1
|
Chan NH, Hawkins CC, Rodrigues BV, Cornet MC, Gonzalez FF, Wu YW. Neuroprotection for neonatal hypoxic-ischemic encephalopathy: A review of novel therapies evaluated in clinical studies. Dev Med Child Neurol 2024. [PMID: 39563426 DOI: 10.1111/dmcn.16184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024]
Abstract
Therapeutic hypothermia is an effective therapy for moderate-to-severe hypoxic-ischemic encephalopathy (HIE) in infants born at term or near-term in high-resource settings. Yet there remains a substantial proportion of infants who do not benefit or who will have significant disability despite therapeutic hypothermia. Novel investigational therapies that may confer additional neuroprotection by targeting known pathogenic mechanisms of hypoxic-ischemic brain injury are under development. This review focuses on putative neuroprotective agents that have shown promise in animal models of HIE, and that have been translated to clinical studies in neonates with HIE. We include agents that have been studied both with and without concurrent therapeutic hypothermia. Our review therefore addresses not just neonatal HIE in high-resource countries where therapeutic hypothermia is the standard of care, but also neonatal HIE in low- and middle-income countries where therapeutic hypothermia has been shown to be ineffective, and where the greatest burden of HIE-related morbidity and mortality exists.
Collapse
Affiliation(s)
- Natalie H Chan
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Cheryl C Hawkins
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Benjamin V Rodrigues
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Marie-Coralie Cornet
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Fernando F Gonzalez
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Yvonne W Wu
- Department of Neurology, University of California, San Francisco, California, USA
| |
Collapse
|
2
|
Notarbartolo V, Badiane BA, Angileri VM, Piro E, Giuffrè M. Antioxidant Therapy in Neonatal Hypoxic Ischemic Encephalopathy: Adjuvant or Future Alternative to Therapeutic Hypothermia? Metabolites 2024; 14:630. [PMID: 39590867 PMCID: PMC11596076 DOI: 10.3390/metabo14110630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Oxidative stress-related diseases in newborns arise from pro-oxidant/antioxidant imbalance in both term and preterm neonates. Pro-oxidant/antioxidant imbalance has shown to be present in different pathological conditions such as hypoxic ischemic encephalopathy (HIE), retinopathy of prematurity (ROP), bronchopulmonary dysplasia (BPD), necrotizing enterocolitis (NEC), and patent ductus arteriosus (PDA). METHODS AND RESULTS We performed a narrative review according to the most recent available literature (2012-2024), using Scopus and PubMed as electronic databases. Many observational and experimental studies in vitro and in vivo have evaluated the effectiveness of antioxidant therapies such as melatonin, erythropoietin (EPO), allopurinol, N-acetylcisteine (NAS), and nitric oxide synthase (NOS) inhibitors in these diseases. Perinatal asphyxia is one of the most important causes of mortality and morbidity in term and near-term newborns. Therapeutic hypothermia (TH) is the gold standard treatment for neonates with moderate-severe perinatal asphyxia, resulting in a reduction in the mortality and neurodevelopmental disability rates. CONCLUSIONS According to the most recent literature and clinical trials, melatonin, allopurinol, NAS, NOS inhibitors, magnesium sulfate, and stem cells stand out as promising as both adjuvants and future probable alternatives to TH in the treatment of HIE.
Collapse
Affiliation(s)
- Veronica Notarbartolo
- Neonatology and Neonatal Intensive Care Unit, University Hospital “Paolo Giaccone”, 90127 Palermo, Italy
| | - Bintu Ayla Badiane
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (B.A.B.); (E.P.); (M.G.)
| | - Vita Maria Angileri
- Neonatal Intensive Care Unit with Neonatology, “G.F. Ingrassia” Hospital Unit, 90131 Palermo, Italy;
| | - Ettore Piro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (B.A.B.); (E.P.); (M.G.)
| | - Mario Giuffrè
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (B.A.B.); (E.P.); (M.G.)
| |
Collapse
|
3
|
Tan S, Alimujiang G, Rejiafu N. A bibliometric study on clinical research in neonatal encephalopathy. Front Pediatr 2024; 12:1403671. [PMID: 39554309 PMCID: PMC11563830 DOI: 10.3389/fped.2024.1403671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
This research presents a comprehensive review of studies on neonatal encephalopathy conducted between 2005 and 2024, utilizing knowledge graph analysis through CiteSpace and VOSviewer software. A search of the Web of Science core database identified 893 articles, with the United States emerging as a prominent contributor in terms of publication volume. Key co-occurrence keywords identified include "Hypoxic-ischemic encephalopathy", "Neonatal encephalopathy", and "Therapeutic hypothermia". Notable contributors, such as Seetha Shankaran and Floris Groenendaal, have significantly advanced research in this area. Leading institutions in this field include the University of Washington, while the journal Pediatrics is recognized as a leading publication in the domain of neonatal encephalopathy. These findings provide a solid foundation for guiding future research endeavors.
Collapse
Affiliation(s)
- Shujun Tan
- Neonatal Center, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Neonatal Center, Xinjiang Hospital of Beijing Children's Hospital, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Neonatal Center, The Seventh People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Gulizuohere Alimujiang
- Neonatal Center, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Neonatal Center, Xinjiang Hospital of Beijing Children's Hospital, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Neonatal Center, The Seventh People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Nuerya Rejiafu
- Neonatal Center, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Neonatal Center, Xinjiang Hospital of Beijing Children's Hospital, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Neonatal Center, The Seventh People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
4
|
Bindal P, Kumar V, Kapil L, Singh C, Singh A. Therapeutic management of ischemic stroke. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2651-2679. [PMID: 37966570 DOI: 10.1007/s00210-023-02804-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/19/2023] [Indexed: 11/16/2023]
Abstract
Stroke is the third leading cause of years lost due to disability and the second-largest cause of mortality worldwide. Most occurrences of stroke are brought on by the sudden occlusion of an artery (ischemic stroke), but sometimes they are brought on by bleeding into brain tissue after a blood vessel has ruptured (hemorrhagic stroke). Alteplase is the only therapy the American Food and Drug Administration has approved for ischemic stroke under the thrombolysis category. Current views as well as relevant clinical research on the diagnosis, assessment, and management of stroke are reviewed to suggest appropriate treatment strategies. We searched PubMed and Google Scholar for the available therapeutic regimes in the past, present, and future. With the advent of endovascular therapy in 2015 and intravenous thrombolysis in 1995, the therapeutic options for ischemic stroke have expanded significantly. A novel approach such as vagus nerve stimulation could be life-changing for many stroke patients. Therapeutic hypothermia, the process of cooling the body or brain to preserve organ integrity, is one of the most potent neuroprotectants in both clinical and preclinical contexts. The rapid intervention has been linked to more favorable clinical results. This study focuses on the pathogenesis of stroke, as well as its recent advancements, future prospects, and potential therapeutic targets in stroke therapy.
Collapse
Affiliation(s)
- Priya Bindal
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Lakshay Kapil
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, HNB Garhwal University (A Central University), Chauras Campus, Distt. Tehri Garhwal, Uttarakhand, 246174, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India.
| |
Collapse
|
5
|
Jiang Z, Wang W, Zhao Y, Li T, Xin D, Gai C, Liu D, Wang Z. Mitochondria-targeted cerium vanadate nanozyme suppressed hypoxia-ischemia injury in neonatal mice via intranasal administration. J Control Release 2024; 365:1074-1088. [PMID: 38101752 DOI: 10.1016/j.jconrel.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Oxidative stress is a major obstacle for neurological functional recovery after hypoxia-ischemia (HI) brain damage. Nanozymes with robust anti-oxidative stress properties offer a therapeutic option for HI injury. However, insufficiency of nanozyme accumulation in the HI brain by noninvasive administration hinders their application. Herein, we reported a cerium vanadate (CeVO4) nanozyme to realize a noninvasive therapy for HI brain in neonatal mice by targeting brain neuron mitochondria. CeVO4 nanozyme with superoxide dismutase activity mainly co-located with neuronal mitochondria 1 h after administration. Pre- and post-HI administrations of CeVO4 nanozyme were able to attenuate acute brain injury, by inhibiting caspase-3 activation, microglia activation, and proinflammation cytokine production in the lesioned cortex 2 d after HI injury. Moreover, CeVO4 nanozyme administration led to short- and long-term functional recovery following HI insult without any potential toxicities in peripheral organs of mice even after prolonged delivery for 4 weeks. These beneficial effects of CeVO4 nanozyme were associated with suppressed oxidative stress and up-regulated nuclear factor erythroid-2-related factor 2 (Nrf2) expression. Finally, we found that Nrf2 inhibition with ML385 abolished the protective effects of CeVO4 nanozyme on HI injury. Collectively, this strategy may provide an applicative perspective for CeVO4 nanozyme therapy in HI brain damage via noninvasive delivery.
