1
|
Manoharan SD, Abdul Hamid H, Md Hashim NF, Cheema MS, Chiroma SM, Mustapha M, Mehat MZ. Could protein phosphatase 2A and glycogen synthase kinase-3 beta be targeted by natural compounds to ameliorate Alzheimer's pathologies? Brain Res 2024; 1829:148793. [PMID: 38309553 DOI: 10.1016/j.brainres.2024.148793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/26/2023] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder that impairs memory and cognitive abilities, primarily in the elderly. The burden of AD extends beyond patients, impacting families and caregivers due to the patients' reliance on assistance for daily tasks. The main features of the pathogenesis of AD are beta-amyloid plaques and neurofibrillary tangles (NFTs), that strongly correlate with oxidative stress and inflammation. NFTs result from misfolded and hyperphosphorylated tau proteins. Various studies have focused on tau phosphorylation, indicating protein phosphatase 2A (PP2A) as the primary tau phosphatase and glycogen synthase kinase-3 beta (GSK-3β) as the leading tau kinase. Experimental evidence suggests that inhibition of PP2A and increased GSK-3β activity contribute to neuroinflammation, oxidative stress, and cognitive impairment. Hence, targeting PP2A and GSK-3β with pharmacological approaches shows promise in treating AD. The use of natural compounds in the drug development for AD have been extensively studied for their antioxidant, anti-inflammatory, anti-cholinesterase, and neuroprotective properties, demonstrating therapeutic advantages in neurological diseases. Alongside the development of PP2A activator and GSK-3β inhibitor drugs, natural compounds are likely to have neuroprotective effects by increasing PP2A activity and decreasing GSK-3β levels. Therefore, based on the preclinical and clinical studies, the potential of PP2A and GSK-3β as therapeutic targets of natural compounds are highlighted in this review.
Collapse
Affiliation(s)
- Sushmitaa Dhevii Manoharan
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Hafizah Abdul Hamid
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Nur Fariesha Md Hashim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Manraj Singh Cheema
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Samaila Musa Chiroma
- Newcastle University Medicine Malaysia (NUMed), Iskandar Puteri 79200, Johor, Malaysia.
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia.
| | - Muhammad Zulfadli Mehat
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| |
Collapse
|
2
|
Lai S, Wang P, Gong J, Zhang S. New insights into the role of GSK-3β in the brain: from neurodegenerative disease to tumorigenesis. PeerJ 2023; 11:e16635. [PMID: 38107562 PMCID: PMC10722984 DOI: 10.7717/peerj.16635] [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: 08/25/2023] [Accepted: 11/18/2023] [Indexed: 12/19/2023] Open
Abstract
Glycogen synthase kinase 3 (GSK-3) is a serine/threonine kinase widely expressed in various tissues and organs. Unlike other kinases, GSK-3 is active under resting conditions and is inactivated upon stimulation. In mammals, GSK-3 includes GSK-3 α and GSK-3β isoforms encoded by two homologous genes, namely, GSK3A and GSK3B. GSK-3β is essential for the control of glucose metabolism, signal transduction, and tissue homeostasis. As more than 100 known proteins have been identified as GSK-3β substrates, it is sometimes referred to as a moonlighting kinase. Previous studies have elucidated the regulation modes of GSK-3β. GSK-3β is involved in almost all aspects of brain functions, such as neuronal morphology, synapse formation, neuroinflammation, and neurological disorders. Recently, several comparatively specific small molecules have facilitated the chemical manipulation of this enzyme within cellular systems, leading to the discovery of novel inhibitors for GSK-3β. Despite these advancements, the therapeutic significance of GSK-3β as a drug target is still complicated by uncertainties surrounding the potential of inhibitors to stimulate tumorigenesis. This review provides a comprehensive overview of the intricate mechanisms of this enzyme and evaluates the existing evidence regarding the therapeutic potential of GSK-3β in brain diseases, including Alzheimer's disease, Parkinson's disease, mood disorders, and glioblastoma.
