminogen activator (tPA) from endothelial cells, perivascular astrocytes, microglia and neurons. Owning to its ability to catalyze the conversion of plasminogen into plasmin, in the intravascular space tPA functions as a fibrinolytic enzyme. In contrast, the release of astrocytic, microglial and neuronal tPA have a plethora of effects that not always require the generation of plasmin. In the ischemic brain tPA increases the permeability of the NVU, induces microglial activation, participates in the recycling of glutamate, and has various effects on neuronal survival. These effects are mediated by different receptors, notably subunits of the N-methyl-D-aspartate receptor (NMDAR) and the low-density lipoprotein receptor-related protein-1 (LRP-1). Here we review data on the role of tPA in the NVU under non-ischemic and ischemic conditions, and analyze how this knowledge may lead to the development of potential strategies for the treatment of acute ischemic stroke patients.
Collapse
Affiliation(s)
- Manuel Yepes
- Department of Neurology, Emory University, Atlanta, GA, USA; Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA; Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA.
| |
Collapse
|
8
|
Janciauskiene S, Lechowicz U, Pelc M, Olejnicka B, Chorostowska-Wynimko J. Diagnostic and therapeutic value of human serpin family proteins. Biomed Pharmacother 2024; 175:116618. [PMID: 38678961 DOI: 10.1016/j.biopha.2024.116618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
SERPIN (serine proteinase inhibitors) is an acronym for the superfamily of structurally similar proteins found in animals, plants, bacteria, viruses, and archaea. Over 1500 SERPINs are known in nature, while only 37 SERPINs are found in humans, which participate in inflammation, coagulation, angiogenesis, cell viability, and other pathophysiological processes. Both qualitative or quantitative deficiencies or overexpression and/or abnormal accumulation of SERPIN can lead to diseases commonly referred to as "serpinopathies". Hence, strategies involving SERPIN supplementation, elimination, or correction are utilized and/or under consideration. In this review, we discuss relationships between certain SERPINs and diseases as well as putative strategies for the clinical explorations of SERPINs.
Collapse
Affiliation(s)
- Sabina Janciauskiene
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany; Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland
| | - Magdalena Pelc
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland
| | - Beata Olejnicka
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka St, Warsaw 01-138, Poland.
| |
Collapse
|
9
|
Challa SR, Nalamolu KR, Fornal CA, Baker IM, Mohandass A, Mada SR, Wang BC, Pinson DM, Lahoti S, Klopfenstein JD, Veeravalli KK. The paradox of tPA in ischemic stroke: tPA knockdown following recanalization improves functional and histological outcomes. Exp Neurol 2024; 374:114727. [PMID: 38360257 PMCID: PMC10986679 DOI: 10.1016/j.expneurol.2024.114727] [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: 12/28/2023] [Revised: 02/01/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
Previous studies have demonstrated that endogenous tissue-type plasminogen activator (tPA) is upregulated in the brain after an acute ischemic stroke (AIS). While mixed results were observed in genetic models, the pharmacological inhibition of endogenous tPA showed beneficial effects. Treatment with exogenous recombinant tPA exacerbated brain damage in rodent models of stroke. Despite the detrimental effects of tPA in ischemic stroke, recombinant tPA is administered to AIS patients to recanalize the occluded blood vessels because the benefits of its administration outweigh the risks associated with tPA upregulation and increased activity. We hypothesized that tPA knockdown following recanalization would ameliorate sensorimotor deficits and reduce brain injury. Young male and female rats (2-3 months old) were subjected to transient focal cerebral ischemia by occlusion of the right middle cerebral artery. Shortly after reperfusion, rats from appropriate cohorts were administered a nanoparticle formulation containing tPA shRNA or control shRNA plasmids (1 mg/kg) intravenously via the tail vein. Infarct volume during acute and chronic phases, expression of matrix metalloproteinases (MMPs) 1, 3, and 9, enlargement of cerebral ventricle volume, and white matter damage were all reduced by shRNA-mediated gene silencing of tPA following reperfusion. Additionally, recovery of somatosensory and motor functions was improved. In conclusion, our results provide evidence that reducing endogenous tPA following recanalization improves functional outcomes and reduces post-stroke brain damage.
Collapse
Affiliation(s)
- Siva Reddy Challa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA; Department of Pharmacology, KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, AP, India
| | - Koteswara Rao Nalamolu
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Casimir A Fornal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Isidra M Baker
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Adithya Mohandass
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Sahil Reddy Mada
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Billy C Wang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA; Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL, USA; Pediatric Critical Care Medicine, OSF HealthCare Saint Francis Medical Center, Peoria, IL, USA
| | - David M Pinson
- Department of Health Sciences Education and Pathology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Sourabh Lahoti
- Department of Neurology, University of Illinois College of Medicine Peoria, Peoria, IL, USA; Illinois Neurological Institute, OSF HealthCare Saint Francis Medical Center, Peoria, IL, USA
| | - Jeffrey D Klopfenstein
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA; Illinois Neurological Institute, OSF HealthCare Saint Francis Medical Center, Peoria, IL, USA; Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA; Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL, USA; Department of Neurology, University of Illinois College of Medicine Peoria, Peoria, IL, USA; Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL, USA.
| |
Collapse
|
10
|
Yepes M. Fibrinolytic and Non-fibrinolytic Roles of Tissue-type Plasminogen Activator in the Ischemic Brain. Neuroscience 2024; 542:69-80. [PMID: 37574107 DOI: 10.1016/j.neuroscience.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/03/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
The neurovascular unit (NVU) is assembled by endothelial cells (ECs) and pericytes, and encased by a basement membrane (BM) surveilled by microglia and surrounded by perivascular astrocytes (PVA), which in turn are in contact with synapses. Cerebral ischemia induces the rapid release of the serine proteinase tissue-type plasminogen activator (tPA) from endothelial cells, perivascular astrocytes, microglia and neurons. Owning to its ability to catalyze the conversion of plasminogen into plasmin, in the intravascular space tPA functions as a fibrinolytic enzyme. In contrast, the release of astrocytic, microglial and neuronal tPA have a plethora of effects that not always require the generation of plasmin. In the ischemic brain tPA increases the permeability of the NVU, induces microglial activation, participates in the recycling of glutamate, and has various effects on neuronal survival. These effects are mediated by different receptors, notably subunits of the N-methyl-D-aspartate receptor (NMDAR) and the low-density lipoprotein receptor-related protein-1 (LRP-1). Here we review data on the role of tPA in the NVU under non-ischemic and ischemic conditions, and analyze how this knowledge may lead to the development of potential strategies for the treatment of acute ischemic stroke patients.
