1
|
Dolan F, Wintermark P. Updates in Treatment of Hypoxic-Ischemic Encephalopathy. Clin Perinatol 2025; 52:321-343. [PMID: 40350214 DOI: 10.1016/j.clp.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Worldwide, hypoxic-ischemic encephalopathy (HIE) remains one of the leading causes of nervous system disabilities. Therapeutic hypothermia is the current standard-of-care treatment in high-income countries, because it was shown to reduce associated mortality and morbidities. However, up to 29% of treated neonates still experience adverse neurodevelopmental outcomes, and its efficacy in low- and middle-income countries remains debated, highlighting the need for adjunct or alternative therapies. Treatments for HIE targeting neuroprotection and/or neurorestoration are under investigation. Meanwhile, attentive daily management of multiorgan failure during the first days of life continues to be essential to limit further brain injury.
Collapse
Affiliation(s)
- Florence Dolan
- Division of Newborn Medicine, Department of Pediatrics, Montreal Children's Hospital, Research Institute of the McGill University Health Center, McGill University, 1001 boul. Décarie, Site Glen Block E, EM0.3244, Montréal, Quebec H4A 3J1, Canada
| | - Pia Wintermark
- Division of Newborn Medicine, Department of Pediatrics, Montreal Children's Hospital, Research Institute of the McGill University Health Center, McGill University, 1001 boul. Décarie, Site Glen Block E, EM0.3244, Montréal, Quebec H4A 3J1, Canada.
| |
Collapse
|
2
|
Constantin MF, de la Villarmois EA, Bravo JL, Marcotti A, Ghersi M, Castro F, Calfa GD, Rubianes MD, Pérez MF. Sildenafil promotes dual memory effects in young rats: involvement of dopamine reuptake. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06806-x. [PMID: 40383851 DOI: 10.1007/s00213-025-06806-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 05/03/2025] [Indexed: 05/20/2025]
Abstract
RATIONALE Sildenafil, a phosphodiesterase-5 inhibitor, crosses the blood-brain barrier, enhancing cGMP signaling, dopamine neurotransmission, and hippocampal plasticity-key mechanisms for learning and memory. OBJECTIVES This study aimed to (1) determine whether sildenafil influences hippocampal dopamine levels by modulating dopamine transporter (DAT) function in naïve young rats; (2) assess sildenafil-induced dopamine increases by evaluating its impact on hippocampal-dependent memories in non-aversive and aversive tasks; and (3) examine the effects of acute sildenafil administration on hippocampal synaptic plasticity. METHODS DAT function was assessed through ex-vivo dopamine reuptake analysis in the hippocampus and nucleus accumbens of rats sacrificed 2 h post-administration. Memory effects were evaluated by administering sildenafil 2 h before training in non-aversive (novel object recognition-NOR, Y-maze, Barnes maze) and aversive (step-down inhibitory avoidance, fear conditioning) tasks. To examine D3 receptor (D3R) involvement, a subset of animals received the selective D3R antagonist FAUC-365 before NOR training. Synaptic plasticity was analyzed via electrophysiology and dendritic spine density. RESULTS Sildenafil reduced dopamine reuptake, likely by inhibiting DAT. It impaired NOR performance, an effect prevented by D3R antagonism, while leaving working and long-term spatial memory unaffected. Additionally, sildenafil enhanced aversive memory expression, facilitated hippocampal long-term potentiation, and increased dendritic spine density. CONCLUSIONS Sildenafil differentially affected hippocampal-dependent memory, potentially by increasing dopamine transmission. In young, healthy individuals, sildenafil may impair recognition memory and alter responses to non-threatening stimuli, influencing cognitive and emotional processes.