Collapse
Affiliation(s)
- Shenjin Lai
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Peng Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jingru Gong
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Shuaishuai Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| |
Collapse
|
3
|
Voicu V, Brehar FM, Toader C, Covache-Busuioc RA, Corlatescu AD, Bordeianu A, Costin HP, Bratu BG, Glavan LA, Ciurea AV. Cannabinoids in Medicine: A Multifaceted Exploration of Types, Therapeutic Applications, and Emerging Opportunities in Neurodegenerative Diseases and Cancer Therapy. Biomolecules 2023; 13:1388. [PMID: 37759788 PMCID: PMC10526757 DOI: 10.3390/biom13091388] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
In this review article, we embark on a thorough exploration of cannabinoids, compounds that have garnered considerable attention for their potential therapeutic applications. Initially, this article delves into the fundamental background of cannabinoids, emphasizing the role of endogenous cannabinoids in the human body and outlining their significance in studying neurodegenerative diseases and cancer. Building on this foundation, this article categorizes cannabinoids into three main types: phytocannabinoids (plant-derived cannabinoids), endocannabinoids (naturally occurring in the body), and synthetic cannabinoids (laboratory-produced cannabinoids). The intricate mechanisms through which these compounds interact with cannabinoid receptors and signaling pathways are elucidated. A comprehensive overview of cannabinoid pharmacology follows, highlighting their absorption, distribution, metabolism, and excretion, as well as their pharmacokinetic and pharmacodynamic properties. Special emphasis is placed on the role of cannabinoids in neurodegenerative diseases, showcasing their potential benefits in conditions such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. The potential antitumor properties of cannabinoids are also investigated, exploring their potential therapeutic applications in cancer treatment and the mechanisms underlying their anticancer effects. Clinical aspects are thoroughly discussed, from the viability of cannabinoids as therapeutic agents to current clinical trials, safety considerations, and the adverse effects observed. This review culminates in a discussion of promising future research avenues and the broader implications for cannabinoid-based therapies, concluding with a reflection on the immense potential of cannabinoids in modern medicine.
Collapse
Affiliation(s)
- Victor Voicu
- Pharmacology, Toxicology and Clinical Psychopharmacology, “Carol Davila” University of Medicine and Pharmacy in Bucharest, 020021 Bucharest, Romania;
- Medical Section within the Romanian Academy, 010071 Bucharest, Romania
| | - Felix-Mircea Brehar
- Neurosurgery Department, Emergency Clinical Hospital Bagdasar-Arseni, 041915 Bucharest, Romania
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Antonio Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Andrei Bordeianu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Luca-Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.-A.C.-B.); (A.D.C.); (A.B.); (H.P.C.); (B.-G.B.); (L.-A.G.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
4
|
Alipour N, Fallahnezhad S, Bagheri J, Babaloo H, Tahmasebi F, Sazegar G, Haghir H. Increased Apoptosis in Subcortical Regions of The Visual Pathway in Offspring Born to Diabetic Rats. CELL JOURNAL 2023; 25:564-569. [PMID: 37641418 PMCID: PMC10542209 DOI: 10.22074/cellj.2023.1989649.1232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/04/2023] [Accepted: 06/13/2023] [Indexed: 08/31/2023]
Abstract
OBJECTIVE Diabetes in pregnancy is a prevalent disease that can affect the central nervous system of the fetus by hyperglycemia. This study aimed to investigate the impact of maternal diabetes on neuronal apoptosis in the superior colliculus (SC) and the lateral geniculate nucleus (LGN) in male neonates born to diabetic mothers. MATERIALS AND METHODS In this experimental study, female adult rats were separated into three groups: control, diabetic (induced using an intraperitoneal injection of streptozotocin), and insulin-treated diabetic [diabetes controlled by subcutaneous neutral protamine hagedorn (NPH)-insulin injection]. Male neonates from each group were euthanized on 0, 7, and 14 postnatal days (P0, P7, and P14, respectively), and apoptotic cells were identified using TUNEL staining. RESULTS The numerical density per unit area (NA) of apoptotic cells was significantly higher in SC and the dorsal LGN (dLGN) in neonates born to the diabetic rats compared to the control group at P0, P7, and P14. However, insulin treatment normalized the number of apoptotic cells. CONCLUSION This study demonstrated that maternal diabetes increased apoptosis in dLGN and SC of male neonates at P0, P7, and P14.