Collapse
Affiliation(s)
- Manuel Yepes
- Department of Neurology, Emory University, Atlanta, GA, USA; Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA; Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA.
| |
Collapse
|
11
|
He W, Gu L, Yang J, Zhang R, Long J, Peng W, Liang B, Zhu L, Lv M, Nan A, Su L. Exosomal circCNOT6L Regulates Astrocyte Apoptotic Signals Induced by Hypoxia Exposure Through miR99a-5p/SERPINE1 and Alleviates Ischemic Stroke Injury. Mol Neurobiol 2023; 60:7118-7135. [PMID: 37531026 DOI: 10.1007/s12035-023-03518-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 07/12/2023] [Indexed: 08/03/2023]
Abstract
Circular RNAs are involved in intervention strategies for treating ischemic stroke (IS). However, circCNOT6L (hsa_circ_0006168) has not yet been reported in IS. Thus, we aimed to explore the potential role of circCNOT6L and its molecular mechanism in IS. In this study, we first found that the expression of both exosomal circCNOT6L (P = 0.0006) and plasma circCNOT6L (P = 0.0054) was down-regulated in IS patients compared with controls. Clinically, a negative correlation was observed between the relative expression level of circCNOT6L and the National Institutes of Health Stroke Scale (NIHSS) score and infarct volume of the brain. Simultaneously, the relative expression level of circCNOT6L was negatively associated with multiple risk factors for IS, such as mean platelet volume (MPV), red cell distribution width (RDW), very low-density lipoprotein (VLDL), and serum potassium, whereas it was positively correlated with high-density lipoprotein (HDL). In vitro, circCNOT6L silencing blocked cell viability and proliferation, while it promoted cell apoptosis of astrocytes undergoing oxygen-glucose deprivation/reperfusion (OGD/R) treatment. Mechanistically, the RNA antisense purification (RAP) assay and luciferase reporter assay revealed that circCNOT6L acts as a miRNA sponge to absorb miR-99a-5p and then regulates the expression of serine proteinase inhibitor (SERPINE1). In the further rescue experiment, overexpressing SERPINE1 could rescue the cell apoptotic signals due to circCNOT6L depletion. In conclusion, CircCNOT6L attenuated the cell apoptotic signal of astrocytes via the miR99a-5p/SERPINE1 axis and then alleviated injury after hypoxia induced by ischemic stroke.
Collapse
Affiliation(s)
- Wanting He
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lian Gu
- First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jialei Yang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ruirui Zhang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Jianxiong Long
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenyi Peng
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Baoyun Liang
- First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Lulu Zhu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Miao Lv
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Aruo Nan
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China.
| | - Li Su
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
12
|
Chmelova M, Androvic P, Kirdajova D, Tureckova J, Kriska J, Valihrach L, Anderova M, Vargova L. A view of the genetic and proteomic profile of extracellular matrix molecules in aging and stroke. Front Cell Neurosci 2023; 17:1296455. [PMID: 38107409 PMCID: PMC10723838 DOI: 10.3389/fncel.2023.1296455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/08/2023] [Indexed: 12/19/2023] Open
Abstract
Introduction Modification of the extracellular matrix (ECM) is one of the major processes in the pathology of brain damage following an ischemic stroke. However, our understanding of how age-related ECM alterations may affect stroke pathophysiology and its outcome is still very limited. Methods We conducted an ECM-targeted re-analysis of our previously obtained RNA-Seq dataset of aging, ischemic stroke and their interactions in young adult (3-month-old) and aged (18-month-old) mice. The permanent middle cerebral artery occlusion (pMCAo) in rodents was used as a model of ischemic stroke. Altogether 56 genes of interest were chosen for this study. Results We identified an increased activation of the genes encoding proteins related to ECM degradation, such as matrix metalloproteinases (MMPs), proteases of a disintegrin and metalloproteinase with the thrombospondin motifs (ADAMTS) family and molecules that regulate their activity, tissue inhibitors of metalloproteinases (TIMPs). Moreover, significant upregulation was also detected in the mRNA of other ECM molecules, such as proteoglycans, syndecans and link proteins. Notably, we identified 8 genes where this upregulation was enhanced in aged mice in comparison with the young ones. Ischemia evoked a significant downregulation in only 6 of our genes of interest, including those encoding proteins associated with the protective function of ECM molecules (e.g., brevican, Hapln4, Sparcl1); downregulation in brevican was more prominent in aged mice. The study was expanded by proteome analysis, where we observed an ischemia-induced overexpression in three proteins, which are associated with neuroinflammation (fibronectin and vitronectin) and neurodegeneration (link protein Hapln2). In fibronectin and Hapln2, this overexpression was more pronounced in aged post-ischemic animals. Conclusion Based on these results, we can conclude that the ratio between the protecting and degrading mechanisms in the aged brain is shifted toward degradation and contributes to the aged tissues' increased sensitivity to ischemic insults. Altogether, our data provide fresh perspectives on the processes underlying ischemic injury in the aging brain and serve as a freely accessible resource for upcoming research.
Collapse
Affiliation(s)
- Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Peter Androvic
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences – BIOCEV, Vestec, Czechia
| | - Denisa Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Lukas Valihrach
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences – BIOCEV, Vestec, Czechia
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
13
|
Chitranshi N, Rajput R, Godinez A, Pushpitha K, Mirzaei M, Basavarajappa D, Gupta V, Sharma S, You Y, Galliciotti G, Salekdeh GH, Baker MS, Graham SL, Gupta VK. Neuroserpin gene therapy inhibits retinal ganglion cell apoptosis and promotes functional preservation in glaucoma. Mol Ther 2023; 31:2056-2076. [PMID: 36905120 PMCID: PMC10362384 DOI: 10.1016/j.ymthe.2023.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/27/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Our research has proven that the inhibitory activity of the serine protease inhibitor neuroserpin (NS) is impaired because of its oxidation deactivation in glaucoma. Using genetic NS knockout (NS-/-) and NS overexpression (NS+/+ Tg) animal models and antibody-based neutralization approaches, we demonstrate that NS loss is detrimental to retinal structure and function. NS ablation was associated with perturbations in autophagy and microglial and synaptic markers, leading to significantly enhanced IBA1, PSD95, beclin-1, and LC3-II/LC3-I ratio and reduced phosphorylated neurofilament heavy chain (pNFH) levels. On the other hand, NS upregulation promoted retinal ganglion cell (RGC) survival in wild-type and NS-/- glaucomatous mice and increased pNFH expression. NS+/+Tg mice demonstrated decreased PSD95, beclin-1, LC3-II/LC3-I ratio, and IBA1 following glaucoma induction, highlighting its protective role. We generated a novel reactive site NS variant (M363R-NS) resistant to oxidative deactivation. Intravitreal administration of M363R-NS was observed to rescue the RGC degenerative phenotype in NS-/- mice. These findings demonstrate that NS dysfunction plays a key role in the glaucoma inner retinal degenerative phenotype and that modulating NS imparts significant protection to the retina. NS upregulation protected RGC function and restored biochemical networks associated with autophagy and microglial and synaptic function in glaucoma.
