1
|
Thapa R, Moglad E, Afzal M, Gupta G, Bhat AA, Hassan Almalki W, Kazmi I, Alzarea SI, Pant K, Singh TG, Singh SK, Ali H. The role of sirtuin 1 in ageing and neurodegenerative disease: A molecular perspective. Ageing Res Rev 2024; 102:102545. [PMID: 39423873 DOI: 10.1016/j.arr.2024.102545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Sirtuin 1 (SIRT1), an NAD+-dependent deacetylase, has emerged as a key regulator of cellular processes linked to ageing and neurodegeneration. SIRT1 modulates various signalling pathways, including those involved in autophagy, oxidative stress, and mitochondrial function, which are critical in the pathogenesis of neurodegenerative diseases. This review explores the therapeutic potential of SIRT1 in several neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and Amyotrophic Lateral Sclerosis (ALS). Preclinical studies have demonstrated that SIRT1 activators, such as resveratrol, SRT1720, and SRT2104, can alleviate disease symptoms by reducing oxidative stress, enhancing autophagic flux, and promoting neuronal survival. Ongoing clinical trials are evaluating the efficacy of these SIRT1 activators, providing hope for future therapeutic strategies targeting SIRT1 in neurodegenerative diseases. This review explores the role of SIRT1 in ageing and neurodegenerative diseases, with a particular focus on its molecular mechanisms, therapeutic potential, and clinical applications.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf 72341, Saudi Arabia
| | - Kumud Pant
- Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| |
Collapse
|
2
|
Cho YE, Kim J, Vorn R, Cho H, Baek W, Park H, Yun S, Kim HS, Cashion AK, Gill J, Koo BN, Lee H. Extracellular Vesicle MicroRNAs as Predictive Biomarkers in Postoperative Delirium After Spine Surgery: Preliminary Study. J Gerontol A Biol Sci Med Sci 2024; 79:glae162. [PMID: 38970345 PMCID: PMC11398910 DOI: 10.1093/gerona/glae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Indexed: 07/08/2024] Open
Abstract
Postoperative delirium (POD) can cause poor patient outcomes in older adults who undergo surgery. In this study, we tested plasma extracellular vesicle (EV) miRNAs obtained before the delirium event to find predictive POD biomarkers after spine surgery. We recruited patients who are more than 70 years old and have undergone spine surgery. Finally, POD patients (n = 31) were included, with no-POD patients matched in age, sex, medical history, and type of surgery (n = 31). Peripheral blood was collected from patients in the operating room after the operation was completed. EVs were isolated from plasma, and the 798 miRNA expression level from EVs was measured using a NanoString platform. Sixty-two patients were included in the study; all were Korean, 67.7% were females, and the median age was 75 years. Preoperative medical history was not statistically different between no-POD and POD patients except for hypertension and the American Society of Anesthesiologists physical status. From the miRNA profiling, we identified 142 significantly differentially expressed miRNAs in POD patients compared with no-POD patients, which are associated with psychological/neurological disorders. The top 10 differentially expressed miRNAs including miR-548ar-5p and miR-627-5p were all upregulated in POD patients and the results were validated using qRT-PCR from the independent sets of samples (n = 96). We demonstrated the potential of plasma EV-miRNAs as predictive biomarkers to identify the risk group of POD after spine surgery. It also provides opportunities for future studies investigating the role of EV-miRNAs in delirium pathology.