Collapse
Affiliation(s)
- Nasim Alipour
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Somaye Fallahnezhad
- Nervous System Stem Cell Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Anatomical Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Javad Bagheri
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamideh Babaloo
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Tahmasebi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghasem Sazegar
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Haghir
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Genetic Research Center (MGRC), School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Kalra P, Khan H, Singh TG, Grewal AK. Mechanistic insights on impact of Adenosine monophosphate-activated protein kinase (AMPK) mediated signalling pathways on cerebral ischemic injury. Neurosci Res 2023; 190:17-28. [PMID: 36403790 DOI: 10.1016/j.neures.2022.11.006] [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: 04/15/2022] [Revised: 09/23/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Cerebral ischemia is the primary cause of morbidity and mortality worldwide due to the perturbations in the blood supply to the brain. The brain triggers a cascade of complex metabolic and cellular defects in response to ischemic stress. However, due to the disease heterogeneity and complexity, ischemic injury's metabolic and cellular pathologies remain elusive, and the link between various pathological mechanisms is difficult to determine. Efforts to develop effective treatments for these disorders have yielded limited efficacy, with no proper cure available to date. Recent clinical and experimental research indicates that several neuronal diseases commonly coexist with metabolic dysfunction, which may aggravate neurological symptoms. As a result, it stands to a reason that metabolic hormones could be a potential therapeutic target for major NDDs. Moreover, fasting signals also influence the circadian clock, as AMPK phosphorylates and promotes the degradation of the photo-sensing receptor (cryptochrome). Here, the interplay of AMPK signaling between metabolic regulation and neuronal death and its role for pathogenesis and therapeutics has been studied. We have also highlighted a significant signaling pathway, i.e., the adenosine monophosphate-activated protein kinase (AMPK) involved in the relationship between the metabolism and ischemia, which could be used as a target for future studies therapeutics, and review some of the clinical progress in this area.
Collapse
Affiliation(s)
- Palak Kalra
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India.
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| |
Collapse
|
6
|
Prajjwal P, Asharaf S, Makhanasa D, Yamparala A, Tariq H, Aleti S, Gadam S, Vora N. Association of Alzheimer's dementia with oral bacteria, vitamin B12, folate, homocysteine levels, and insulin resistance along with its pathophysiology, genetics, imaging, and biomarkers. Dis Mon 2023; 69:101546. [PMID: 36931946 DOI: 10.1016/j.disamonth.2023.101546] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Alzheimer's disease is a prevalent form of dementia, particularly among the elderly population. It is characterized by progressive cognitive decline and neurodegeneration. Despite numerous studies, the exact cause of Alzheimer's disease remains uncertain, and various theories have been proposed, including Aβ amyloid deposition in the brain and tau protein hyper-phosphorylation. This review article explores the potential pathogenesis of Alzheimer's disease, focusing on the effects of derangements in the levels of vitamin B12, folate, and homocysteine, as well as the impact of oral bacteria causing periodontitis and insulin resistance, and their relationship to Alzheimer's. Studies have shown that high levels of homocysteine and low levels of vitamin B12 and folate, are associated with an increased risk of developing Alzheimer's disease. The article also explores the link between Alzheimer's disease and oral bacteria, specifically dental infections and periodontitis, which contribute to the inflammatory processes in the nervous system of Alzheimer's patients. There could be derangement in the insulin signaling further causing disruption in glucose metabolism within the brain, suggesting that Alzheimer's disease may represent a form of type 2 diabetes mellitus associated with the brain, commonly known as type 3 diabetes. Neuroimaging techniques, including MRI, PET, and tau PET, can identify the predictive characteristics of Alzheimer's disease, with amyloid PET being the most useful in ruling out the disease. The article concludes by stressing the importance of understanding genetic and neuroimaging factors in the diagnosing and treating Alzheimer's disease.