Collapse
Affiliation(s)
- Zige Jiang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Wenhan Wang
- State Key Laboratory of Crystal Materials, Shandong University, 27 Shanda Nanlu, Jinan, Shandong 250100, PR China
| | - Yijing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Tingting Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Danqing Xin
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Chengcheng Gai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
6
|
Pavlov V, Papazovska Cherepnalkovski A, Marcic M, Marcic L, Kuzmanic Samija R. The Association of Different Genetic Variants with the Development of Hypoxic-Ischemic Encephalopathy. Biomedicines 2023; 11:2795. [PMID: 37893168 PMCID: PMC10603921 DOI: 10.3390/biomedicines11102795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The aim of this study is to investigate the frequency of six tag SNPs (single nucleotide polymorphisms) within specific genes (F2, F5, F7, MTHFR, NOS2A, PAI 2-1, PAI 2-2, and PAI 3-3): F2 (rs1799963), F5 (rs6025), F7 (rs6046), NOS 2 (rs1137933), PAI 2 (SERPINB2) (rs6103), MTHFR (rs1801133). The study also investigates their association with the development and severity of HIE. The genes F2, F5, and F7 code for proteins involved in blood clotting. MTHFR is a gene that plays a significant role in processing amino acids, the fundamental building blocks of proteins. NOS2A, PAI 2-1, PAI 2-2, and PAI 3-3 are genes involved in the regulation of various physiological processes, such as the relaxation of smooth muscle, regulation of central blood pressure, vasodilatation, and synaptic plasticity. Changes in these genes may be associated with brain injury. This retrospective study included 279 participants, of which 132 participants had Hypoxic-Ischemic Encephalopathy (HIE) and 147 subjects were in the control group. Our study found that certain genetic variants in the rs61103 and rs1137933 polymorphisms were associated with hypoxic-ischemic encephalopathy (HIE) and the findings of the magnetic resonance imaging. There was a correlation between Apgar scores and the degree of damage according to the ultrasound findings. These results highlight the complex relationship between genetic factors, clinical parameters, and the severity of HIE.
Collapse
Affiliation(s)
- Vesna Pavlov
- Department of Neonatology, Clinic for Gynecology and Obstetrics, Clinical Hospital Center Split, 21000 Split, Croatia;
| | - Anet Papazovska Cherepnalkovski
- Department of Neonatology, Clinic for Gynecology and Obstetrics, Clinical Hospital Center Split, 21000 Split, Croatia;
- University Department of Health Studies, University of Split, 21000 Split, Croatia
| | - Marino Marcic
- Department of Neurology, Clinical Hospital Center Split, 21000 Split, Croatia;
| | - Ljiljana Marcic
- Department of Radiology, Polyclinic Medikol, 10000 Zagreb, Croatia;
| | | |
Collapse
|
7
|
Zhang Q, Yao M, Qi J, Song R, Wang L, Li J, Zhou X, Chang D, Huang Q, Li L, Wang N. Puerarin inhibited oxidative stress and alleviated cerebral ischemia-reperfusion injury through PI3K/Akt/Nrf2 signaling pathway. Front Pharmacol 2023; 14:1134380. [PMID: 37284311 PMCID: PMC10240043 DOI: 10.3389/fphar.2023.1134380] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 05/09/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction: Puerarin (PUE) is a natural compound isolated from Puerariae Lobatae Radix, which has a neuroprotective effect on IS. We explored the therapeutic effect and underlying mechanism of PUE on cerebral I/R injury by inhibiting oxidative stress related to the PI3K/Akt/Nrf2 pathway in vitro and in vivo. Methods: The middle cerebral artery occlusion and reperfusion (MCAO/R) rats and oxygen-glucose deprivation and reperfusion (OGD/R) were selected as the models, respectively. The therapeutic effect of PUE was observed using triphenyl tetrazolium and hematoxylin-eosin staining. Tunel-NeuN staining and Nissl staining to quantify hippocampal apoptosis. The reactive oxygen species (ROS) level was detected by flow cytometry and immunofluorescence. Biochemical method to detect oxidative stress levels. The protein expression related to PI3K/Akt/Nrf2 pathway was detected by using Western blotting. Finally, co-immunoprecipitation was used to study the molecular interaction between Keap1 and Nrf2. Results: In vivo and vitro studies showed that PUE improved neurological deficits in rats, as well as decreased oxidative stress. Immunofluorescence and flow cytometry indicated that the release of ROS can be inhibited by PUE. In addition, the Western blotting results showed that PUE promoted the phosphorylation of PI3K and Akt, and enabled Nrf2 to enter the nucleus, which further activated the expression of downstream antioxidant enzymes such as HO-1. The combination of PUE with PI3K inhibitor LY294002 reversed these results. Finally, co-immunoprecipitation results showed that PUE promoted Nrf2-Keap1 complex dissociation. Discussion: Taken together, PUE can activate Nrf2 via PI3K/Akt and promote downstream antioxidant enzyme expression, which could further ameliorate oxidative stress, against I/R-induced Neuron injury.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China
| | - Min Yao
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China
| | - Jiajia Qi
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China
| | - Rui Song
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lei Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China
| | - Jiacheng Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China
| | - Xian Zhou
- National Institute of Complementary Medicine, Western Sydney University, Westmead, NSW, Australia
| | - Dennis Chang
- National Institute of Complementary Medicine, Western Sydney University, Westmead, NSW, Australia
| | - Qi Huang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, China
| | - Lili Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China
- Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Traditional Chinese Medicine, Hefei, China
| | - Ning Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei, China
- Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Traditional Chinese Medicine, Hefei, China
| |
Collapse
|
8
|
Perrone S, Grassi F, Caporilli C, Boscarino G, Carbone G, Petrolini C, Gambini LM, Di Peri A, Moretti S, Buonocore G, Esposito SMR. Brain Damage in Preterm and Full-Term Neonates: Serum Biomarkers for the Early Diagnosis and Intervention. Antioxidants (Basel) 2023; 12:antiox12020309. [PMID: 36829868 PMCID: PMC9952571 DOI: 10.3390/antiox12020309] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/14/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
The Brain is vulnerable to numerous insults that can act in the pre-, peri-, and post-natal period. There is growing evidence that demonstrate how oxidative stress (OS) could represent the final common pathway of all these insults. Fetuses and newborns are particularly vulnerable to OS due to their inability to active the antioxidant defenses. Specific molecules involved in OS could be measured in biologic fluids as early biomarkers of neonatal brain injury with an essential role in neuroprotection. Although S-100B seems to be the most studied biomarker, its use in clinical practice is limited by the complexity of brain damage etiopathogenesis and the time of blood sampling in relation to the brain injury. Reliable early specific serum markers are currently lacking in clinical practice. It is essential to determine if there are specific biomarkers that can help caregivers to monitor the progression of the disease in order to active an early neuroprotective strategy. We aimed to describe, in an educational review, the actual evidence on serum biomarkers for the early identification of newborns at a high risk of neurological diseases. To move the biomarkers from the bench to the bedside, the assays must be not only be of a high sensitivity but suitable for the very rapid processing and return of the results for the clinical practice to act on. For the best prognosis, more studies should focus on the association of these biomarkers to the type and severity of perinatal brain damage.