Collapse
Affiliation(s)
- Maria Florencia Constantin
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, IFEC-CONICET, Universidad Nacional de Córdoba. Haya de La Torre y Medina Allende S/N, Ciudad Universitaria, 5000, Córdoba, CP, Argentina
| | - Emilce Artur de la Villarmois
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, IFEC-CONICET, Universidad Nacional de Córdoba. Haya de La Torre y Medina Allende S/N, Ciudad Universitaria, 5000, Córdoba, CP, Argentina
| | - José Leonardo Bravo
- Departamento de Fisicoquímica, Facultad de Ciencias Químicas, INFIQC-CONICET, Universidad Nacional de Córdoba. Haya de La Torre y Medina Allende S/N, Ciudad Universitaria, Córdoba, 5000, CP, Argentina
| | - Aida Marcotti
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, IFEC-CONICET, Universidad Nacional de Córdoba. Haya de La Torre y Medina Allende S/N, Ciudad Universitaria, 5000, Córdoba, CP, Argentina
| | - Marisa Ghersi
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, IFEC-CONICET, Universidad Nacional de Córdoba. Haya de La Torre y Medina Allende S/N, Ciudad Universitaria, 5000, Córdoba, CP, Argentina
| | - Facundo Castro
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, IFEC-CONICET, Universidad Nacional de Córdoba. Haya de La Torre y Medina Allende S/N, Ciudad Universitaria, 5000, Córdoba, CP, Argentina
| | - Gastón Diego Calfa
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, IFEC-CONICET, Universidad Nacional de Córdoba. Haya de La Torre y Medina Allende S/N, Ciudad Universitaria, 5000, Córdoba, CP, Argentina
| | - María Dolores Rubianes
- Departamento de Fisicoquímica, Facultad de Ciencias Químicas, INFIQC-CONICET, Universidad Nacional de Córdoba. Haya de La Torre y Medina Allende S/N, Ciudad Universitaria, Córdoba, 5000, CP, Argentina
| | - Mariela Fernanda Pérez
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, IFEC-CONICET, Universidad Nacional de Córdoba. Haya de La Torre y Medina Allende S/N, Ciudad Universitaria, 5000, Córdoba, CP, Argentina.
| |
Collapse
|
3
|
Altahrawi AY, James AW, Shah ZA. The Role of Oxidative Stress and Inflammation in the Pathogenesis and Treatment of Vascular Dementia. Cells 2025; 14:609. [PMID: 40277934 PMCID: PMC12026122 DOI: 10.3390/cells14080609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Vascular dementia (VaD) is a heterogeneous group of brain disorders caused by cerebrovascular pathologies and the second most common cause of dementia, accounting for over 20% of cases and posing an important global health concern. VaD can be caused by cerebral infarction or injury in critical brain regions, including the speech area of the dominant hemisphere or arcuate fasciculus of the dominant hemisphere, leading to notable cognitive impairment. Although the exact causes of dementia remain multifactorial and complex, oxidative stress (reactive oxygen species), neuroinflammation (TNFα, IL-6, and IL-1β), and inflammasomes are considered central mechanisms in its pathology. These conditions contribute to neuronal damage, synaptic dysfunction, and cognitive decline. Thus, antioxidants and anti-inflammatory agents have emerged as potential therapeutic targets in dementia. Recent studies emphasize that cerebrovascular disease plays a dual role: first, as a primary cause of cognitive impairment and then as a contributor to the manifestation of dementia driven by other factors, such as Alzheimer's disease and other neurodegenerative conditions. This comprehensive review of VaD focuses on molecular mechanisms and their consequences. We provided up-to-date knowledge about epidemiology, pathophysiological mechanisms, and current therapeutic approaches for VaD.