Collapse
Affiliation(s)
| | - Shahnaz Asharaf
- Internal Medicine, Travancore Medical College, Kollam, Kerala, India
| | | | | | - Halla Tariq
- Internal Medicine, Multan Medical and Dental College, Multan, Pakistan
| | - Soumya Aleti
- Internal Medicine, Berkshire Medical Center, Pittsfield, MA, USA
| | - Srikanth Gadam
- Internal Medicine, Postdoctoral Research Fellow, Mayo Clinic, USA
| | - Neel Vora
- Internal Medicine, B. J. Medical College, Ahmedabad, India
| |
Collapse
|
7
|
Fokina EA, Zakharova IO, Bayunova LV, Avrova DK, Ilyasov IO, Avrova NF. Intranasal Insulin Decreases Autophagic and Apoptotic Death of Neurons in the Rat Hippocampal C1 Region and Frontal Cortex under Forebrain Ischemia–Reperfusion. J EVOL BIOCHEM PHYS+ 2023. [DOI: 10.1134/s0022093023010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
|
8
|
Zakharova IO, Zorina II, Bayunova LV, Shpakov AO, Avrova NF. Protective and Antioxidant Effects of Insulin on Rat Brain Cortical Neurons in an in vitro Model of Oxygen and Glucose Deprivation. J EVOL BIOCHEM PHYS+ 2023. [DOI: 10.1134/s0022093023010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
9
|
Methylmercury promotes oxidative stress and autophagy in rat cerebral cortex: Involvement of PI3K/AKT/mTOR or AMPK/TSC2/mTOR pathways and attenuation by N-acetyl-L-cysteine. Neurotoxicol Teratol 2023; 95:107137. [PMID: 36403891 DOI: 10.1016/j.ntt.2022.107137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 11/13/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022]
Abstract
Methylmercury (MeHg) is a potent neurotoxicant that could induce oxidative stress and autophagy. However, the underlying mechanisms through which MeHg affects the central nervous system have not been fully elucidated, and little has been known of the interaction between oxidative stress and autophagy. Therefore, rats were administrated with different MeHg concentrations to evaluate the neurotoxic effects and autophagy in cerebral cortex. Moreover, we have investigated the neuroprotective role of N-acetyl-L-cysteine (NAC) against MeHg-induced neurotoxicity in order to estimate the regulation effects of oxidative stress on autophagy. A total of 64 rats, 40 of which were randomly divided into control and MeHg-treated (4, 8 and 12 μ mol/kg) groups. The remaining 24 rats were divided into control, NAC control (1 mmol/kg), 12 μ mol/kg MeHg, and NAC pretreatment. Administration of 12 μ mol/kg MeHg significantly increased behavioral and pathological abnormalities, and autophagy levels. In addition, the oxidative stress levels increased, together with abnormal expression of autophagy-related molecules. Pretreatment with NAC significantly prevented MeHg-induced oxidative stress and PI3K/AKT/mTOR or AMPK/TSC2/mTOR-mediated autophagy. In conclusion, the present study suggested that oxidative stress can regulate autophagy through PI3K/AKT/mTOR or AMPK/TSC2/mTOR pathways. This study provides a theoretical basis for the study and treatment of MeHg-induced neurotoxicity.
Collapse
|
10
|
Kubis-Kubiak A, Wiatrak B, Piwowar A. Hyper-glycemia and insulinemia induce morphological changes and modulate secretion of S100B, S100A8, amyloid β 1–40 and amyloid β 1–42, in a model of human dopaminergic neurons. Biomed Pharmacother 2022; 156:113869. [DOI: 10.1016/j.biopha.2022.113869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/02/2022] Open
|
11
|
Zhang J, Li X, Cheng W, Li Y, Shi T, Jiang Y, Wang T, Wang H, Ren D, Zhang R, Zheng Y, Tang J. Chronic carbon black nanoparticles exposure increases lung cancer risk by affecting the cell cycle via circulatory inflammation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 305:119293. [PMID: 35421554 DOI: 10.1016/j.envpol.2022.119293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/22/2022] [Accepted: 04/09/2022] [Indexed: 06/14/2023]
Abstract