Collapse
Affiliation(s)
- Serafina Perrone
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Correspondence:
| | - Federica Grassi
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Caporilli
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Giovanni Boscarino
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Giulia Carbone
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Petrolini
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Lucia Maria Gambini
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Antonio Di Peri
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Sabrina Moretti
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | | |
Collapse
|
9
|
Pluta R, Furmaga-Jabłońska W, Januszewski S, Tarkowska A. Melatonin: A Potential Candidate for the Treatment of Experimental and Clinical Perinatal Asphyxia. Molecules 2023; 28:1105. [PMID: 36770769 PMCID: PMC9919754 DOI: 10.3390/molecules28031105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Perinatal asphyxia is considered to be one of the major causes of brain neurodegeneration in full-term newborns. The worst consequence of perinatal asphyxia is neurodegenerative brain damage, also known as hypoxic-ischemic encephalopathy. Hypoxic-ischemic encephalopathy is the leading cause of mortality in term newborns. To date, due to the complex mechanisms of brain damage, no effective or causal treatment has been developed that would ensure complete neuroprotection. Although hypothermia is the standard of care for hypoxic-ischemic encephalopathy, it does not affect all changes associated with encephalopathy. Therefore, there is a need to develop effective treatment strategies, namely research into new agents and therapies. In recent years, it has been pointed out that natural compounds with neuroprotective properties, such as melatonin, can be used in the treatment of hypoxic-ischemic encephalopathy. This natural substance with anti-inflammatory, antioxidant, anti-apoptotic and neurofunctional properties has been shown to have pleiotropic prophylactic or therapeutic effects, mainly against experimental brain neurodegeneration in hypoxic-ischemic neonates. Melatonin is a natural neuroprotective hormone, which makes it promising for the treatment of neurodegeneration after asphyxia. It is supposed that melatonin alone or in combination with hypothermia may improve neurological outcomes in infants with hypoxic-ischemic encephalopathy. Melatonin has been shown to be effective in the last 20 years of research, mainly in animals with perinatal asphyxia but, so far, no clinical trials have been performed on a sufficient number of newborns. In this review, we summarize the advantages and limitations of melatonin research in the treatment of experimental and clinical perinatal asphyxia.
Collapse
Affiliation(s)
- Ryszard Pluta
- Ecotech-Complex Analytical and Programme Centre for Advanced Environmentally-Friendly Technologies, Marie Curie-Skłodowska University in Lublin, 20-612 Lublin, Poland
| | - Wanda Furmaga-Jabłońska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Agata Tarkowska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
10
|
Iannuzzi C, Liccardo M, Sirangelo I. Overview of the Role of Vanillin in Neurodegenerative Diseases and Neuropathophysiological Conditions. Int J Mol Sci 2023; 24:ijms24031817. [PMID: 36768141 PMCID: PMC9915872 DOI: 10.3390/ijms24031817] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/11/2023] [Accepted: 01/14/2023] [Indexed: 01/18/2023] Open
Abstract
Nowadays, bioactive natural products play key roles in drug development due to their safety profile and strong antioxidant power. Vanillin is a natural phenolic compound found in several vanilla beans and widely used for food, cosmetic, and pharmaceutical products. Besides its industrial applications, vanillin possesses several beneficial effects for human health, such as antioxidant activity in addition to anti-inflammatory, anti-mutagenic, anti-metastatic, and anti-depressant properties. Moreover, vanillin exhibits neuroprotective effects on multiple neurological disorders and neuropathophysiological conditions. This study reviews the mechanisms of action by which vanillin prevents neuroinflammation and neurodegeneration in vitro and in vivo systems, in order to provide the latest views on the beneficial properties of this molecule in chronic neurodegenerative diseases and neuropathophysiological conditions.
Collapse
|
11
|
Zironi I, Aicardi G. Hypoxia Depresses Synaptic Transmission in the Primary Motor Cortex of the Infant Rat-Role of Adenosine A 1 Receptors and Nitric Oxide. Biomedicines 2022; 10:2875. [PMID: 36359395 PMCID: PMC9687150 DOI: 10.3390/biomedicines10112875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 09/08/2024] Open
Abstract
The acute and long-term consequences of perinatal asphyxia have been extensively investigated, but only a few studies have focused on postnatal asphyxia. In particular, electrophysiological changes induced in the motor cortex by postnatal asphyxia have not been examined so far, despite the critical involvement of this cortical area in epilepsy. In this study, we exposed primary motor cortex slices obtained from infant rats in an age window (16-18 day-old) characterized by high incidence of hypoxia-induced seizures associated with epileptiform motor behavior to 10 min of hypoxia. Extracellular field potentials evoked by horizontal pathway stimulation were recorded in layers II/III of the primary motor cortex before, during, and after the hypoxic event. The results show that hypoxia reversibly depressed glutamatergic synaptic transmission and neuronal excitability. Data obtained in the presence of specific blockers suggest that synaptic depression was mediated by adenosine acting on pre-synaptic A1 receptors to decrease glutamate release, and by a nitric oxide (NO)/cGMP postsynaptic pathway. These effects are neuroprotective because they limit energy failure. The present findings may be helpful in the preclinical search for therapeutic strategies aimed at preventing acute and long-term neurological consequences of postnatal asphyxia.
Collapse
Affiliation(s)
- Isabella Zironi
- Department of Physics and Astronomy, University of Bologna, 40127 Bologna, Italy
| | - Giorgio Aicardi
- Department for Life Quality Studies, University of Bologna, 40127 Bologna, Italy
| |
Collapse
|
12
|
Hahn KR, Kwon HJ, Yoon YS, Kim DW, Hwang IK. Phosphoglycerate kinase 1 protects against ischemic damage in the gerbil hippocampus. Aging (Albany NY) 2022; 14:8886-8899. [PMID: 36260875 PMCID: PMC9740370 DOI: 10.18632/aging.204343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 10/10/2022] [Indexed: 12/14/2022]
Abstract
Phosphoglycerate kinase 1 (PGK1) is a metabolic enzyme that converts 1,3-diphosphoglycerate to 3-phosphoglycerate. In the current study, we synthesized a PEP-1-PGK1 fusion protein that can cross the blood-brain barrier and cell membrane, and the effects of PEP-1-PGK1 against oxidative stress were investigated HT22 cells and ischemic gerbil brain. The PEP-1-PGK1 protein and its control protein (Con-PGK1) were treated and permeability was evaluated HT22 cells. The PEP-1-PGK1 was introduced into HT22 cells depending on its concentration and incubation time and was gradually degraded over 36 h after treatment. PEP-1-PGK1, but not Con-PGK1, significantly ameliorated H2O2-induced cell damage and reactive oxygen species formation in HT22 cells. Additionally, PEP-1-PGK1, but not Con-PGK1, mitigated ischemia-induced hyperlocomotion 1 d after ischemia and 4 d after ischemia of neuronic cell death. PEP-1-PGK1 treatment significantly alleviated the raised lactate and succinate dehydrogenase activities in the early (15 min to 6 h) and late (4 and 7 d) stages of ischemia, respectively. In addition, PEP-1-PGK1 treatment ameliorated the decrease in ATP and pH levels in the late stage (2-7 d) of ischemia. Nuclear factor erythroid-2-related factor 2 (Nrf2) levels accelerated the ischemia-induced increase in the hippocampus 1 d after ischemia after PEP-1-PGK1 treatment. Neuroprotective and ameliorative effects were prominent at a low concentration (0.1 mg/kg), but not at a high concentration (1 mg/kg), of PEP-1-PGK1. Collectively, low concentrations of PEP-1-PGK1 prevented neuronal stress by increasing energy production.