Collapse
Affiliation(s)
- Aseel Y. Altahrawi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Antonisamy William James
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA;
| | - Zahoor A. Shah
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA;
| |
Collapse
|
4
|
Singh NK, Singh P, Varshney P, Singh A, Bhushan B. Multimodal action of phosphodiesterase 5 inhibitors against neurodegenerative disorders: An update review. J Biochem Mol Toxicol 2024; 38:e70021. [PMID: 39425458 DOI: 10.1002/jbt.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/03/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024]
Abstract
Phosphodiesterase type 5 (PDE5) is an enzyme primarily found in the smooth muscle of the corpus cavernosum and also highly expressed in the substantia nigra, cerebellum, caudate, hippocampal regions and cerebellar purkinje cells, responsible for selectively breaking down cyclic guanosine monophosphate (cGMP) into 5'-GMP and regulate intracellular cGMP levels. As a second messenger, cyclic GMP enhances signals at postsynaptic receptors and triggers downstream effector molecules, leading to changes in gene expression and neuronal responses. Additionally, cGMP signaling transduction cascade, present in the brain, is also essential for learning and memory processes. Mechanistically, PDE5 inhibitors share structural similarities with cGMP, competitively binding to PDE5 and inhibiting cGMP hydrolysis. This action enhances the effects of nitric oxide, resulting in anti-inflammatory and neuroprotective effects. Neurodegenerative disorders entail the progressive loss of neuron structure, culminating in neuronal cell death, with currently available drugs providing only limited symptomatic relief, rendering neurodegeneration considered incurable. PDE5 inhibitors have recently emerged as a potential therapeutic approach for neurodegeneration, neuroinflammation, and diseases involving cognitive impairment. This review elucidates the principal roles of 3',5'-cyclic adenosine monophosphate (cAMP) and cGMP signaling pathways in neuronal functions, believed to play pivotal roles in the pathogenesis of various neurodegenerative disorders. It provides an updated assessment of PDE5 inhibitors as disease-modifying agents for conditions such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, cerebral ischemia, Huntington's disease, and neuroinflammation. The paper aims to review the current understanding of PDE5 inhibitors, which concurrently regulate both cAMP and cGMP signaling pathways, positing that they may exert complementary and synergistic effects in modifying neurodegeneration, thus presenting a novel direction in therapeutic discovery. Moreover, the review provides critical about biological functions, therapeutic potentials, limitations, challenges, and emerging applications of selective PDE5 inhibitors. This comprehensive overview aims to guide future academic and industrial endeavors in this field.
Collapse
Affiliation(s)
- Niraj Kumar Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| | - Pranjul Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| | - Prachi Varshney
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| | - Ashini Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| | - Bharat Bhushan
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Chaumuhan, Mathura, India
| |
Collapse
|
5
|
Yu YH, Kim GW, Lee YR, Park DK, Song B, Kim DS. Effects of Sildenafil on Cognitive Function Recovery and Neuronal Cell Death Protection after Transient Global Cerebral Ischemia in Gerbils. Biomedicines 2024; 12:2077. [PMID: 39335590 PMCID: PMC11429064 DOI: 10.3390/biomedicines12092077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Cerebral ischemic stroke is a major cause of death worldwide due to brain cell death resulting from ischemia-reperfusion injury. However, effective treatment approaches for patients with ischemic stroke are still lacking in clinical practice. This study investigated the potential neuroprotective effects of sildenafil, a phosphodiesterase-5 inhibitor, in a gerbil model of global brain ischemia. We investigated the effects of sildenafil on the expression of glial fibrillary acidic protein and aquaporin-4, which are markers related to astrocyte activation and water homeostasis, respectively. Immunofluorescence analysis showed that the number of cells co-expressing these markers, which was elevated in the ischemia-induced group, was significantly reduced in the sildenafil-treated groups. This suggests that sildenafil may have a potential mitigating effect on astrocyte activation induced by ischemia. Additionally, we performed various behavioral tests, including the open-field test, novel object recognition, Barnes maze, Y-maze, and passive avoidance tests, to evaluate sildenafil's effect on cognitive function impaired by ischemia. Overall, the results suggest that sildenafil may serve as a neuroprotective agent, potentially alleviating delayed neuronal cell death and improving cognitive function impaired by ischemia.