Collapse
Affiliation(s)
- Young-Eun Cho
- College of Nursing, The University of Iowa, Iowa City, Iowa, USA
| | - Jeongmin Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Rany Vorn
- School of Nursing, Johns Hopkins University, Baltimore, Maryland, USA
- National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Hyeonmi Cho
- Mo-Im Kim Nursing Research Institute, College of Nursing, Yonsei University, Seoul, Republic of Korea
| | - Wonhee Baek
- College of Nursing, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Hyunki Park
- Mo-Im Kim Nursing Research Institute, College of Nursing, Yonsei University, Seoul, Republic of Korea
| | - Sijung Yun
- Predictiv Care, Inc, Sunnyvale, California, USA
| | - Hyung-Suk Kim
- National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Ann K Cashion
- College of Nursing, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jessica Gill
- School of Nursing, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, Anesthesia Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyangkyu Lee
- Mo-Im Kim Nursing Research Institute, College of Nursing, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Zhou JZ, Deng J, Luo DX, Mai JW, Wu JY, Duan YJ, Dong B, Xin WJ, Xu T, Wei JY. Sex differences in functional and structural alterations of hippocampus region in chronic pain: a DTI and resting-state fMRI study. Front Neurosci 2024; 18:1428666. [PMID: 39308951 PMCID: PMC11412943 DOI: 10.3389/fnins.2024.1428666] [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: 05/06/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction It is well known that there are significant differences in the prevalence of chronic pain between males and females. Human and animal imaging studies have shown that chronic pain profoundly alters the structure and function of brain regions. However, there is limited research on the sex-specific mechanisms underlying the brain plasticity and adaptive changes associated with chronic pain. In this article, we conducted a multimodal study to evaluate how nerve injury-induced chronic pain affects the brain. Methods Male and female Sprague-Dawley (SD) rats with spared nerve injury (SNI) model underwent resting-state functional magnetic resonance imaging (rs-fMRI) (male sham group: n = 18; male SNI group: n = 18; female sham group: n = 20; female SNI group: n = 18) and magnetic resonance diffusion tensor imaging (DTI) (male sham group: n = 23; male SNI group: n = 21; female sham group: n = 20; female SNI group: n = 21) scanning. ICA method, Fractional amplitude of low-frequency fluctuations (fALFF), immunofluorescence staining, and graph theory analysis was utilized to extract the rs-fMRI changes of brain regions of each group. Results Using SNI model, which promotes long-lasting mechanical allodynia, we found that neuropathic pain deeply modified the intrinsic organization of the brain functional network in male and female rats (main effect of operation: F = 298.449, P < 0.001). 64 independent components (ICs) in the brain were divided and assigned to 16 systems. In male rats, we observed significant alterations in the microstructure of the hippocampal cornu ammonis 1 and cornu ammonis 2 (CA1/CA2) region, as indicated by increased mean diffusivity (MD) (CA1_L: P = 0.02; CA1_R: P = 0.031; CA2_L: P = 0.035; CA2_R: P = 0.015) and radial diffusivity (RD) (CA1_L: P = 0.028; CA1_R: P = 0.033; CA2_L: P = 0.037; CA2_R: P = 0.038) values, along with enhanced activating transcription factor 3 (ATF3) expression. Conversely, in female rats, we found significant increases in the fractional amplitude of low frequency fluctuations (fALFF) value within the hippocampal dentate gyrus (DG) (F = 5.419, P = 0.023), accompanied by elevated c-Fos signal (F = 6.269, P = 0.031). Furthermore, graph theory analysis revealed notable differences in the small-world network of the hippocampal system in female rats, characterized by reduced small-world attributes and increased inter-nodal transmission efficiency. Discussion Our study indicates sex differences in structural and functional alterations in the hippocampal system in rats under chronic pain conditions. The results suggest that the hippocampus system plays an important role in the different mechanisms of chronic pain in different sexes. These findings provide reliable insights to explore the complex mechanisms underlying sex differences in chronic pain.
Collapse
Affiliation(s)
- Jun-Zhi Zhou
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
| | - Jie Deng
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - De-Xing Luo
- Department of Anesthesiology, Huizhou Central People’s Hospital, Huizhou, China
| | - Jing-Wen Mai
- Department of Anesthesiology, Huizhou Central People’s Hospital, Huizhou, China
| | - Jia-Yan Wu
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
| | - Yu-Juan Duan
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
| | - Bo Dong
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
| | - Wen-Jun Xin
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Ting Xu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Jia-You Wei
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Zhuhai, China
| |
Collapse
|
4
|
Sidorina A, Catesini G, Sacchetti E, Rizzo C, Dionisi-Vici C. Propionic Acidemia, Methylmalonic Acidemia, and Cobalamin C Deficiency: Comparison of Untargeted Metabolomic Profiles. Metabolites 2024; 14:428. [PMID: 39195524 DOI: 10.3390/metabo14080428] [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: 06/07/2024] [Revised: 07/12/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Methylmalonic acidemia (MMA), propionic acidemia (PA), and cobalamin C deficiency (cblC) share a defect in propionic acid metabolism. In addition, cblC is also involved in the process of homocysteine remethylation. These three diseases produce various phenotypes and complex downstream metabolic effects. In this study, we used an untargeted metabolomics approach to investigate the biochemical differences and the possible connections among the pathophysiology of each disease. The significantly changed metabolites in the untargeted urine metabolomic profiles of 21 patients (seven MMA, seven PA, seven cblC) were identified through statistical analysis (p < 0.05; log2FC > |1|) and then used for annotation. Annotated features were associated with different metabolic pathways potentially involved in the disease's development. Comparative statistics showed markedly different metabolomic profiles between MMA, PA, and cblC, highlighting the characteristic species for each disease. The most affected pathways were related to the metabolism of organic acids (all diseases), amino acids (all diseases), and glycine and its conjugates (in PA); the transsulfuration pathway; oxidative processes; and neurosteroid hormones (in cblC). The untargeted metabolomics study highlighted the presence of significant differences between the three diseases, pointing to the most relevant contrast in the cblC profile compared to MMA and PA. Some new biomarkers were proposed for PA, while novel data regarding the alterations of steroid hormone profiles and biomarkers of oxidative stress were obtained for cblC disease. The elevation of neurosteroids in cblC may indicate a potential connection with the development of ocular and neuronal deterioration.