Collapse
Affiliation(s)
- Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, South Korea,Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| |
Collapse
|
13
|
Rasineni GK, Panigrahy N, Rath SN, Chinnaboina M, Konanki R, Chirla DK, Madduri S. Diagnostic and Therapeutic Roles of the "Omics" in Hypoxic-Ischemic Encephalopathy in Neonates. Bioengineering (Basel) 2022; 9:498. [PMID: 36290466 PMCID: PMC9598631 DOI: 10.3390/bioengineering9100498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Perinatal asphyxia and neonatal encephalopathy remain major causes of neonatal mortality, despite the improved availability of diagnostic and therapeutic tools, contributing to neurological and intellectual disabilities worldwide. An approach using a combination of clinical data, neuroimaging, and biochemical parameters is the current strategy towards the improved diagnosis and prognosis of the outcome in neonatal hypoxic-ischemic encephalopathy (HIE) using bioengineering methods. Traditional biomarkers are of little use in this multifactorial and variable phenotype-presenting clinical condition. Novel systems of biology-based "omics" approaches (genomics, transcriptome proteomics, and metabolomics) may help to identify biomarkers associated with brain and other tissue injuries, predicting the disease severity in HIE. Biomarker studies using omics technologies will likely be a key feature of future neuroprotective treatment methods and will help to assess the successful treatment and long-term efficacy of the intervention. This article reviews the roles of different omics as biomarkers of HIE and outlines the existing knowledge of our current understanding of the clinical use of different omics molecules as novel neonatal brain injury biomarkers, which may lead to improved interventions related to the diagnostic and therapeutic aspects of HIE.
Collapse
Affiliation(s)
- Girish Kumar Rasineni
- LCMS Division, Tenet Medcorp Pvt. Ltd., 54 Kineta Towers Road No 3, Banjara Hills, Hyderabad 500034, India
| | | | - Subha Narayan Rath
- Regenerative Medicine and Stem Cell Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana 502284, India
| | - Madhurarekha Chinnaboina
- LCMS Division, Tenet Medcorp Pvt. Ltd., 54 Kineta Towers Road No 3, Banjara Hills, Hyderabad 500034, India
| | - Ramesh Konanki
- Department of Pediatric Neurology, Rainbow Children’s Hospital, Hyderabad 500034, India
| | - Dinesh Kumar Chirla
- Department of Neonatology, Rainbow Children’s Hospital, Hyderabad 500034, India
| | - Srinivas Madduri
- Bioengineering and Regenerative Medicine, Department of Biomedical Engineering, University of Basel, University Hospital Basel, 4001 Basel, Switzerland
- Department of Surgery, Bioengineering and Neuroregeneration, University of Geneva, University Hospital Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
14
|
Zdolińska-Malinowska I, Boruczkowski D, Hołowaty D, Krajewski P, Snarski E. Rationale for the Use of Cord Blood in Hypoxic-Ischaemic Encephalopathy. Stem Cells Int 2022; 2022:9125460. [PMID: 35599846 PMCID: PMC9117076 DOI: 10.1155/2022/9125460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 02/16/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022] Open
Abstract
Hypoxic-ischaemic encephalopathy (HIE) is a severe complication of asphyxia at birth. Therapeutic hypothermia, the standard method for HIE prevention, is effective in only 50% of the cases. As the understanding of the immunological basis of these changes increases, experiments have begun with the use of cord blood (CB) because of its neuroprotective properties. Mechanisms for the neuroprotective effects of CB stem cells include antiapoptotic and anti-inflammatory actions, stimulation of angiogenesis, production of trophic factors, and mitochondrial donation. In several animal models of HIE, CB decreased oxidative stress, cell death markers, CD4+ T cell infiltration, and microglial activation; restored normal brain metabolic activity; promoted neurogenesis; improved myelination; and increased the proportion of mature oligodendrocytes, neuron numbers in the motor cortex and somatosensory cortex, and brain weight. These observations translate into motor strength, limb function, gait, and cognitive function and behaviour. In humans, the efficacy and safety of CB administration were reported in a few early clinical studies which confirmed the feasibility and safety of this intervention for up to 10 years. The results of these studies showed an improvement in the developmental outcomes over hypothermia. Two phase-2 clinical studies are ongoing under the United States regulations, namely one controlled study and one blinded study.
Collapse
Affiliation(s)
| | - Dariusz Boruczkowski
- Polski Bank Komórek Macierzystych S.A. (FamiCord Group), Jana Pawła II 29, 00-86 Warsaw, Poland
| | - Dominika Hołowaty
- Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza Square 1/3, 02-015 Warsaw, Poland
| | - Paweł Krajewski
- Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza Square 1/3, 02-015 Warsaw, Poland
| | - Emilian Snarski
- Polski Bank Komórek Macierzystych S.A. (FamiCord Group), Jana Pawła II 29, 00-86 Warsaw, Poland
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
15
|
Roles of Nitric Oxide in Brain Ischemia and Reperfusion. Int J Mol Sci 2022; 23:ijms23084243. [PMID: 35457061 PMCID: PMC9028809 DOI: 10.3390/ijms23084243] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 01/27/2023] Open
Abstract
Brain ischemia and reperfusion (I/R) is one of the most severe clinical manifestations of ischemic stroke, placing a significant burden on both individuals and society. The only FDA-approved clinical treatment for ischemic stroke is tissue plasminogen activator (t-PA), which rapidly restores cerebral blood flow but can have severe side effects. The complex pathological process of brain I/R has been well-established in the past few years, including energy metabolism disorders, cellular acidosis, doubling of the synthesis or release of excitotoxic amino acids, intracellular calcium homeostasis, free radical production, and activation of apoptotic genes. Recently, accumulating evidence has shown that NO may be strongly related to brain I/R and involved in complex pathological processes. This review focuses on the role of endogenous NO in pathological processes in brain I/R, including neuronal cell death and blood brain barrier disruption, to explore how NO impacts specific signaling cascades and contributes to brain I/R injury. Moreover, NO can rapidly react with superoxide to produce peroxynitrite, which may also mediate brain I/R injury, which is discussed here. Finally, we reveal several therapeutic approaches strongly associated with NO and discuss their potential as a clinical treatment for ischemic stroke.
Collapse
|
16
|
Ovali F. Hemodynamic changes and evaluation during hypoxic-ischemic encephalopathy and therapeutic hypothermia. Early Hum Dev 2022; 167:105563. [PMID: 35248984 DOI: 10.1016/j.earlhumdev.2022.105563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
Multiorgan damage is a hallmark of hypoxic-ischemic encephalopathy and cardiovascular and hemodynamic changes during asphyxia contribute significantly to the brain damage. The main insult to the heart is myocardial damage and associated ventricular dysfunction, which is manifested by reduced preload and afterload. The immature myocardium reacts to asphyxia by bradycardia and reduced contractile capacity. Pulmonary hypertension aggrevates cardiac dysfunction. Hypothermia is the only effective treatment for HIE but it may also affect the heart and peripheral vascular system leading to bradycardia and peripheral vasoconstriction. In fact, these effects might be cardioprotective also. Rewarming after hypothermia may increase the heart rate and cardiac metabolism, augmenting the cardiac output. Monitoring of patient with HIE during and after hypothermia is possible by using near-infrared spectroscopy, echocardiography and electrocardiography. Cerebral effects may be monitored by magnetic resonance imaging also. Management should include the physiological status of the patient and appropriate treatments, including inotropes, vasopressors or rarely fluid boluses. Dopamine should not be used unless absolutely necessary. Drugs like melatonin and magnesium are under investigation. All treatments should be evidence-based and targeted echocardiography should be used more often in these vulnerable infants.