Collapse
Affiliation(s)
- Yeon Hee Yu
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Gun Woo Kim
- Research Supporting Center for Medical Science, College of Medicine, Dong-A, Busan 49201, Republic of Korea
| | - Yu Ran Lee
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Dae-Kyoon Park
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Beomjong Song
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| |
Collapse
|
6
|
Wintermark P, Lapointe A, Steinhorn R, Rampakakis E, Burhenne J, Meid AD, Bajraktari-Sylejmani G, Khairy M, Altit G, Adamo MT, Poccia A, Gilbert G, Saint-Martin C, Toffoli D, Vachon J, Hailu E, Colin P, Haefeli WE. Feasibility and Safety of Sildenafil to Repair Brain Injury Secondary to Birth Asphyxia (SANE-01): A Randomized, Double-blind, Placebo-controlled Phase Ib Clinical Trial. J Pediatr 2024; 266:113879. [PMID: 38142044 DOI: 10.1016/j.jpeds.2023.113879] [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: 10/05/2023] [Revised: 11/21/2023] [Accepted: 12/17/2023] [Indexed: 12/25/2023]
Abstract
OBJECTIVE To test feasibility and safety of administering sildenafil in neonates with neonatal encephalopathy (NE), developing brain injury despite therapeutic hypothermia (TH). STUDY DESIGN We performed a randomized, double-blind, placebo-controlled phase Ib clinical trial between 2016 and 2019 in neonates with moderate or severe NE, displaying brain injury on day-2 magnetic resonance imaging (MRI) despite TH. Neonates were randomized (2:1) to 7-day sildenafil or placebo (2 mg/kg/dose enterally every 12 hours, 14 doses). Outcomes included feasibility and safety (primary outcomes), pharmacokinetics (secondary), and day-30 neuroimaging and 18-month neurodevelopment assessments (exploratory). RESULTS Of the 24 enrolled neonates, 8 were randomized to sildenafil and 3 to placebo. A mild decrease in blood pressure was reported in 2 of the 8 neonates after initial dose, but not with subsequent doses. Sildenafil plasma steady-state concentration was rapidly reached, but decreased after TH discontinuation. Twelve percent of neonates (1/8) neonates died in the sildenafil group and 0% (0/3) in the placebo group. Among surviving neonates, partial recovery of injury, fewer cystic lesions, and less brain volume loss on day-30 magnetic resonance imaging were noted in 71% (5/7) of the sildenafil group and in 0% (0/3) of the placebo group. The rate of death or survival to 18 months with severe neurodevelopmental impairment was 57% (4/7) in the sildenafil group and 100% (3/3) in the placebo group. CONCLUSIONS Sildenafil was safe and well-absorbed in neonates with NE treated with TH. Optimal dosing needs to be established. Evaluation of a larger number of neonates through subsequent phases II and III trials is required to establish efficacy. CLINICAL TRIAL REGISTRATION ClinicalTrials.govNCT02812433.
Collapse
Affiliation(s)
- Pia Wintermark
- Division of Newborn Medicine, Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Research Institute of the McGill University Health Center, McGill University, Montreal, Quebec, Canada.