As a widely used pure elemental carbon in colloidal particles, carbon black was listed as a group 2B carcinogen by IARC in 2010. The most available mechanism information about carbon black and carcinogenesis are from in vivo or in vitro studies. However, few studies concerned the nanoparticle's real-ambient exposure causing systemic change and further affecting the target organ. Herein, we used an ex vivo biosensor assay to investigate the transcriptome change of primary bronchial epithelial cells after treatment with the plasma from workers with long-term occupational carbon black exposure history. Based on ex vivo biosensor assay and transcriptome sequencing, we found the effect of internal systemic environment on epithelial cells after carbon black exposure was an inflammatory response, which mainly activates cell cycle-related pathways. After exposure to carbon black, the internal systemic environment could activate cancer-related pathways like epithelial-mesenchymal transition, hypoxia, TNF-α signaling via NF-κB. The hub genes in the carbon black group (CDC20 and PLK1) and their correlation with the systemic environment were uncovered by constructing the protein-protein interaction network. Inflammatory cytokines, especially CRP, were strongly correlated with the expression of CDC20 and PLK1. Besides, we also find a strong correlation between CDC20 and cytokinesis-block micronucleus endpoints in peripheral blood (rho = 0.591, P < 0.001). Our results show that long-term carbon black exposure might activate cell cycle-related pathways through circulating inflammation and increase the risk of cancer, while the oxidative stress caused by diesel exhaust particles are mainly related to PAHs exposure. After exposure to carbon black, the systemic environment could activate cancer-related pathways like diesel exhaust particles, increasing the risk of lung cancer. These attempts might provide a further understanding of the indirect effect of chronic occupational inhaled carbon black exposure on pulmonary carcinogenesis.
Collapse
Affiliation(s)
- Jianzhong Zhang
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Xin Li
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Wenting Cheng
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Yanting Li
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Teng Shi
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Yingying Jiang
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Tao Wang
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Hongmei Wang
- The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Dunqiang Ren
- The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Rong Zhang
- School of Public Health, Hebei Medical Univeristy, Shijiazhuang, 050017, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Jinglong Tang
- School of Public Health, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
12
|
Bellia C, Lombardo M, Meloni M, Della-Morte D, Bellia A, Lauro D. Diabetes and cognitive decline. Adv Clin Chem 2022; 108:37-71. [PMID: 35659061 DOI: 10.1016/bs.acc.2021.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Epidemiologic studies have documented an association between diabetes and increased risk of cognitive decline in the elderly. Based on animal model studies, several mechanisms have been proposed to explain such an association, including central insulin signaling, neurodegeneration, brain amyloidosis, and neuroinflammation. Nevertheless, the exact mechanisms in humans remain poorly defined. It is reasonable, however, that many pathways may be involved in these patients leading to cognitive impairment. A major aim of clinicians is identifying early onset of neurologic signs and symptoms in elderly diabetics to improve quality of life of those with cognitive impairment and reduce costs associated with long-term complications. Several biomarkers have been proposed to identify diabetics at higher risk of developing dementia and diagnose early stage dementia. Although biomarkers of brain amyloidosis, neurodegeneration and synaptic plasticity are commonly used to diagnose dementia, especially Alzheimer disease, their role in diabetes remains unclear. The aim of this review is to explore the molecular mechanisms linking diabetes with cognitive decline and present the most important findings on the clinical use of biomarkers for diagnosing and predicting early cognitive decline in diabetics.