Collapse
Affiliation(s)
- Nitin Chitranshi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.
| | - Rashi Rajput
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Angela Godinez
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Kanishka Pushpitha
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Melbourne, VIC, Australia
| | - Samridhi Sharma
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Yuyi You
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Giovanna Galliciotti
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ghasem H Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Mark S Baker
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
14
|
Toomer KH, Gerber GF, Zhang Y, Daou L, Tushek M, Hooper JE, Francischetti IMB. SARS-CoV-2 infection results in upregulation of Plasminogen Activator Inhibitor-1 and Neuroserpin in the lungs, and an increase in fibrinolysis inhibitors associated with disease severity. EJHAEM 2023; 4:324-338. [PMID: 37206290 PMCID: PMC10188457 DOI: 10.1002/jha2.654] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/14/2023] [Accepted: 01/21/2023] [Indexed: 05/21/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection results in coagulation activation although it is usually not associated with consumption coagulopathy. D-dimers are also commonly elevated despite systemic hypofibrinolysis. To understand these unusual features of coronavirus disease 2019 (COVID-19) coagulopathy, 64 adult patients with SARS-CoV-2 infection (36 moderate and 28 severe) and 16 controls were studied. We evaluated the repertoire of plasma protease inhibitors (Serpins, Kunitz, Kazal, Cystatin-like) targeting the fibrinolytic system: Plasminogen Activator Inhibitor-1 (PAI-1), Tissue Plasminogen Activator/Plasminogen Activator Inhibitor-1 complex (t-PA/PAI-1), α-2-Antiplasmin, Plasmin-α2-Antiplasmin Complex, Thrombin-activatable Fibrinolysis Inhibitor (TAFI)/TAFIa, Protease Nexin-1 (PN-1), and Neuroserpin (the main t-PA inhibitor of the central nervous system). Inhibitors of the common (Antithrombin, Thrombin-antithrombin complex, Protein Z [PZ]/PZ inhibitor, Heparin Cofactor II, and α2-Macroglobulin), Protein C ([PC], Protein C inhibitor, and Protein S), contact (Kallistatin, Protease Nexin-2/Amyloid Beta Precursor Protein, and α-1-Antitrypsin), and complement (C1-Inhibitor) pathways, in addition to Factor XIII, Histidine-rich glycoprotein (HRG) and Vaspin were also investigated by enzyme-linked immunosorbent assay. The association of these markers with disease severity was evaluated by logistic regression. Pulmonary expression of PAI-1 and Neuroserpin in the lungs from eight post-mortem cases was assessed by immunohistochemistry. Results show that six patients (10%) developed thrombotic events, and mortality was 11%. There was no significant reduction in plasma anticoagulants, in keeping with a compensated state. However, an increase in fibrinolysis inhibitors (PAI-1, Neuroserpin, PN-1, PAP, and t-PA/PAI-1) was consistently observed, while HRG was reduced. Furthermore, these markers were associated with moderate and/or severe disease. Notably, immunostains demonstrated overexpression of PAI-1 in epithelial cells, macrophages, and endothelial cells of fatal COVID-19, while Neuroserpin was found in intraalveolar macrophages only. These results imply that the lungs in SARS-CoV-2 infection provide anti-fibrinolytic activity resulting in a shift toward a local and systemic hypofibrinolytic state predisposing to (immuno)thrombosis, often in a background of compensated disseminated intravascular coagulation.
Collapse
Affiliation(s)
- Kevin H. Toomer
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Gloria F. Gerber
- Division of HematologyDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Yifan Zhang
- Department of BiostatisticsJohns Hopkins University Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Laetitia Daou
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Michael Tushek
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jody E. Hooper
- Department of PathologyStanford University School of MedicinePalo AltoCaliforniaUSA
| | | |
Collapse
|
15
|
Satapathy S, Wilson MR. Roles of constitutively secreted extracellular chaperones in neuronal cell repair and regeneration. Neural Regen Res 2023; 18:769-772. [PMID: 36204835 PMCID: PMC9700095 DOI: 10.4103/1673-5374.353483] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 12/05/2022] Open
Abstract
Protein quality control involves many processes that jointly act to regulate the expression, localization, turnover, and degradation of proteins, and has been highlighted in recent studies as critical to the differentiation of stem cells during regeneration. The roles of constitutively secreted extracellular chaperones in neuronal injury and disease are poorly understood. Extracellular chaperones are multifunctional proteins expressed by many cell types, including those of the nervous system, known to facilitate protein quality control processes. These molecules exert pleiotropic effects and have been implicated as playing important protective roles in a variety of stress conditions, including tissue damage, infections, and local tissue inflammation. This article aims to provide a critical review of what is currently known about the functions of extracellular chaperones in neuronal repair and regeneration and highlight future directions for this important research area. We review what is known of four constitutively secreted extracellular chaperones directly implicated in processes of neuronal damage and repair, including transthyretin, clusterin, α2-macroglobulin, and neuroserpin, and propose that investigation into the effects of these and other extracellular chaperones on neuronal repair and regeneration has the potential to yield valuable new therapies.