| | - Anie Lapointe
- Department of Neonatology, Sainte-Justine Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Robin Steinhorn
- Department of Pediatrics, University of California San Diego, and Rady Children's Hospital, San Diego, CA
| | | | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas D Meid
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Gzona Bajraktari-Sylejmani
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - May Khairy
- Department of Pediatrics, McGill University, Montreal, Québec, Canada
| | - Gabriel Altit
- Division of Newborn Medicine, Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Research Institute of the McGill University Health Center, McGill University, Montreal, Quebec, Canada
| | - Marie-Therese Adamo
- Pharmacy Department, McGill University Health Center, Montreal, Québec, Canada
| | - Alishia Poccia
- Research Institute of the McGill University Health Center, McGill University, Montreal, Quebec, Canada
| | - Guillaume Gilbert
- MR Clinical Science, Philips Healthcare, Mississauga, Ontario, Canada
| | | | - Daniela Toffoli
- Department of Ophthalmology, McGill University, Montreal, Québec, Canada
| | - Julie Vachon
- Member of the Ordre des Psychologues du Quebec, Montreal, Québec, Canada
| | - Elizabeth Hailu
- Division of Newborn Medicine, Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Patrick Colin
- Patrick Colin Consultant Inc, Montreal, Québec, Canada
| | - Walter E Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
7
|
Julien P, Zinni M, Bonnel N, El Kamouh M, Odorcyk F, Peters L, Gautier EF, Leduc M, Broussard C, Baud O. Synergistic effect of sildenafil combined with controlled hypothermia to alleviate microglial activation after neonatal hypoxia-ischemia in rats. J Neuroinflammation 2024; 21:31. [PMID: 38263116 PMCID: PMC10804557 DOI: 10.1186/s12974-024-03022-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/12/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND AND PURPOSE The only validated treatment to prevent brain damage associated with hypoxia-ischemia (HI) encephalopathy of the newborn is controlled hypothermia with limited benefits. Additional putative neuroprotective drug candidates include sildenafil citrate, a phosphodiesterase-type 5 inhibitor. The main objective of this preclinical study is to assess its ability to reduce HI-induced neuroinflammation, in particular through its potential effect on microglial activation. METHODS HI was induced in P10 Sprague-Dawley rats by unilateral carotid permanent artery occlusion and hypoxia (HI) and treated by either hypothermia (HT) alone, Sildenafil (Sild) alone or combined treatment (SildHT). Lesion size and glial activation were analyzed by immunohistochemistry, qRT-PCR, and proteomic analyses performed at P13. RESULTS None of the treatments was associated with a significant early reduction in lesion size 72h after HI, despite significant changes in tissue loss distribution. Significant reductions in both Iba1 + (within the ipsilateral hemisphere) and GFAP + cells (within the ipsilateral hippocampus) were observed in SildHT group, but not in the other treatment groups. In microglia-sorted cells, pro-inflammatory markers, i.e. Il1b, Il6, Nos2, and CD86 were significantly downregulated in SildHT treatment group only. These changes were restricted to the ipsilateral hemisphere, were not evidenced in sorted astrocytes, and were not sex dependent. Proteomic analyses in sorted microglia refined the pro-inflammatory effect of HI and confirmed a biologically relevant impact of SildHT on specific molecular pathways including genes related to neutrophilic functions. CONCLUSIONS Our findings suggest that Sildenafil combined with controlled hypothermia produces maximum effect in mitigating microglial activation induced by HI through complex proteomic regulation. The reduction of neuroinflammation induced by Sildenafil may represent an interesting therapeutic strategy for neonatal neuroprotection.
Collapse
Affiliation(s)
- Pansiot Julien
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France
| | - Manuela Zinni
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France
| | - Natacha Bonnel
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France
| | - Marina El Kamouh
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France
| | - Felipe Odorcyk
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France
| | - Lea Peters
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France
| | - Emilie-Fleur Gautier
- Institut Cochin, Proteom'IC Facility, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Marjorie Leduc
- Institut Cochin, Proteom'IC Facility, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Cédric Broussard
- Institut Cochin, Proteom'IC Facility, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Olivier Baud
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France.
- Laboratory of Child Growth and Development, University of Geneva, Geneva, Switzerland.