Collapse
Affiliation(s)
- Fahri Ovali
- Istanbul Medeniyet University, Medical Faculty, Department of Pediatrics, Division of Neonatology, Göztepe, İstanbul, Turkey.
| |
Collapse
|
17
|
Perrone S, Lembo C, Gironi F, Petrolini C, Catalucci T, Corbo G, Buonocore G, Gitto E, Esposito SMR. Erythropoietin as a Neuroprotective Drug for Newborn Infants: Ten Years after the First Use. Antioxidants (Basel) 2022; 11:antiox11040652. [PMID: 35453337 PMCID: PMC9031072 DOI: 10.3390/antiox11040652] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 01/27/2023] Open
Abstract
Protective strategies against perinatal brain injury represent a major challenge for modern neonatology. Erythropoietin (Epo) enhances endogenous mechanisms of repair and angiogenesis. In order to analyse the newest evidence on the role of Epo in prematurity, hypoxic ischemic encephalopathy (HIE) and perinatal stroke, a critical review using 2020 PRISMA statement guidelines was conducted. This review uncovered 26 clinical trials examining the use of Epo for prematurity and brain injury-related outcomes. The effects of Epo on prematurity were analysed in 16 clinical trials. Erythropoietin was provided until 32–35 weeks of corrected postnatal age with a dosage between 500–3000 UI/kg/dose. Eight trials reported the Epo effects on HIE term newborn infants: Erythropoietin was administered in the first weeks of life, at different multiple doses between 250–2500 UI/kg/dose, as either an adjuvant therapy with hypothermia or a substitute for hypothermia. Two trials investigated Epo effects in perinatal stroke. Erythropoietin was administered at a dose of 1000 IU/kg for three days. No beneficial effect in improving morbidity was observed after Epo administration in perinatal stroke. A positive effect on neurodevelopmental outcome seems to occur when Epo is used as an adjuvant therapy with hypothermia in the HIE newborns. Administration of Epo in preterm infants still presents inconsistencies with regard to neurodevelopmental outcome. Clinical trials show significant differences mainly in target population and intervention scheme. The identification of specific markers and their temporal expression at different time of recovery after hypoxia-ischemia in neonates might be implemented to optimize the therapeutic scheme after hypoxic-ischemic injury in the developing brain. Additional studies on tailored regimes, accounting for the risk stratification of brain damage in newborns, are required.
Collapse
Affiliation(s)
- Serafina Perrone
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (C.P.); (S.M.R.E.)
- Correspondence:
| | - Chiara Lembo
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Federica Gironi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Chiara Petrolini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (C.P.); (S.M.R.E.)
| | - Tiziana Catalucci
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Giulia Corbo
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Eloisa Gitto
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | | |
Collapse
|
18
|
Plinia trunciflora Extract Administration Prevents HI-Induced Oxidative Stress, Inflammatory Response, Behavioral Impairments, and Tissue Damage in Rats. Nutrients 2022; 14:nu14020395. [PMID: 35057576 PMCID: PMC8779767 DOI: 10.3390/nu14020395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/14/2022] Open
Abstract
The disruption of redox homeostasis and neuroinflammation are key mechanisms in the pathogenesis of brain hypoxia–ischemia (HI); medicinal plants have been studied as a therapeutic strategy, generally associated with the prevention of oxidative stress and inflammatory response. This study evaluates the neuroprotective role of the Plinia trunciflora fruit extract (PTE) in neonatal rats submitted to experimental HI. The HI insult provoked a marked increase in the lipoperoxidation levels and glutathione peroxidase (GPx) activity, accompanied by a decrease in the brain concentration of glutathione (GSH). Interestingly, PTE was able to prevent most of the HI-induced pro-oxidant effects. It was also observed that HI increased the levels of interleukin-1β in the hippocampus, and that PTE-treatment prevented this effect. Furthermore, PTE was able to prevent neuronal loss and astrocyte reactivity induced by HI, as demonstrated by NeuN and GFAP staining, respectively. PTE also attenuated the anxiety-like behavior and prevented the spatial memory impairment caused by HI. Finally, PTE prevented neural tissue loss in the brain hemisphere, the hippocampus, cerebral cortex, and the striatum ipsilateral to the HI. Taken together our results provide good evidence that the PTE extract has the potential to be investigated as an adjunctive therapy in the treatment of brain insult caused by neonatal hypoxia–ischemia.
Collapse
|
19
|
Fiani B, Chacon D, Jarrah R, Barthelmass M, Covarrubias C. Neuroprotective strategies of cerebrolysin for the treatment of infants with neonatal hypoxic-ischemic encephalopathy. Acta Neurol Belg 2021; 121:1401-1406. [PMID: 34494216 DOI: 10.1007/s13760-021-01795-y] [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: 08/11/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Perinatal asphyxia (PA) is a devastating neonatal condition characterized by a lack of oxygen supporting the organ systems. PA can lead to hypoxic-ischemic encephalopathy (HIE), a brain dysfunction due to oxygen deprivation with a complex neurological sequela. The pathophysiology of HIE and PA is not entirely understood, with therapeutic hypothermia being the standard treatment with only limited value. However, alternative neuroprotective therapies can be a potential treatment modality. METHODS In this review, we will characterize the biochemical mechanisms of PA and HIE, while also giving insight into cerebrolysin, a neuroprotective treatment used for HIE and PA. RESULTS We found that cerebrolysin has up to 6-month treatment window post-ischemic insult. Cerebrolysin injections of 0.1 ml/kg of body weight twice per week were found to provide gross motor and speech deficit improvement. CONCLUSION Our literature search emphasizes the positive effects of cerebrolysin for general improvement outcomes. Nevertheless, biomarker establishment is warranted to improve patient outcomes.
Collapse
Affiliation(s)
- Brian Fiani
- Department of Neurosurgery, Desert Regional Medical Center, 1150 N. Indian Canyon Dr., Palm Springs, CA, 92262, USA.
| | - Daniel Chacon
- School of Medicine, Ross University, Bridgetown, Barbados
| | - Ryan Jarrah
- Department of Neuro-Informatics, Mayo Clinic, Rochester, MN, USA
| | - Michaela Barthelmass
- School of Medicine, California University of Sciences and Medicine, Colton, CA, USA
| | - Claudia Covarrubias
- School of Medicine, Universidad Anáhuac Querétaro, Santiago de Querétaro, México
| |
Collapse
|
20
|
Succinate accumulation contributes to oxidative stress and iron accumulation in pentylenetetrazol-induced epileptogenesis and kainic acid-induced seizure. Neurochem Int 2021; 149:105123. [PMID: 34224804 DOI: 10.1016/j.neuint.2021.105123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/17/2021] [Accepted: 06/30/2021] [Indexed: 11/21/2022]
Abstract
This study explored the role of succinate accumulation in the oxidative stress and iron accumulation in both pentylenetetrazol (PTZ)-induced epileptogenesis and kainic acid (KA)-induced status epilepticus (SE). The levels of succinate, oxidative stress, iron content, iron-related protein expression, and the severity of neuronal injury and seizures were measured in both models. We found that increased concentrations of succinate were associated with increased levels of oxidative stress, iron content, iron regulator protein, and iron importer divalent metal transporter 1, as well as decreased levels of iron exporter ferropotin 1. Aggravated neuronal injury was observed in the hippocampi and cortices of both models. The cell-permeable molecule dimethyl malonate (DM), a competitive inhibitor of succinate dehydrogenase (SDH), significantly attenuated succinate accumulation, reduced the oxidative stress and iron levels, and mitigated the severity of the seizures and neuronal injury. Our results thus indicate that the accumulation of succinate due to the reverse catalysis of SDH may exacerbate oxidative stress and thus induce iron accumulation and neuronal injury in both models. Targeting succinate accumulation may achieve neuroprotective and anti-seizure effects.
Collapse
|
21
|
Alzheimer's Disease Associated Presenilin 1 and 2 Genes Dysregulation in Neonatal Lymphocytes Following Perinatal Asphyxia. Int J Mol Sci 2021; 22:ijms22105140. [PMID: 34067945 PMCID: PMC8152038 DOI: 10.3390/ijms22105140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Perinatal asphyxia is mainly a brain disease leading to the development of neurodegeneration, in which a number of peripheral lesions have been identified; however, little is known about the expression of key genes involved in amyloid production by peripheral cells, such as lymphocytes, during the development of hypoxic-ischemic encephalopathy. We analyzed the gene expression of the amyloid protein precursor, β-secretase, presenilin 1 and 2 and hypoxia-inducible factor 1-α by RT-PCR in the lymphocytes of post-asphyxia and control neonates. In all examined periods after asphyxia, decreased expression of the genes of the amyloid protein precursor, β-secretase and hypoxia-inducible factor 1-α was noted in lymphocytes. Conversely, expression of presenilin 1 and 2 genes decreased on days 1–7 and 8–14 but increased after survival for more than 15 days. We believe that the expression of presenilin genes in lymphocytes could be a potential biomarker to determine the severity of the post-asphyxia neurodegeneration or to identify the underlying factors for brain neurodegeneration and get information about the time they occurred. This appears to be the first worldwide data on the role of the presenilin 1 and 2 genes associated with Alzheimer’s disease in the dysregulation of neonatal lymphocytes after perinatal asphyxia.