Collapse
Affiliation(s)
- Sandeep Satapathy
- Blavatnik Institute of Cell Biology, Harvard Medical School, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mark R. Wilson
- Molecular Horizons and The School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, Australia
| |
Collapse
|
16
|
Challa SR, Nalamolu KR, Fornal CA, Mohandass A, Mussman JP, Schaibley C, Kashyap A, Sama V, Wang BC, Klopfenstein JD, Pinson DM, Kunamneni A, Veeravalli KK. The interplay between MMP-12 and t-PA in the brain after ischemic stroke. Neurochem Int 2022; 161:105436. [PMID: 36283468 PMCID: PMC9898869 DOI: 10.1016/j.neuint.2022.105436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022]
Abstract
Tissue-type plasminogen activator (t-PA) expression is known to increase following transient focal cerebral ischemia and reperfusion. Previously, we reported downregulation of t-PA upon suppression of matrix metalloproteinase-12 (MMP-12), following transient focal cerebral ischemia and reperfusion. We now present data on the temporal expression of t-PA in the brain after transient ischemia, as well as the interaction between MMP-12 and t-PA, two proteases associated with the breakdown of the blood-brain barrier (BBB) and ischemic brain damage. We hypothesized that there might be reciprocal interactions between MMP-12 and t-PA in the brain after ischemic stroke. This hypothesis was tested using shRNA-mediated gene silencing and computational modeling. Suppression of t-PA following transient ischemia and reperfusion in rats attenuated MMP-12 expression in the brain. The overall effect of t-PA shRNA administration was to attenuate the degradation of BBB tight junction protein claudin-5, diminish BBB disruption, and reduce neuroinflammation by decreasing the expression of the microglia/macrophage pro-inflammatory M1 phenotype (CD68, iNOS, IL-1β, and TNFα). Reduced BBB disruption and subsequent lack of infiltration of macrophages (the main source of MMP-12 in the ischemic brain) could account for the decrease in MMP-12 expression after t-PA suppression. Computational modeling of in silico protein-protein interactions indicated that MMP-12 and t-PA may interact physically. Overall, our findings demonstrate that MMP-12 and t-PA interact directly or indirectly at multiple levels in the brain following an ischemic stroke. The present findings could be useful in the development of new pharmacotherapies for the treatment of stroke.
Collapse
Affiliation(s)
- Siva Reddy Challa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA; Department of Pharmacology, KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, Andhra Pradesh, India
| | - Koteswara Rao Nalamolu
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Casimir A Fornal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Adithya Mohandass
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Justin P Mussman
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Claire Schaibley
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Aanan Kashyap
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Vinay Sama
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Billy C Wang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA; Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, IL, USA; Children's Hospital of Illinois, OSF HealthCare Saint Francis Medical Center, Peoria, IL, USA
| | - Jeffrey D Klopfenstein
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA; Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA; Illinois Neurological Institute, OSF HealthCare Saint Francis Medical Center, Peoria, IL, USA
| | - David M Pinson
- Department of Health Sciences Education and Pathology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | | | - Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA; Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, IL, USA; Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA; Department of Neurology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA.
| |
Collapse
|
17
|
Su EJ, Lawrence DA. Diabetes and the treatment of ischemic stroke. J Diabetes Complications 2022; 36:108318. [PMID: 36228562 DOI: 10.1016/j.jdiacomp.2022.108318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/23/2022] [Indexed: 11/23/2022]
Abstract
This white paper examines the current challenges for treating ischemic stroke in diabetic patients. The need for a greater understanding of the mechanisms that underlie the relationship between diabetes and the cerebral vascular responses to ischemia is discussed. The critical need to improve the efficacy and safety of thrombolysis is addressed, as is the need for a better characterization the off-target actions of tPA, the only currently approved thrombolytic for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Enming J Su
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel A Lawrence
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Fatima S, Ansari S, Bano S, Ahamad S, Ishqi HM, Tabish M, Gupta D, Rehman SU, Jairajpuri MA. Detection of truncated isoforms of human neuroserpin lacking the reactive center loop: Implications in noninhibitory role. IUBMB Life 2021; 73:941-952. [PMID: 33893722 DOI: 10.1002/iub.2475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/23/2021] [Accepted: 04/13/2021] [Indexed: 12/27/2022]
Abstract
Neuroserpin is a serine protease inhibitor expressed mainly in the brain and at low levels in other tissues like the kidney, testis, heart, and spinal cord. It is involved in the inhibition of tissue plasminogen activator (tPA), plasmin, and to a lesser extent, urokinase-type plasminogen (uPA). Neuroserpin has also been shown to plays noninhibitory roles in the regulation of N-cadherin-mediated cell adhesion. It is involved in neuroprotection from seizure and stroke through tPA-mediated inhibition and also through its other protease targets. Mutations in critical domains of neuroserpin lead to its polymerization and neuronal death. In this study, a novel truncated isoform of human neuroserpin was identified in the brain and liver, which was confirmed by reverse transcriptase-PCR and DNA sequencing using exon-specific primers. Structural characterization of novel isoform using MD simulations studies indicated that it lacks the reactive center loop (RCL) but largely maintains its secondary structure fold. The novel truncated variant was cloned, expressed, and purified. A comparative intrinsic fluorescence and 4,4'-bis-1-anilino naphthalene 8-sulfonate studies revealed a decrease in fluorescence emission intensity and a more exposed hydrophobic surface as compared to the reported isoform. However, the novel isoform has lost its ability for tPA inhibition and complex formation. The absence of RCL indicates a noninhibitory role for the truncated isoform, prompting a detailed search and identification of two smaller isoforms in the human brain. With indications of the noninhibitory role of neuroserpin, identifying novel isoforms that appear to be without the tPA recognition domain is significant.
Collapse
Affiliation(s)
- Sana Fatima
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Shoyab Ansari
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Shadabi Bano
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Shahzaib Ahamad
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Hassan Mubarak Ishqi
- Molecular Genetics Laboratory, National Institute of Immunology, New Delhi, India
- Department of Biochemistry, Faculty of Life Sciences, Aligarh M. University, Aligarh, Uttar Pradesh, India
| | - Mohammad Tabish
- Department of Biochemistry, Faculty of Life Sciences, Aligarh M. University, Aligarh, Uttar Pradesh, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sayeed Ur Rehman
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | | |
Collapse
|
19
|
Yepes M. The Plasminogen Activation System Promotes Neurorepair in the Ischemic Brain. Curr Drug Targets 2020; 20:953-959. [PMID: 30539695 PMCID: PMC6700753 DOI: 10.2174/1389450120666181211144550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 12/26/2022]
Abstract
The plasminogen activation (PA) system was originally thought to exclusively promote the degradation of fibrin by catalyzing the conversion of plasminogen into plasmin via two serine proteinases: tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA). However, experimental evidence accumulated over the last 30 years indicates that tPA and uPA are also found in the central nervous system (CNS), where they have a plethora of functions that not always require plasmin generation or fibrin degradation. For example, plasminogen-dependent and - independent effects of tPA and uPA play a central role in the pathophysiological events that underlie one of the leading causes of mortality and disability in the world: cerebral ischemia. Indeed, recent work indicates that while the rapid release of tPA from the presynaptic compartment following the onset of cerebral ischemia protects the synapse from the deleterious effects of the ischemic injury, the secretion of uPA and its binding to its receptor (uPAR) during the recovery phase promotes the repair of synapses that have been lost to the acute ischemic insult. This restorative role of uPA has high translational significance because to this date there is no effective approach to induce neurorepair in the ischemic brain. Here we will discuss recent evidence that bridges the gap between basic research in the field of the PA system and the bedside of ischemic stroke patients, indicating that uPA and uPAR are potential targets for the development of therapeutic strategies to promote neurological recovery among ischemic stroke survivors.