- Division of Neonatology and Pediatric Intensive Care, Département de Pédiatrie, Hôpitaux Universitaires de Genève, Laboratoire de Développement et Croissance, Children's University Hospital of Geneva, Geneva, Switzerland.
| |
Collapse
|
8
|
You Q, Lan XB, Liu N, Du J, Ma L, Yang JM, Niu JG, Peng XD, Jin GL, Yu JQ. Neuroprotective strategies for neonatal hypoxic-ischemic brain damage: Current status and challenges. Eur J Pharmacol 2023; 957:176003. [PMID: 37640219 DOI: 10.1016/j.ejphar.2023.176003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) is a prominent contributor to both immediate mortality and long-term impairment in newborns. The elusive nature of the underlying mechanisms responsible for neonatal HIBD presents a significant obstacle in the effective clinical application of numerous pharmaceutical interventions. This comprehensive review aims to concentrate on the potential neuroprotective agents that have demonstrated efficacy in addressing various pathogenic factors associated with neonatal HIBD, encompassing oxidative stress, calcium overload, mitochondrial dysfunction, endoplasmic reticulum stress, inflammatory response, and apoptosis. In this review, we conducted an analysis of the precise molecular pathways by which these drugs elicit neuroprotective effects in animal models of neonatal hypoxic-ischemic brain injury (HIBD). Our objective was to provide a comprehensive overview of potential neuroprotective agents for the treatment of neonatal HIBD in animal experiments, with the ultimate goal of enhancing the feasibility of clinical translation and establishing a solid theoretical foundation for the clinical management of neonatal HIBD.
Collapse
Affiliation(s)
- Qing You
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Xiao-Bing Lan
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Ning Liu
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; Ningxia Special Traditional Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Juan Du
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Lin Ma
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Jia-Mei Yang
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Jian-Guo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, 750004, China.
| | - Xiao-Dong Peng
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Gui-Lin Jin
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fuzhou, 350108, Fujian, China; Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, 350108, Fujian, China.
| | - Jian-Qiang Yu
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; Ningxia Special Traditional Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| |
Collapse
|
9
|
Sela TC, Zahavi A, Friedman-Gohas M, Weiss S, Sternfeld A, Ilguisonis A, Badash D, Geffen N, Ofri R, BarKana Y, Goldenberg-Cohen N. Azithromycin and Sildenafil May Have Protective Effects on Retinal Ganglion Cells via Different Pathways: Study in a Rodent Microbead Model. Pharmaceuticals (Basel) 2023; 16:ph16040486. [PMID: 37111243 PMCID: PMC10142588 DOI: 10.3390/ph16040486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/12/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Decreased blood flow to the optic nerve (ON) and neuroinflammation are suggested to play an important role in the pathophysiology of glaucoma. This study investigated the potential neuroprotective effect of azithromycin, an anti-inflammatory macrolide, and sildenafil, a selective phosphodiesterase-5 inhibitor, on retinal ganglion cell survival in a glaucoma model, which was induced by microbead injection into the right anterior chamber of 50 wild-type (WT) and 30 transgenic toll-like receptor 4 knockout (TLR4KO) mice. Treatment groups included intraperitoneal azithromycin 0.1 mL (1 mg/0.1 mL), intravitreal sildenafil 3 µL, or intraperitoneal sildenafil 0.1 mL (0.24 μg/3 µL). Left eyes served as controls. Microbead injection increased intraocular pressure (IOP), which peaked on day 7 in all groups and on day 14 in azithromycin-treated mice. Furthermore, the retinas and ON of microbead-injected eyes showed a trend of increased expression of inflammatory- and apoptosis-related genes, mainly in WT and to a lesser extent in TLR4KO mice. Azithromycin reduced the BAX/BCL2 ratio, TGFβ, and TNFα levels in the ON and CD45 expression in WT retina. Sildenafil activated TNFα-mediated pathways. Both azithromycin and sildenafil exerted a neuroprotective effect in WT and TLR4KO mice with microbead-induced glaucoma, albeit via different pathways, without affecting IOP. The relatively low apoptotic effect observed in microbead-injected TLR4KO mice suggests a role of inflammation in glaucomatous damage.