Collapse
Affiliation(s)
- Chiara Bellia
- Department of Biomedicine, Neurosciences, and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Mauro Lombardo
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Rome, Italy
| | - Marco Meloni
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - David Della-Morte
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Rome, Italy; Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; Department of Neurology and Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Alfonso Bellia
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Davide Lauro
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
13
|
Li F, Yang H, Cao Y, Li D, Ma J, Liu P. DOX-loaded silver nanotriangles and photothermal therapy exert a synergistic antibreast cancer effect via ROS/ERK1/2 signaling pathway. NANOTECHNOLOGY 2021; 33:075101. [PMID: 34749347 DOI: 10.1088/1361-6528/ac378c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/08/2021] [Indexed: 06/13/2023]
Abstract
The combination of multiple therapies has been proved to be more effective than a single therapy for many cancers. This study aimed to investigate the synergistic antibreast cancer effect of doxorubicin-loaded silver nanotriangles (DOX-AgNTs) combined with near-infrared (NIR) irradiation and explore the underlying mechanism. AgNTs were prepared by a chemical method and DOX was loaded via electrostatic adsorption. Characterization was performed by transmission electron microscopy, ultraviolet-visible spectroscopy and dynamic light scattering. The viability of MDA-MB-231 cells was detected by using MTT assay to evaluate the synergistic anticancer effect of DOX-AgNTs combined with NIR irradiation. The intracellular reactive oxygen species (ROS) level and cell apoptosis were analyzed by flow cytometry. Mitochondrial membrane potential (MMP) was measured with fluorescence microscopy. The mechanism was further investigated with ROS scavenger N-acetylcysteine and specific inhibitors of extracellular signal-regulated kinase 1/2 (ERK1/2), C-jun N-terminal kinase and p38 pathways. Characterization results revealed that the prepared AgNTs were mostly triangular and the mean edge length was about 126 nm. The combination of DOX-AgNTs and NIR exhibited a superior synergistic anticancer effect over single DOX-AgNTs or photothermal therapy (PTT). N-acetylcysteine and ERK1/2 inhibitor U0126 were found to significantly rescue the decreased cell viability, declined MMP and increased apoptosis induced by the combined treatment. Our results suggested that DOX-AgNTs combined with PTT performed a synergistic antibreast cancer effect. The synergy might be closely associated with the excessive production of ROS, changed MMP and the activation of ERK1/2 signaling pathway. These findings might provide a new perspective for the development of breast cancer treatments with excellent efficacy.
Collapse
Affiliation(s)
- Fan Li
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Huiquan Yang
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Yuyu Cao
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Dongdong Li
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Jing Ma
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Peidang Liu
- School of Medicine, Southeast University, Nanjing, People's Republic of China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
14
|
Insulin and α-Tocopherol Enhance the Protective Effect of Each Other on Brain Cortical Neurons under Oxidative Stress Conditions and in Rat Two-Vessel Forebrain Ischemia/Reperfusion Injury. Int J Mol Sci 2021; 22:ijms222111768. [PMID: 34769198 PMCID: PMC8584186 DOI: 10.3390/ijms222111768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/15/2021] [Accepted: 10/27/2021] [Indexed: 12/18/2022] Open
Abstract
Clinical trials show that insulin administered intranasally is a promising drug to treat neurodegenerative diseases, but at high doses its use may result in cerebral insulin resistance. Identifying compounds which could enhance the protective effects of insulin, may be helpful to reduce its effective dose. Our aim was thus to study the efficiency of combined use of insulin and α-tocopherol (α-T) to increase the viability of cultured cortical neurons under oxidative stress conditions and to normalize the metabolic disturbances caused by free radical reaction activation in brain cortex of rats with two-vessel forebrain ischemia/reperfusion injury. Immunoblotting, flow cytometry, colorimetric, and fluorometric techniques were used. α-T enhanced the protective and antioxidative effects of insulin on neurons in oxidative stress, their effects were additive. At the late stages of oxidative stress, the combined action of insulin and α-T increased Akt-kinase activity, inactivated GSK-3beta and normalized ERK1/2 activity in cortical neurons, it was more effective than either drug action. In the brain cortex, ischemia/reperfusion increased the lipid peroxidation product content and caused Na+,K+-ATPase oxidative inactivation. Co-administration of insulin (intranasally, 0.25 IU/rat) and α-T (orally, 50 mg/kg) led to a more pronounced normalization of the levels of Schiff bases, conjugated dienes and trienes and Na+,K+-ATPase activity than administration of each drug alone. Thus, α-T enhances the protective effects of insulin on cultured cortical neurons in oxidative stress and in the brain cortex of rats with cerebral ischemia/reperfusion injury.