Collapse
Affiliation(s)
- Manuel Yepes
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center; Atlanta, GA, United States.,Department of Neurology & Center for Neurodegenerative Disease, Emory University School of Medicine; Atlanta, GA, United States.,Department of Neurology, Veterans Affairs Medical Center; Atlanta, GA, United States
| |
Collapse
|
20
|
Adorjan I, Tyler T, Bhaduri A, Demharter S, Finszter CK, Bako M, Sebok OM, Nowakowski TJ, Khodosevich K, Møllgård K, Kriegstein AR, Shi L, Hoerder‐Suabedissen A, Ansorge O, Molnár Z. Neuroserpin expression during human brain development and in adult brain revealed by immunohistochemistry and single cell RNA sequencing. J Anat 2019; 235:543-554. [PMID: 30644551 PMCID: PMC6704272 DOI: 10.1111/joa.12931] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2018] [Indexed: 01/08/2023] Open
Abstract
Neuroserpin is a serine-protease inhibitor mainly expressed in the CNS and involved in the inhibition of the proteolytic cascade. Animal models confirmed its neuroprotective role in perinatal hypoxia-ischaemia and adult stroke. Although neuroserpin may be a potential therapeutic target in the treatment of the aforementioned conditions, there is still no information in the literature on its distribution during human brain development. The present study provides a detailed description of the changing spatiotemporal patterns of neuroserpin focusing on physiological human brain development. Five stages were distinguished within our examined age range which spanned from the 7th gestational week until adulthood. In particular, subplate and deep cortical plate neurons were identified as the main sources of neuroserpin production between the 25th gestational week and the first postnatal month. Our immunohistochemical findings were substantiated by single cell RNA sequencing data showing specific neuronal and glial cell types expressing neuroserpin. The characterization of neuroserpin expression during physiological human brain development is essential for forthcoming studies which will explore its involvement in pathological conditions, such as perinatal hypoxia-ischaemia and adult stroke in human.
Collapse
Affiliation(s)
- Istvan Adorjan
- Department of Anatomy, Histology and EmbryologySemmelweis UniversityBudapestHungary
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
- Neuropathology UnitNuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Teadora Tyler
- Department of Anatomy, Histology and EmbryologySemmelweis UniversityBudapestHungary
| | - Aparna Bhaduri
- Department NeurologyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Samuel Demharter
- Biotech Research and Innovation CentreUniversity of CopenhagenCopenhagenDenmark
| | | | - Maria Bako
- Department of Anatomy, Histology and EmbryologySemmelweis UniversityBudapestHungary
| | - Oliver Marcell Sebok
- Department of Anatomy, Histology and EmbryologySemmelweis UniversityBudapestHungary
| | | | | | - Kjeld Møllgård
- Department of Cellular and Molecular MedicineThe Panum InstituteFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | | | - Lei Shi
- Joint Laboratory for Neuroscience and Innovative Drug ResearchJinan UniversityGuangzhouChina
| | | | - Olaf Ansorge
- Neuropathology UnitNuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Zoltán Molnár
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
21
|
Zhu J, Wan Y, Xu H, Wu Y, Hu B, Jin H. The role of endogenous tissue-type plasminogen activator in neuronal survival after ischemic stroke: friend or foe? Cell Mol Life Sci 2019; 76:1489-1506. [PMID: 30656378 PMCID: PMC11105644 DOI: 10.1007/s00018-019-03005-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/19/2018] [Accepted: 01/03/2019] [Indexed: 12/29/2022]
Abstract
Endogenous protease tissue-type plasminogen activator (tPA) has highly efficient fibrinolytic activity and its recombinant variants alteplase and tenecteplase are established as highly effective thrombolytic drugs for ischemic stroke. Endogenous tPA is constituted of five functional domains through which it interacts with a variety of substrates, binding proteins and receptors, thus having enzymatic and cytokine-like effects to act on all cell types of the brain. In the past 2 decades, numerous studies have explored the clinical relevance of endogenous tPA in neurological diseases, especially in ischemic stroke. tPA is released from many cells within the brain parenchyma exposed to ischemia conditions in vitro and in vivo, which is believed to control neuronal fate. Some studies proved that tPA could induce blood-brain barrier disruption, neural excitotoxicity and inflammation, while others indicated that tPA also has anti-excitotoxic, neurotrophic and anti-apoptotic effects on neurons. Therefore, more work is needed to elucidate how tPA mediates such opposing functions that may amplify tPA from a therapeutic means into a key therapeutic target in endogenous neuroprotection after stroke. In this review, we summarize the biological characteristics and pleiotropic functions of tPA in the brain. Then we focus on possible hypotheses about why and how endogenous tPA mediates ischemic neuronal death and survival. Finally, we analyze how endogenous tPA affects neuron fate in ischemic stroke in a comprehensive view.