Collapse
Affiliation(s)
- Tal Corina Sela
- Clalit Health Services, Tel Aviv 6209804, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Alon Zahavi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Ophthalmology, Rabin Medical Center-Beilinson Hospital, Petach Tikva 4941492, Israel
- Laboratory of Eye Research, Felsenstein Medical Research Center, Petach Tikva 4941492, Israel
| | - Moran Friedman-Gohas
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Krieger Eye Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Shirel Weiss
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Krieger Eye Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Amir Sternfeld
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Ophthalmology, Rabin Medical Center-Beilinson Hospital, Petach Tikva 4941492, Israel
- Krieger Eye Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Astrid Ilguisonis
- Krieger Eye Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Danielle Badash
- Krieger Eye Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Noa Geffen
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Ophthalmology, Rabin Medical Center-Beilinson Hospital, Petach Tikva 4941492, Israel
| | - Ron Ofri
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Yaniv BarKana
- The Glaucoma Innovations and Research Laboratory, The Sam Rothberg Glaucoma Center, Sheba Medical Center, Tel Hashomer 5262000, Israel
| | - Nitza Goldenberg-Cohen
- Krieger Eye Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
- Department of Ophthalmology, Bnai Zion Medical Center, Haifa 3339419, Israel
- Bruce and Ruth Faculty of Medicine, Technion, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
10
|
Sheweita SA, Alian DME, Haroun M, Nounou MI, Patel A, El-Khordagui L. Chitosan Nanoparticles Alleviated the Adverse Effects of Sildenafil on the Oxidative Stress Markers and Antioxidant Enzyme Activities in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:9944985. [PMID: 36891377 PMCID: PMC9988388 DOI: 10.1155/2023/9944985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/29/2022] [Accepted: 11/25/2022] [Indexed: 02/01/2023]
Abstract
Sildenafil (SF) is widely used for erectile dysfunction and other conditions, though with limitations regarding oral absorption and adverse effects. Despite nanotechnological improvements, the effect of nanocarriers on SF hepatotoxicity has not been documented to date. This study aimed at assessing the impact of chitosan nanoparticles either uncoated (CS NPs) or Tween 80-coated (T-CS NPs) on the effects of SF on oxidative stress markers and antioxidant enzyme activities in rats. Test SF-CS NPs prepared by ionic gelation were uniform positively charged nanospheres (diameter 178-215 nm). SF was administered intraperitoneally to male rats (1.5 mg/kg body weight) in free or nanoencapsulated forms as SF-CS NPs and T-SF-CS NPs for 3 weeks. Free SF significantly suppressed the activity of the antioxidant enzymes glutathione S-transferase (GST), glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT), and superoxide dismutase (SOD), as well as the levels of glutathione (GSH) and thiobarbituric acid reactive substances (TBARS) as in an indirect measure of free radicals. Interestingly, SF-CS NPs and T-SF-CS-NPs treatments significantly attenuated the inhibitory effects of SF on the activity of these enzymes whereas, GST activity was inhibited. Moreover, the protein expression of GST was downregulated upon treatment of rats with free SF, SF-CS-NPs, and T-SF CS-NPs. In contrast, the activity and protein expression of GPx was induced by SF-CS NPs and T-SF-CS-NPs treatments. The histopathological study showed that SF induced multiple adverse effects on the rat liver architecture which were markedly suppressed particularly by T-SF-CS NPs. In conclusion, chitosan nanoencapsulation of SF counteracted the adverse effects of SF on the activity of antioxidant enzymes and liver architecture. Findings might have significant implications in improving the safety and efficacy of SF treatment of the widely expanding disease conditions.
Collapse
Affiliation(s)
- Salah A. Sheweita
- Department of Clinical Biochemistry, Faculty of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Egypt
| | - Dalia M. Elsayed Alian
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Egypt
| | - Medhat Haroun
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Egypt
| | | | - Ayyub Patel
- Department of Clinical Biochemistry, Faculty of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Labiba El-Khordagui
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt
| |
Collapse
|