Collapse
|
22
|
Liu J, Gong Z, Wu J, Liu S, Wang X, Wang J, Xu J, Li J, Zhao Y. Hypoxic postconditioning-induced neuroprotection increases neuronal autophagy via activation of the SIRT1/FoxO1 signaling pathway in rats with global cerebral ischemia. Exp Ther Med 2021; 22:695. [PMID: 33986859 PMCID: PMC8111876 DOI: 10.3892/etm.2021.10127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 09/02/2020] [Indexed: 11/27/2022] Open
Abstract
Hypoxic postconditioning (HPC) has been reported to be a beneficial and promising treatment for global cerebral ischemia (GCI). However, its neuroprotective mechanism remains unclear. The aim of the present study was to determine whether the protective effects of HPC in a rat model of GCI were due to the upregulation of autophagy via the silent information regulator transcript-1 (SIRT1)/Forkhead box protein 1 (FoxO1) pathway. Morris water maze test revealed that HPC attenuated cognitive damage in GCI rats. HPC also significantly increased the levels of the autophagy-related protein LC3-II, SIRT1 and FoxO1 compared with those in the GCI group. However, the HPC-induced LC3-II upregulation was blocked by the SIRT1 inhibitor EX527. These results suggested that the beneficial effects of HPC on GCI rats were due to the upregulation of ischemiainduced autophagy and involved the SIRT1/FoxO1 signaling pathway.
Collapse
Affiliation(s)
- Junjie Liu
- College of Clinical Medicine, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China.,Department of Neurosurgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Zehua Gong
- College of Clinical Medicine, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China.,Department of Neurosurgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Juan Wu
- College of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Shaopeng Liu
- College of Clinical Medicine, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Xue Wang
- College of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Jingyao Wang
- College of Clinical Medicine, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Jiwei Xu
- Department of Neurosurgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Jianmin Li
- College of Clinical Medicine, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China.,Department of Neurosurgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Yaning Zhao
- College of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
23
|
Wei W, Lu M, Lan XB, Liu N, Su WK, Dushkin AV, Yu JQ. Neuroprotective Effects of Oxymatrine on PI3K/Akt/mTOR Pathway After Hypoxic-Ischemic Brain Damage in Neonatal Rats. Front Pharmacol 2021; 12:642415. [PMID: 33927621 PMCID: PMC8077028 DOI: 10.3389/fphar.2021.642415] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Oxymatrine (OMT), a quinolizidine alkaloid extracted from traditional Chinese herb Sophora flavescens Ait, has drawn attention because of its beneficial bioactivities against hypoxic-ischemic brain damage (HIBD). However, the underlying molecular mechanism remains unclear. In this study, we determined the in vivo and in vitro effects of OMT on seven-day old Sprague-Dawley rats with HIBD and in a rat model of primary hippocampal neuron oxygen glucose deprivation reoxygenation (OGD/R). This study was aimed to evaluate whether OMT exerted neuroprotective effects mediated by the (phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin) PI3K/Akt/mTOR pathway after HIBD. Experimental results showed that the alkaloid significantly improved the early neurofunctional development, brain water content, abnormal pathological changes, and necrosis of neurons after HIBD. Moreover, OMT enhanced the cell viability and stabilized the mitochondrial permeability transition pore in the primary hippocampal neurons after OGD/R. OMT significantly decreased the autophagosome generation, elevated the expression of PI3K, Akt, and mTOR, and simultaneously reversed the mRNA expression of microtubule-associated protein 1-light chain 3 (LC3), Beclin-1, and sequestosomel (P62) induced by hypoxia and ischemia. However, these protective effects against HIBD could be suppressed when rapamycin, a specific inhibitor of mTOR, was included. Hence, the OMT exerted neuroprotective effects against HIBD by attenuating excessive autophagy by mediating the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Wei Wei
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Min Lu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Xiao-bing Lan
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Ning Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Wei-ke Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Alexandr V. Dushkin
- Institute of Solid State Chemistry and Mechanochemistry, Novosibirsk, Russia
| | - Jian-qiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
- Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
24
|
Rey-Funes M, Contartese DS, Peláez R, García-Sanmartín J, Narro-Íñiguez J, Soliño M, Fernández JC, Sarotto A, Ciranna NS, López-Costa JJ, Dorfman VB, Larrayoz IM, Loidl CF, Martínez A. Hypothermic Shock Applied After Perinatal Asphyxia Prevents Retinal Damage in Rats. Front Pharmacol 2021; 12:651599. [PMID: 33897437 PMCID: PMC8060653 DOI: 10.3389/fphar.2021.651599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 02/22/2021] [Indexed: 01/10/2023] Open
Abstract
Perinatal asphyxia (PA) can cause retinopathy and different degrees of visual loss, including total blindness. In a rat model of PA, we have previously shown a protective effect of hypothermia on the retina when applied simultaneously with the hypoxic insult. In the present work, we evaluated the possible protective effect of hypothermia on the retina of PA rats when applied immediately after delivery. Four experimental groups were studied: Rats born naturally as controls (CTL), animals that were exposed to PA for 20 min at 37°C (PA), animals exposed to PA for 20 min at 15°C (HYP), and animals that were exposed to PA for 20 min at 37°C and, immediately after birth, kept for 15 min at 8°C (HYP-PA). To evaluate the integrity of the visual pathway, animals were subjected to electroretinography at 45 days of age. Molecular (real time PCR) and histological (immunohistochemistry, immunofluorescence, TUNEL assay) techniques were applied to the eyes of all experimental groups collected at 6, 12, 24, and 48 h, and 6 days after birth. PA resulted in a significant reduction in the amplitude of the a- and b-wave and oscillatory potentials (OP) of the electroretinogram. All animals treated with hypothermia had a significant correction of the a-wave and OP, but the b-wave was fully corrected in the HYP group but only partially in the HYP-PA group. The number of TUNEL-positive cells increased sharply in the ganglion cell layer of the PA animals and this increase was significantly prevented by both hypothermia treatments. Expression of the cold-shock proteins, cold-inducible RNA binding protein (CIRP) and RNA binding motif protein 3 (RBM3), was undetectable in retinas of the CTL and PA groups, but they were highly expressed in ganglion neurons and cells of the inner nuclear layer of the HYP and HYP-PA groups. In conclusion, our results suggest that a post-partum hypothermic shock could represent a useful and affordable method to prevent asphyxia-related vision disabling sequelae.
Collapse
Affiliation(s)
- Manuel Rey-Funes
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Biología Celular, Histología, Embriología y Genética, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniela S Contartese
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rafael Peláez
- Biomarkers and Molecular Signaling Group, Center for Biomedical Research of La Rioja, Logroño, Spain
| | - Josune García-Sanmartín
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja, Logroño, Spain
| | - Judit Narro-Íñiguez
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja, Logroño, Spain
| | - Manuel Soliño
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Carlos Fernández
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Aníbal Sarotto
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nicolás S Ciranna
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan José López-Costa
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Biología Celular, Histología, Embriología y Genética, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Verónica B Dorfman
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo, Universidad Maimónides, Buenos Aires, Argentina
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling Group, Center for Biomedical Research of La Rioja, Logroño, Spain
| | - C Fabián Loidl
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Biología Celular, Histología, Embriología y Genética, Instituto de Biología Celular y Neurociencia "Prof, E. De Robertis" (IBCN), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alfredo Martínez
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja, Logroño, Spain
| |
Collapse
|
25
|
Mohsenpour H, Pesce M, Patruno A, Bahrami A, Pour PM, Farzaei MH. A Review of Plant Extracts and Plant-Derived Natural Compounds in the Prevention/Treatment of Neonatal Hypoxic-Ischemic Brain Injury. Int J Mol Sci 2021; 22:E833. [PMID: 33467663 PMCID: PMC7830094 DOI: 10.3390/ijms22020833] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Neonatal hypoxic-ischemic (HI) brain injury is one of the major drawbacks of mortality and causes significant short/long-term neurological dysfunction in newborn infants worldwide. To date, due to multifunctional complex mechanisms of brain injury, there is no well-established effective strategy to completely provide neuroprotection. Although therapeutic hypothermia is the proven treatment for hypoxic-ischemic encephalopathy (HIE), it does not completely chang outcomes in severe forms of HIE. Therefore, there is a critical need for reviewing the effective therapeutic strategies to explore the protective agents and methods. In recent years, it is widely believed that there are neuroprotective possibilities of natural compounds extracted from plants against HIE. These natural agents with the anti-inflammatory, anti-oxidative, anti-apoptotic, and neurofunctional regulatory properties exhibit preventive or therapeutic effects against experimental neonatal HI brain damage. In this study, it was aimed to review the literature in scientific databases that investigate the neuroprotective effects of plant extracts/plant-derived compounds in experimental animal models of neonatal HI brain damage and their possible underlying molecular mechanisms of action.