Collapse
Affiliation(s)
- Jiayi Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yan Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Hexiang Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yulang Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
22
|
Effect of Combination Therapy with Neuroprotective and Vasoprotective Agents on Cerebral Ischemia. Can J Neurol Sci 2018; 45:325-331. [DOI: 10.1017/cjn.2018.8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AbstractBecause most tested drugs are active against only one of the damaging processes associated with stroke, other mechanisms may cause cellular death. Thus, a combination of protective agents targeting different pathophysiological mechanisms may obtain better effects than a single agent. The major objective of this study was to investigate the effect of combination therapy with vascular endothelial growth factor (VEGF) and nerve growth factor (NGF) after controlled ischemic brain injury in rabbits.Methods:Animals were randomly assigned to one of the following groups: sham group, saline-treated control group or NGF+VEGF-treated group. Animals received an intracerebral microinjection of VEGF and NGF or saline at 5 or 8 hours after ischemia. The two specified time points of administration were greater than or equal to the existing therapeutic time window for monoterapy with VEGF or NGF alone (3 or 5 hours of ischemia). Infarct volume, water content, neurological deficits, neural cell apoptosis and the expression of caspase-3 and Bcl-2 were measured.Results:Compared with saline-treated controls, the combination therapy of VEGF and NGF significantly reduced infarct volume, water content, neural cell apoptosis and the expression of caspase-3, up-regulated the expression of Bcl-2 and improved functional recovery (bothp<0.01) when administered 5 or 8 hours after ischemia. The earlier the administration the better the neuroprotection.Conclusions:These results showed that the combination therapy with VEGF and NGF provided neuroprotective effects. In addition, the time window of combination treatment should be at least 8 hours after ischemia, which was wider than monotherapy.RÉSUMÉ:Les effets d’une polythérapie combinant agents neuro-protecteurs et agents vasoprotecteurs dans les cas d’ischémie cérébrale.Contexte:Étant donné que la plupart des médicaments préalablement testés tendent à n’agir contre seulement un des processus de dommage associés aux AVC, il est possible que d’autres processus entraînent une mort cellulaire. À cet effet, il se pourrait qu’une combinaison d’agents protecteurs ciblant divers mécanismes physiopathologiques permette d’obtenir de meilleurs résultats qu’un simple agent. Après avoir suscité de façon contrôlée des lésions cérébrales ischémiques chez des lapins, l’objectif principal de la présente étude a donc été de se pencher sur l’impact d’une polythérapie combinant la protéine dite « facteur de croissance de l’endothélium vasculaire » (ou « VEGF » en anglais) avec le « facteur de croissance des nerfs » (ou « NGF » en anglais).Méthodes:Les animaux ont été attribués au hasard à l’un des groupes suivants : ceux ayant reçu un traitement fictif ; ceux, du groupe témoin, ayant bénéficié d’un traitement à base de solution saline ; et finalement ceux ayant été traités au moyen des VEGF et NGF. À noter que les lapins ont reçu une micro-injection intracérébrale de VEGF et de NGF ou de solution saline 5 heures ou 8 heures à la suite de leur AVC. Ces deux délais d’administration des VEGF et NGF sont équivalents ou supérieurs aux délais actuels d’administration des VEGF ou NGF à titre de monothérapie (3 heures ou 5 heures à la suite d’un AVC). Tant le volume des infarctus, le contenu en eau, les déficits neurologiques ainsi causés, l’apoptose des neurones que l’expression des protéases caspase 3 et des protéines Bcl-2 ont été mesurés.Résultats:Si on la compare au traitement à base de solution saline administré au groupe témoin, la polythérapie à base de VEGF et de NGF, lorsqu’administrée 5 heures ou 8 heures à la suite de l’AVC, a su réduire de façon notable le volume des infarctus, le contenu en eau, l’apoptose des neurones et l’expression des protéases caspase 3. Elle a également permis de réguler à la hausse l’expression des protéines Bcl-2 en plus d’entraîner une amélioration de la récupération fonctionnelle (p< 0,01 pour ces deux aspects). Ainsi donc, plus tôt l’on opte pour cette polythérapie, meilleure sera la neuroprotection encourue.Conclusions:Ces résultats démontrent que la polythérapie à base de VEGF et de NGF procure des effets neuroprotecteurs. Quant au délai d’administration de ce traitement combinatoire, il devrait être d’au moins 8 heures à la suite d’un AVC, ce qui est plus élevé que la monothérapie.
Collapse
|
23
|
Tissue Plasminogen Activator Neurotoxicity is Neutralized by Recombinant ADAMTS 13. Sci Rep 2016; 6:25971. [PMID: 27181025 PMCID: PMC4867598 DOI: 10.1038/srep25971] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 04/22/2016] [Indexed: 12/29/2022] Open
Abstract
Tissue plasminogen activator (tPA) is an effective treatment for ischemic stroke, but its neurotoxicity is a significant problem. Here we tested the hypothesis that recombinant ADAMTS 13 (rADAMTS 13) would reduce tPA neurotoxicity in a mouse model of stroke. We show that treatment with rADAMTS 13 in combination with tPA significantly reduced infarct volume compared with mice treated with tPA alone 48 hours after stroke. The combination treatment significantly improved neurological deficits compared with mice treated with tPA or vehicle alone. These neuroprotective effects were associated with significant reductions in fibrin deposits in ischemic vessels and less severe cell death in ischemic brain. The effect of rADAMTS13 on tPA neurotoxicity was mimicked by the N-methyl-D-aspartate (NMDA) receptor antagonist M-801, and was abolished by injection of NMDA. Moreover, rADAMTS 13 prevents the neurotoxicity effect of tPA, by blocking its interaction with the NMDA receptor NR2B and the attendant phosphorylation of NR2B and activation of ERK1/2. Finally, the NR2B-specific NMDA receptor antagonist ifenprodil abolished tPA neurotoxicity and rADAMTS 13 treatment had no further beneficial effect. Our data suggest that the combination of rADAMTS 13 and tPA may provide a novel treatment of ischemic stroke by diminishing the neurotoxic effects of exogenous tPA.
Collapse
|
24
|
Jandial R, Choy C, Levy ML. Plasmin in brain stroma inhibits metastatic colonization. Neurosurgery 2014; 75:N10-1. [PMID: 25033355 DOI: 10.1227/neu.0000000000000436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Affiliation(s)
- Rahul Jandial
- City of Hope, Duarte, California Rady Children's Hospital San Diego, San Diego, California
| | | | | |
Collapse
|
25
|
Lee TW, Montgomery JM, Birch NP. The serine protease inhibitor neuroserpin regulates the growth and maturation of hippocampal neurons through a non-inhibitory mechanism. J Neurochem 2012; 121:561-74. [PMID: 22191421 DOI: 10.1111/j.1471-4159.2011.07639.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuroserpin is a brain-specific serine protease inhibitor that is expressed in the developing and adult nervous system. Its expression profile led to suggestions that it played roles in neuronal growth and connectivity. In this study, we provide direct evidence to support a role for neuroserpin in axon and dendritic growth. We report that axon growth is enhanced while axon and dendrite diameter are reduced following neuroserpin treatment of hippocampal neurons. More complex effects are seen on dendritic growth and branching with neuroserpin-stimulating dendritic growth and branching in young neurons but switching to an inhibitory response in older neurons. The protease inhibitory activity of neuroserpin is not required to activate changes in neuronal morphology and a proportion of responses are modulated by an antagonist to the LRP1 receptor. Collectively, these findings support a key role for neuroserpin as a regulator of neuronal development through a non-inhibitory mechanism and suggest a basis for neuroserpin's effects on complex emotional behaviours and recent link to schizophrenia.