Collapse
|
15
|
Shi H, Fang Y, Huang L, Gao L, Lenahan C, Okada T, Travis ZD, Xie S, Tang H, Lu Q, Liu R, Tang J, Cheng Y, Zhang JH. Activation of Galanin Receptor 1 with M617 Attenuates Neuronal Apoptosis via ERK/GSK-3β/TIP60 Pathway After Subarachnoid Hemorrhage in Rats. Neurotherapeutics 2021; 18:1905-1921. [PMID: 34086200 PMCID: PMC8609084 DOI: 10.1007/s13311-021-01066-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2021] [Indexed: 02/07/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a devastating cerebrovascular disease. Neuronal apoptosis plays an important pathological role in early brain injury after SAH. Galanin receptor 1 (GalR1) activation was recently shown to be anti-apoptotic in the setting of ischemic stroke. This study aimed to explore the anti-neuronal apoptosis effect of GalR1 activation after SAH, as well as the underlying mechanisms. GalR1 CRISPR and GalR1 selective agonist, M617, was administered, respectively. Extracellular-signal-regulated kinase (ERK) inhibitor (U0126) and glycogen synthase kinase 3-beta (GSK3-β) CRISPR were administered to investigate the involvement of the ERK/GSK3-β pathway in GalR1-mediated neuroprotection after SAH. Outcome assessments included neurobehavioral tests, western blot, and immunohistochemistry. The results showed that endogenous ligand galanin (Gal) and GalR1 were markedly increased in the ipsilateral brain hemisphere at 12 h and 24 h after SAH. GalR1 were expressed mainly in neurons, but expression was also observed in some astrocytes and microglia. GalR1 CRISPR knockdown exacerbated neurological deficits and neuronal apoptosis 24 h after SAH. Moreover, activation of GalR1 with M617 significantly improved short- and long-term neurological deficits but decreased neuronal apoptosis after SAH. Furthermore, GalR1 activation dysregulated the protein levels of phosphorylated ERK and GSK-3β, but downregulated the phosphorylated Tat-interactive protein 60 (TIP60) and cleaved caspase-3 at 24 h after SAH. GalR1 CRISPR, U0126, and GSK-3β CRISPR abolished the beneficial effects of GalR1 activation at 24 h after SAH in rats. Collectively, the present study demonstrated that activation of GalR1 using M617 attenuated neuronal apoptosis through the ERK/GSK-3β/TIP60 pathway after SAH in rats. GalR1 may serve as a promising therapeutic target for SAH patients.
Collapse
Affiliation(s)
- Hui Shi
- Department of Neurosurgery, Chongqing Medical University, Yongchuan Hospital, Yongchuan, Chongqing, China
| | - Yuanjian Fang
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lei Huang
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Ling Gao
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Takeshi Okada
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Zachary D Travis
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Shucai Xie
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Hong Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Qin Lu
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Rui Liu
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Yuan Cheng
- Department of Neurosurgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - John H Zhang
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA.
- Department of Neurosurgery and Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA.
| |
Collapse
|
16
|
Inflammasome NLRP3 Potentially Links Obesity-Associated Low-Grade Systemic Inflammation and Insulin Resistance with Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22115603. [PMID: 34070553 PMCID: PMC8198882 DOI: 10.3390/ijms22115603] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/16/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common form of neurodegenerative dementia. Metabolic disorders including obesity and type 2 diabetes mellitus (T2DM) may stimulate amyloid β (Aβ) aggregate formation. AD, obesity, and T2DM share similar features such as chronic inflammation, increased oxidative stress, insulin resistance, and impaired energy metabolism. Adiposity is associated with the pro-inflammatory phenotype. Adiposity-related inflammatory factors lead to the formation of inflammasome complexes, which are responsible for the activation, maturation, and release of the pro-inflammatory cytokines including interleukin-1β (IL-1β) and interleukin-18 (IL-18). Activation of the inflammasome complex, particularly NLRP3, has a crucial role in obesity-induced inflammation, insulin resistance, and T2DM. The abnormal activation of the NLRP3 signaling pathway influences neuroinflammatory processes. NLRP3/IL-1β signaling could underlie the association between adiposity and cognitive impairment in humans. The review includes a broadened approach to the role of obesity-related diseases (obesity, low-grade chronic inflammation, type 2 diabetes, insulin resistance, and enhanced NLRP3 activity) in AD. Moreover, we also discuss the mechanisms by which the NLRP3 activation potentially links inflammation, peripheral and central insulin resistance, and metabolic changes with AD.