Collapse
Affiliation(s)
- Hadi Mohsenpour
- Department of Pediatrics, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah 75333–67427, Iran;
| | - Mirko Pesce
- Department of Medicine and Aging Sciences, University G. d’Annunzio, 66100 Chieti, Italy
| | - Antonia Patruno
- Department of Medicine and Aging Sciences, University G. d’Annunzio, 66100 Chieti, Italy
| | - Azam Bahrami
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 67158-47141, Iran;
| | - Pardis Mohammadi Pour
- Department of Pharmacognosy, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran;
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 67158-47141, Iran;
| |
Collapse
|
26
|
Ac2-26 Alleviates Brain Injury after Cardiac Arrest and Cardiopulmonary Resuscitation in Rats via the eNOS Pathway. Mediators Inflamm 2020; 2020:3649613. [PMID: 32908448 PMCID: PMC7450310 DOI: 10.1155/2020/3649613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022] Open
Abstract
Background Brain injury is the leading cause of death following cardiac arrest (CA) and cardiopulmonary resuscitation (CPR). Ac2-26 and endothelial nitric oxide synthase (eNOS) have been shown to reduce neuroinflammation. This study is aimed at determining the mechanism by which Ac2-26 protects against inflammation during brain injury following CA and CPR. Methods Sixty-four rats were randomized into sham, saline, Ac2-26, and Ac2-26+L-NIO (endothelial nitric oxide synthase (eNOS) inhibitor) groups. Rats received Ac2-26, Ac2-26+L-NIO, or saline after CPR. Neurologic function was assessed at baseline, 24, and 72 hours after CPR. At 72 hours after resuscitation, serum and brain tissues were collected. Results Blood-brain barrier (BBB) permeability increased, and the number of surviving neurons and neurological function decreased in the saline group compared to the sham group. Anti-inflammatory and proinflammatory factors, neuron-specific enolase (NSE) levels, and the expression of eNOS, phosphorylated (p)-eNOS, inducible nitric oxide synthase (iNOS), and oxidative stress-related factors in the three CA groups significantly increased (P < 0.05). BBB permeability decreased, and the number of surviving neurons and neurological function increased in the Ac2-26 group compared to the saline group (P < 0.05). Ac2-26 increased anti-inflammatory and reduced proinflammatory markers, raised NSE levels, increased the expression of eNOS and p-eNOS, and reduced the expression of iNOS and oxidative stress-related factors compared to the saline group (P < 0.05). The effect of Ac2-26 on brain injury was reversed by L-NIO (P < 0.05). Conclusions Ac2-26 reduced brain injury after CPR by inhibiting oxidative stress and neuroinflammation and protecting the BBB. The therapeutic effect of Ac2-26 on brain injury was largely dependent on the eNOS pathway.
Collapse
|
27
|
Rodríguez M, Valez V, Cimarra C, Blasina F, Radi R. Hypoxic-Ischemic Encephalopathy and Mitochondrial Dysfunction: Facts, Unknowns, and Challenges. Antioxid Redox Signal 2020; 33:247-262. [PMID: 32295425 DOI: 10.1089/ars.2020.8093] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Hypoxic-ischemic events due to intrapartum complications represent the second cause of neonatal mortality and initiate an acute brain disorder known as hypoxic-ischemic encephalopathy (HIE). In HIE, the brain undergoes primary and secondary energy failure phases separated by a latent phase in which partial neuronal recovery is observed. A hypoxic-ischemic event leads to oxygen restriction causing ATP depletion, neuronal oxidative stress, and cell death. Mitochondrial dysfunction and enhanced oxidant formation in brain cells are characteristic phenomena associated with energy failure. Recent Advances: Mitochondrial sources of oxidants in neurons include complex I of the mitochondrial respiratory chain, as a key contributor to O2•- production via succinate by a reverse electron transport mechanism. The reaction of O2•- with nitric oxide (•NO) yields peroxynitrite, a mitochondrial and cellular toxin. Quantitation of the redox state of cytochrome c oxidase, through broadband near-infrared spectroscopy, represents a promising monitoring approach to evaluate mitochondrial dysfunction in vivo in humans, in conjunction with the determination of cerebral oxygenation and their correlation with the severity of brain injury. Critical Issues: The energetic failure being a key phenomenon in HIE connected with the severity of the encephalopathy, measurement of mitochondrial dysfunction in vivo provides an approach to assess evolution, prognosis, and adequate therapies. Restoration of mitochondrial redox homeostasis constitutes a key therapeutic goal. Future Directions: While hypothermia is the only currently accepted therapy in clinical management to preserve mitochondrial function, other mitochondria-targeted and/or redox-based treatments are likely to synergize to ensure further efficacy.
Collapse
Affiliation(s)
- Marianela Rodríguez
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay.,Departamento de Neonatología, Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Valeria Valez
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Carolina Cimarra
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Fernanda Blasina
- Departamento de Neonatología, Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO) and Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
28
|
Smits A, Annaert P, Van Cruchten S, Allegaert K. A Physiology-Based Pharmacokinetic Framework to Support Drug Development and Dose Precision During Therapeutic Hypothermia in Neonates. Front Pharmacol 2020; 11:587. [PMID: 32477113 PMCID: PMC7237643 DOI: 10.3389/fphar.2020.00587] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/16/2020] [Indexed: 12/21/2022] Open
Abstract
Therapeutic hypothermia (TH) is standard treatment for neonates (≥36 weeks) with perinatal asphyxia (PA) and hypoxic-ischemic encephalopathy. TH reduces mortality and neurodevelopmental disability due to reduced metabolic rate and decreased neuronal apoptosis. Since both hypothermia and PA influence physiology, they are expected to alter pharmacokinetics (PK). Tools for personalized dosing in this setting are lacking. A neonatal hypothermia physiology-based PK (PBPK) framework would enable precision dosing in the clinic. In this literature review, the stepwise approach, benefits and challenges to develop such a PBPK framework are covered. It hereby contributes to explore the impact of non-maturational PK covariates. First, the current evidence as well as knowledge gaps on the impact of PA and TH on drug absorption, distribution, metabolism and excretion in neonates is summarized. While reduced renal drug elimination is well-documented in neonates with PA undergoing hypothermia, knowledge of the impact on drug metabolism is limited. Second, a multidisciplinary approach to develop a neonatal hypothermia PBPK framework is presented. Insights on the effect of hypothermia on hepatic drug elimination can partly be generated from in vitro (human/animal) profiling of hepatic drug metabolizing enzymes and transporters. Also, endogenous biomarkers may be evaluated as surrogate for metabolic activity. To distinguish the impact of PA versus hypothermia on drug metabolism, in vivo neonatal animal data are needed. The conventional pig is a well-established model for PA and the neonatal Göttingen minipig should be further explored for PA under hypothermia conditions, as it is the most commonly used pig strain in nonclinical drug development. Finally, a strategy is proposed for establishing and fine-tuning compound-specific PBPK models for this application. Besides improvement of clinical exposure predictions of drugs used during hypothermia, the developed PBPK models can be applied in drug development. Add-on pharmacotherapies to further improve outcome in neonates undergoing hypothermia are under investigation, all in need for dosing guidance. Furthermore, the hypothermia PBPK framework can be used to develop temperature-driven PBPK models for other populations or indications. The applicability of the proposed workflow and the challenges in the development of the PBPK framework are illustrated for midazolam as model drug.