Collapse
Affiliation(s)
- Tet Woo Lee
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | | |
Collapse
|
26
|
Shichi K, Fujita-Hamabe W, Harada S, Mizoguchi H, Yamada K, Nabeshima T, Tokuyama S. Involvement of matrix metalloproteinase-mediated proteolysis of neural cell adhesion molecule in the development of cerebral ischemic neuronal damage. J Pharmacol Exp Ther 2011; 338:701-10. [PMID: 21602423 DOI: 10.1124/jpet.110.178079] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Neural cell adhesion molecule (NCAM) is a membrane protein abundantly expressed in the central nervous system. Recently, it has been reported that dysfunction of NCAM is linked to human brain disorders. Furthermore, NCAM is one of the proteolysis targets of matrix metalloproteinase (MMP), whose activation is implicated in neuronal damage. The aim of this study was to elucidate the involvement of MMP-mediated proteolysis of NCAM in the development of ischemic neuronal damage. Male ddY and MMP-9 knockout (KO) C57BL/6J mice were subjected to 2 h of middle cerebral artery occlusion (MCAO). In MCAO model mice, development of infarction and behavioral abnormality were clearly observed on days 1 and 3 after MCAO. Protein levels of MMP-2 and MMP-9 were significantly increased on days 1 and 3 after MCAO. In addition, full-length NCAM (180 kDa) was significantly decreased, but its metabolite levels increased on day 1 by ischemic stress per se. NCAM small interfering RNA significantly increased the neuronal damage induced by MCAO. MMP inhibition or MMP-9 gene KO attenuated the infarction, behavioral abnormalities, and decrease of NCAM (180 kDa) observed after MCAO in mice. The present findings clearly suggest that MMP-2/MMP-9-mediated NCAM proteolysis is implicated in the exacerbation of ischemic neuronal damage.
Collapse
Affiliation(s)
- Kanako Shichi
- Department of Clinical Pharmacy, Kobe Gakuin University School of Pharmaceutical Sciences, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Su EJ, Geyer M, Wahl M, Mann K, Ginsburg D, Brohmann H, Petersen KU, Lawrence DA. The thrombomodulin analog Solulin promotes reperfusion and reduces infarct volume in a thrombotic stroke model. J Thromb Haemost 2011; 9:1174-82. [PMID: 21645225 PMCID: PMC3111949 DOI: 10.1111/j.1538-7836.2011.04269.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Accepted: 02/24/2011] [Indexed: 01/02/2023]
Abstract
BACKGROUND Currently there is no approved anticoagulant for treating acute stroke. This is largely because of concern for hemorrhagic complications, and suggests a critical need for safer anticoagulants. Solulin is a soluble analog of the endothelial cell receptor thrombomodulin, able to bind free thrombin and convert it to an activator of the anticoagulant, protein C. OBJECTIVE Solulin was tested for its ability to inhibit middle cerebral artery occlusion (MCAO) induced by photothrombosis, and to restore MCA patency after establishment of stable occlusion. METHODS Cerebral blood flow (CBF) was monitored by laser Doppler for 1.5 h after occlusion and again 72 h later. RESULTS Solulin treatment 30 min before thrombosis resulted in an approximately 50% increase in time to form a stable occlusion. When administered 30 or 60 min after MCAO, Solulin significantly improved CBF within 90 min of treatment. In contrast, none of the vehicle-treated mice showed restoration of CBF in the first 90 min and only 17% did so by 72 h. Solulin treatment was associated with a significant reduction in infarct volume, and was well tolerated with no overt hemorrhage observed in any treatment group. Mechanistic studies in mice homozygous for the factor (F)V Leiden mutation, suggest that Solulin's efficacy derives primarily from the anticoagulant activity of the thrombin-Solulin complex and not from direct anti-inflammatory or neuroprotective effects of Solulin or activated protein C. CONCLUSIONS Our data indicate that Solulin is a safe and effective anticoagulant that is able to antagonize active thrombosis in acute ischemic stroke, and to reduce infarct volume.
Collapse
Affiliation(s)
- E J Su
- Department of Internal Medicine Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI 48109-5644, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Weiss TW, Samson AL, Niego B, Daniel PB, Medcalf RL. Oncostatin M is a neuroprotective cytokine that inhibits excitotoxic injury in vitro and in vivo. FASEB J 2006; 20:2369-71. [PMID: 17023520 DOI: 10.1096/fj.06-5850fje] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Oncostatin M (OsM) is a member of the interleukin (IL)-6 family of cytokines and is well known for its role in inflammation, cell proliferation, and hematopoiesis. OsM, together with its glycoprotein 130 containing receptor complex, is expressed and regulated in most cells of the central nervous system (CNS), yet the function of OsM within this compartment is poorly understood. Here we have investigated the effect of OsM using in vitro and in vivo models of excitotoxic injury. Using primary cultures of mouse cortical neurons, OsM was shown to reduce N-methyl-D-aspartate (NMDA) -induced neuronal death by 50% when added simultaneously with NMDA while pretreatment of neurons with OsM fully prevented NMDA toxicity indicating a profound protective effect of this cytokine. OsM was also shown to inhibit NMDA-mediated increase in levels of free intracellular calcium and to selectively reduce neuronal expression of the NR2C subunit of the NMDA receptor. Finally, using an in vivo model of excitotoxic injury, OsM significantly reduced the NMDA-induced lesion volume when coinjected with NMDA into the mouse striatum. Taken together, these results identify OsM as a powerful neuroprotective cytokine and provide a rational foundation to explore the therapeutic potential for OsM in diseases of the CNS.