Collapse
|
17
|
Al Hussein Al Awamlh S, Wareham LK, Risner ML, Calkins DJ. Insulin Signaling as a Therapeutic Target in Glaucomatous Neurodegeneration. Int J Mol Sci 2021; 22:4672. [PMID: 33925119 PMCID: PMC8124776 DOI: 10.3390/ijms22094672] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 01/28/2023] Open
Abstract
Glaucoma is a multifactorial disease that is conventionally managed with treatments to lower intraocular pressure (IOP). Despite these efforts, many patients continue to lose their vision. The degeneration of retinal ganglion cells (RGCs) and their axons in the optic tract that characterizes glaucoma is similar to neurodegeneration in other age-related disorders of the central nervous system (CNS). Identifying the different molecular signaling pathways that contribute to early neuronal dysfunction can be utilized for neuroprotective strategies that prevent degeneration. The discovery of insulin and its receptor in the CNS and retina led to exploration of the role of insulin signaling in the CNS. Historically, insulin was considered a peripherally secreted hormone that regulated glucose homeostasis, with no obvious roles in the CNS. However, a growing number of pre-clinical and clinical studies have demonstrated the potential of modulating insulin signaling in the treatment of neurodegenerative diseases. This review will highlight the role that insulin signaling plays in RGC neurodegeneration. We will focus on how this pathway can be therapeutically targeted to promote RGC axon survival and preserve vision.
Collapse
Affiliation(s)
- Sara Al Hussein Al Awamlh
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.H.A.A.); (L.K.W.); (M.L.R.)
| | - Lauren K. Wareham
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.H.A.A.); (L.K.W.); (M.L.R.)
| | - Michael L. Risner
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.H.A.A.); (L.K.W.); (M.L.R.)
| | - David J. Calkins
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.H.A.A.); (L.K.W.); (M.L.R.)
- Department of Ophthalmology & Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
18
|
Zhang J, Lai ZP, Chen P, Ying Y, Zhuang J, Yu KM. Glycogen synthase kinase-3β inhibitor SB216763 promotes DNA repair in ischemic retinal neurons. Neural Regen Res 2021; 16:394-400. [PMID: 32859805 PMCID: PMC7896226 DOI: 10.4103/1673-5374.290913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glycogen synthase kinase-3β (GSK-3β) has been shown to attenuate DNA damage in nerve cells, thereby enhancing neuronal survival under pathological conditions; however, the underlying mechanism remains unclear. An in vitro serum-starvation retinal neuron model and in vivo ischemia/reperfusion retina injury rat model were established and treated with SB216763, a GSK-3β inhibitor. SB21673 decreased the formation of γ-H2A histone family member X foci and enhanced the viability of ischemic retinal neurons. In addition, SB216763 upregulated expression of phosphorylated-CREB1, a ligase IV transcription factor, and significantly increased the transcriptional activity of ligase IV in ischemic retinal neurons. These results were confirmed in rat retinas following ischemia/reperfusion injury. Furthermore, we found that unlike lithium chlorine (a well-known direct inhibitor of GSK-3β), SB216763 inhibited GSK-3β activity by suppressing its phosphorylation. Taken together, our results suggest that GSK-3β inhibition enhances repair of DNA double-strand breaks by upregulating ligase IV expression in ischemic retinal neurons. This study was approved by the Institutional Animal Care and Use Committee of Zhongshan Ophthalmic Center on February 18, 2018.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhi-Peng Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Pei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yang Ying
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ke-Ming Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
19
|
Zou XH, Sun LH, Yang W, Li BJ, Cui RJ. Potential role of insulin on the pathogenesis of depression. Cell Prolif 2020; 53:e12806. [PMID: 32281722 PMCID: PMC7260070 DOI: 10.1111/cpr.12806] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/22/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
The regulation of insulin on depression and depression-like behaviour has been widely reported. Insulin and activation of its receptor can promote learning and memory, affect the hypothalamic-pituitary-adrenal axis (HPA) balance, regulate the secretion of neurotrophic factors and neurotransmitters, interact with gastrointestinal microbiome, exert neuroprotective effects and have an impact on depression. However, the role of insulin on depression remains largely unclear. Therefore, in this review, we summarized the potential role of insulin on depression. It may provide new insight for clarifying role of insulin on the pathogenesis of depression.
Collapse
Affiliation(s)
- Xiao Han Zou
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Li Hua Sun
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bing Jin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ran Ji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|