Collapse
Affiliation(s)
- Anne Smits
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Steven Van Cruchten
- Applied Veterinary Morphology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Clinical Pharmacy, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| |
Collapse
|
29
|
Biselele T, Bambi J, Betukumesu DM, Ndiyo Y, Tabu G, Kapinga J, Bola V, Makaya P, Tjabbes H, Vis P, Peeters-Scholte C. A Phase IIa Clinical Trial of 2-Iminobiotin for the Treatment of Birth Asphyxia in DR Congo, a Low-Income Country. Paediatr Drugs 2020; 22:95-104. [PMID: 31960360 DOI: 10.1007/s40272-019-00373-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
AIM The main burden of hypoxic-ischemic encephalopathy falls in low-income countries. 2-Iminobiotin, a selective inhibitor of neuronal and inducible nitric oxide synthase, has been shown to be safe and effective in preclinical studies of birth asphyxia. Recently, safety and pharmacokinetics of 2-iminobiotin treatment on top of hypothermia has been described. Since logistics and the standard of medical care are very different in low-resource settings, the aim of this study was to investigate safety and pharmacokinetics of Two-IminoBiotin in the Democratic Republic of Congo (TIBC). METHODS Near-term neonates, born in Kinshasa, Democratic Republic of Congo, with a Thompson score ≥ 7 were eligible for inclusion. Excluded were patients with (1) inability to insert an umbilical venous catheter for administration of the study drug; (2) major congenital or chromosomal abnormalities; (3) birth weight < 1800 g; (4) clear signs of infection; and (5) moribund patients. Neonates received six infusions of 2-iminobiotin 0.16 mg/kg started within 6 h after birth, with 4-h intervals, targeting an AUC0-4h of 365 ng*h/mL. Safety, defined as vital signs, the need for clinical intervention after administration of study drug, occurrence of (serious) adverse events, and pharmacokinetics were assessed. RESULTS After parental consent, seven patients were included with a median Thompson score of 10 (range 8-16). No relevant changes in vital signs were observed over time. There was no need for clinical intervention due to administration of study drug. Three patients died, two after completing the study protocol, one was moribund at inclusion and should not have been included. Pharmacokinetic data of 2-iminobiotin were best described using a two-compartment model. Median AUC0-4h was 664 ng*h/mL (range 414-917). No safety issues attributed to the administration of 2-iminobiotin were found. CONCLUSION The present dosing regimen resulted in higher AUCs than targeted, necessitating a change in the dose regimen in future efficacy trials. No adverse effects that could be attributed to the use of 2-iminobiotin were observed. EudraCT number 2015-003063-12.
Collapse
Affiliation(s)
- Thérèse Biselele
- Neonatal Unit, Department of Pediatrics, University Hospital of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Jephté Bambi
- Neonatal Unit, Department of Pediatrics, University Hospital of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Dieu M Betukumesu
- Neonatal Unit, Department of Pediatrics, University Hospital of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Yoly Ndiyo
- Neonatal Unit, Department of Pediatrics, University Hospital of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Gabriel Tabu
- Neonatal Unit, Department of Pediatrics, Clinique Ngaliema, Kinshasa, Democratic Republic of Congo
| | - Josée Kapinga
- Neonatal Unit, Department of Pediatrics, Clinique Ngaliema, Kinshasa, Democratic Republic of Congo
| | - Valérie Bola
- Neonatal Unit, Department of Pediatrics, Hôpital Saint Joseph, Kinshasa, Democratic Republic of Congo
| | - Pascal Makaya
- Neonatal Unit, Department of Pediatrics, Hôpital Saint Joseph, Kinshasa, Democratic Republic of Congo
| | - Huibert Tjabbes
- Neurophyxia BV, Onderwijsboulevard 225, 5223 DE, 's-Hertogenbosch, The Netherlands
| | - Peter Vis
- LAP&P Consultants, Leiden, The Netherlands
| | | |
Collapse
|
30
|
Gentle SJ, Tipple TE, Patel R. Neonatal comorbidities and gasotransmitters. Nitric Oxide 2020; 97:27-32. [PMID: 32014495 DOI: 10.1016/j.niox.2020.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/10/2019] [Accepted: 01/29/2020] [Indexed: 01/28/2023]
Abstract
Hydrogen sulfide, nitric oxide, and carbon monoxide are endogenously produced gases that regulate various signaling pathways. The role of these transmitters is complex as constitutive production of these molecules may have anti-inflammatory, anti-microbial, and/or vasodilatory effects whereas induced production or formation of secondary metabolites may lead to cellular death. Given this fine line between friend and foe, therapeutic attenuation of these molecules' production has involved both inhibition of endogenous formation and therapeutic supplementation. All three gases have been implicated as regulators of critical aspects of neonatal physiology, and in turn, comorbidities including necrotizing enterocolitis, hypoxic ischemic encephalopathy, and pulmonary hypertension. In this review, we present current perspectives on these associations, highlight areas where insights remain sparse, and identify areas for potential for future investigations.
Collapse
Affiliation(s)
- Samuel J Gentle
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Trent E Tipple
- Section of Neonatal-Perinatal Medicine, University of Oklahoma College of Medicine, Oklahoma City, OK, USA
| | - Rakesh Patel
- Department of Pathology and Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
31
|
Solevåg AL, Schmölzer GM, Cheung PY. Novel interventions to reduce oxidative-stress related brain injury in neonatal asphyxia. Free Radic Biol Med 2019; 142:113-122. [PMID: 31039399 DOI: 10.1016/j.freeradbiomed.2019.04.028] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 04/15/2019] [Accepted: 04/23/2019] [Indexed: 01/10/2023]
Abstract
Perinatal asphyxia-induced brain injury may present as hypoxic-ischemic encephalopathy in the neonatal period, and disability including cerebral palsy in the long term. The brain injury is secondary to both the hypoxic-ischemic event and the reoxygenation-reperfusion following resuscitation. Early events in the cascade of brain injury can be classified as either inflammation or oxidative stress through the generation of free radicals. The objective of this paper is to present efforts that have been made to limit the oxidative stress associated with hypoxic-ischemic encephalopathy. In the acute phase of ischemia/hypoxia and reperfusion/reoxygenation, the outcomes of asphyxiated infants can be improved by optimizing the initial delivery room stabilization. Interventions include limiting oxygen exposure, and shortening the time to return of spontaneous circulation through improved methods for supporting hemodynamics and ventilation. Allopurinol, melatonin, noble gases such as xenon and argon, and magnesium administration also target the acute injury phase. Therapeutic hypothermia, N-acetylcysteine2-iminobiotin, remote ischemic postconditioning, cannabinoids and doxycycline target the subacute phase. Erythropoietin, mesenchymal stem cells, topiramate and memantine could potentially limit injury in the repair phase after asphyxia. To limit the injurious biochemical processes during the different stages of brain injury, determination of the stage of injury in any particular infant remains essential. Currently, therapeutic hypothermia is the only established treatment in the subacute phase of asphyxia-induced brain injury. The effects and side effects of oxidative stress reducing/limiting medications may however be difficult to predict in infants during therapeutic hypothermia. Future neuroprotection in asphyxiated infants may indeed include a combination of therapies. Challenges include timing, dosing and administration route for each neuroprotectant.
Collapse
Affiliation(s)
- A L Solevåg
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, Lørenskog, Norway
| | - G M Schmölzer
- Centre for the Studies of Asphyxia and Resuscitation, Neonatal Research Unit, Royal Alexandra Hospital, Edmonton, Alberta, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - P-Y Cheung
- Centre for the Studies of Asphyxia and Resuscitation, Neonatal Research Unit, Royal Alexandra Hospital, Edmonton, Alberta, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
32
|
Affiliation(s)
- Máximo Vento
- Division of Neonatology, University and Polytechnic Hospital La Fe, Valencia, Spain.
| |
Collapse
|
33
|
Cytotoxicity, Oxidative Stress, and Autophagy in Human Alveolar Epithelial Cell Line (A549 Cells) Exposed to Standardized Urban Dust. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1176:101-108. [DOI: 10.1007/5584_2019_387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|