Collapse
Affiliation(s)
- Thomas W Weiss
- Australian Centre for Blood Diseases, Monash University, 89 Commercial Rd., Prahran 3181, Victoria, Australia
| | | | | | | | | |
Collapse
|
29
|
Lotem J, Sachs L. Epigenetics and the plasticity of differentiation in normal and cancer stem cells. Oncogene 2006; 25:7663-72. [PMID: 16847453 DOI: 10.1038/sj.onc.1209816] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Embryonic stem cells are characterized by their differentiation to all cell types during embryogenesis. In adult life, different tissues also have somatic stem cells, called adult stem cells, which in specific niches can undergo multipotent differentiation. The use of these adult stem cells has considerable therapeutic potential for the regeneration of damaged tissues. In both embryonic and adult stem cells, differentiation is controlled by epigenetic mechanisms, and the plasticity of differentiation in these cells is associated with transcription accessibility for genes expressed in different normal tissues. Abnormalities in genetic and/or epigenetic controls can lead to development of cancer, which is maintained by self-renewing cancer stem cells. Although the genetic abnormalities produce defects in growth and differentiation in cancer stem cells, these cells have not always lost the ability to undergo differentiation through epigenetic changes that by-pass the genomic abnormalities, thus creating the basis for differentiation therapy. Like normal stem cells, cancer stem cells can show plasticity for differentiation. This plasticity of cancer stem cells is also associated with transcription accessibility for genes that are normally expressed in different tissues, including tissues other than those from which the cancers originated. This broad transcription accessibility can also contribute to the behavior of cancer cells by overexpressing genes that promote cell viability, growth and metastasis.
Collapse
Affiliation(s)
- J Lotem
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
30
|
Sheehan JJ, Tsirka SE. Fibrin-modifying serine proteases thrombin, tPA, and plasmin in ischemic stroke: a review. Glia 2005; 50:340-350. [PMID: 15846799 DOI: 10.1002/glia.20150] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ischemic stroke is a sudden loss of circulation to a portion of the brain that results in a loss of neurologic function. Many ischemic strokes are embolic. They result from a thrombus traveling into the central circulation and occluding a blood vessel. Treatment of ischemic stroke with recombinant tissue plasminogen activator (tPA) can improve patient outcomes. However, tPA must be used during a specific time window after the stroke onset to be effective and it risks converting an ischemic stroke into a hemorrhagic one. We explore the basic effects of fibrin-modifying proteases on neurons, astrocytes, and microglia during ischemia. tPA, thrombin, and plasmin can initiate microglial activation and change both neuronal and astrocytic survival. As a result of these functions and of their role in blood homeostasis, all three of these proteases have profound effects on neurons and glial cells in the brain and are capable of altering the development and severity of ischemic stroke.
Collapse
Affiliation(s)
- John J Sheehan
- Program in Molecular and Cellular Pharmacology and Department of Pharmacological Sciences, University Medical Center at Stony Brook, Stony Brook, New York
| | - Stella E Tsirka
- Program in Molecular and Cellular Pharmacology and Department of Pharmacological Sciences, University Medical Center at Stony Brook, Stony Brook, New York
| |
Collapse
|
31
|
Cinelli P, Madani R, Tsuzuki N, Vallet P, Arras M, Zhao CN, Osterwalder T, Rülicke T, Sonderegger P. Neuroserpin, a neuroprotective factor in focal ischemic stroke. Mol Cell Neurosci 2001; 18:443-57. [PMID: 11922137 DOI: 10.1006/mcne.2001.1028] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Because recent studies have indicated that tissue plasminogen activator (tPA) aggravates neurodegenerative processes in many neural pathologies, we studied whether the endogenous tPA antagonist neuroserpin has a neuroprotective effect in an animal model of focal ischemic stroke. After induction of a focal ischemic stroke in the mouse by occlusion of the middle cerebral artery, we found that microglial cells accumulated in the marginal zone of the infarct are the most important source for both plasminogen activators, tPA and uPA. To investigate the effect of neuroserpin on the size and the histology of the infarct we produced transgenic mice overexpressing neuroserpin approximately sixfold in the nervous system. In the brain of these mice the total tPA activity in the uninjured tissue was strongly reduced. After induction of a focal ischemic stroke in the transgenic mice by a permanent occlusion of the middle cerebral artery (MCA), the infarcts were 30% smaller than in the wild-type mice. Immunohistochemical analyses and in situ hybridization revealed an attenuation of the microglial activation in the reactive zone. Concomitantly, the microglial production of tPA and uPA, as well as the PA-activity in the infarct region was markedly reduced. Thus, our results indicate that neuroserpin reduces microglial activation and, therefore, the PA activity and has a neuroprotective role after focal ischemic stroke.
Collapse
Affiliation(s)
- P Cinelli
- Institute of Biochemistry, University of Zurich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hill RM, Brennan SO, Birch NP. Expression, purification, and functional characterization of the serine protease inhibitor neuroserpin expressed in Drosophila S2 cells. Protein Expr Purif 2001; 22:406-13. [PMID: 11483002 DOI: 10.1006/prep.2001.1463] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuroserpin (NS) is a serine protease inhibitor (or serpin) that is widely expressed in the developing and adult nervous systems. It has been implicated in the regulation of proteases involved in processes such as synaptic plasticity, neuronal migration, and axogenesis. To aid in the characterization of this new serpin we have established a high-level expression system in Drosophila S2 cells and developed a purification strategy to obtain neuroserpin for functional studies. Suspension cultures of S2-NS cells secreted recombinant neuroserpin into the medium. High-level expression was maintained when the cells were switched to a nonselection serum-free medium for 3-4 days to facilitate protein purification. Recombinant neuroserpin was purified by sequential chromatography on Macroprep ceramic hydroxyapatite, Type I, POROS HQ20, Resource Q, and Superdex 75 HR 10/30 media. Two secreted forms of neuroserpin were observed with molecular weights of approximately 49 and approximately 50 kDa which may represent alternative glycosylation at three putative N-linked glycosylation sites. Amino acid sequence analysis indicated three NH(2)-terminal sequences. The major sequence was generated by cleavage at the Gly(18)-Ala(19) bond consistent with removal of an 18-amino-acid signal peptide. Two further sequences were identified each with one fewer amino acids at the NH(2)-terminus. All three NH(2)-terminal sequences were also identified by mass spectrometric analysis of neuroserpin following trypsin digestion. Mass spectrometry also confirmed the protein had an intact carboxyl terminus while complex formation assays indicated the inhibitor was functionally active. In summary, Drosophila S2 cells offered a nonlytic stable expression system for the continual production of neuroserpin in high-density suspension cultures.
Collapse
Affiliation(s)
- R M Hill
- Molecular Neuroendocrinology Laboratory, School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | |
Collapse
|