Collapse
Affiliation(s)
- Anna Sidorina
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Giulio Catesini
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Elisa Sacchetti
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Cristiano Rizzo
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Carlo Dionisi-Vici
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| |
Collapse
|
5
|
Liu M, Zhao J, Xue C, Yang J, Ying L. Uncovering the ferroptosis related mechanism of laduviglusib in the cell-type-specific targets of the striatum in Huntington's disease. BMC Genomics 2024; 25:633. [PMID: 38918688 PMCID: PMC11197352 DOI: 10.1186/s12864-024-10534-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder featured by abnormal movements, arising from the extensive neuronal loss and glial dysfunction in the striatum. Although the causes and pathogenetic mechanisms of HD are well established, the development of disease-modifying pharmacological therapies for HD remains a formidable challenge. Laduviglusib has demonstrated neuroprotective effects through the enhancement of mitochondrial function in the striatum of HD animal models. Ferroptosis is a nonapoptotic form of cell death that occurs as a consequence of lethal iron-dependent lipid peroxidation and mitochondrial dysfunction. However, the ferroptosis-related mechanisms underlying the neuroprotective effects of laduviglusib in the striatum of HD patients remain largely uncharted. In this study, we leveraged single-nucleus RNA sequencing data obtained from the striatum of HD patients in stages 2-4 to identify differentially expressed genes within distinct cell-type. We subsequently integrated these differentially expressed genes of HD, laduviglusib target genes and ferroptosis-related genes to predict the ferroptosis-related mechanisms underpinning the neuroprotective effects of laduviglusib in HD patients. The Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses unveiled that the effects of laduviglusib on direct pathway striatal projection neurons (dSPNs) is mainly associated with Th17 cell differentiation pathways. Conversely, its impact on indirect pathway striatal projection neurons (iSPNs) extends to the Neurotrophin signaling pathway, FoxO signaling pathway, and reactive oxygen species pathway. In microglia, laduviglusib appears to contribute to HD pathology via mechanisms related to Th17 cell differentiation and the FoxO signaling pathway. Further, molecular docking results indicated favorable binding of laduviglusib with PARP1 (associated with dSPNs and iSPNs), SCD (associated with astrocytes), ALOX5 (associated with microglia), and HIF1A (associated with dSPNs, iSPNs, and microglia). In addition, the KEGG results suggest that laduviglusib may enhance mitochondrial function and protect against neuronal loss by targeting ferroptosis-related signaling pathways, particularly mediated by ALOX5 in microglia. These findings provide valuable insights into the potential mechanisms through which laduviglusib exerts its effects on distinct cell-types within the HD striatum.
Collapse
Affiliation(s)
- Mei Liu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jinlan Zhao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, School of Pharmaceutical Science, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chengcheng Xue
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jun Yang
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Li Ying
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
6
|
Vassal M, Martins F, Monteiro B, Tambaro S, Martinez-Murillo R, Rebelo S. Emerging Pro-neurogenic Therapeutic Strategies for Neurodegenerative Diseases: A Review of Pre-clinical and Clinical Research. Mol Neurobiol 2024:10.1007/s12035-024-04246-w. [PMID: 38816676 DOI: 10.1007/s12035-024-04246-w] [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: 01/03/2024] [Accepted: 05/14/2024] [Indexed: 06/01/2024]
Abstract
The neuroscience community has largely accepted the notion that functional neurons can be generated from neural stem cells in the adult brain, especially in two brain regions: the subventricular zone of the lateral ventricles and the subgranular zone in the dentate gyrus of the hippocampus. However, impaired neurogenesis has been observed in some neurodegenerative diseases, particularly in Alzheimer's, Parkinson's, and Huntington's diseases, and also in Lewy Body dementia. Therefore, restoration of neurogenic function in neurodegenerative diseases emerges as a potential therapeutic strategy to counteract, or at least delay, disease progression. Considering this, the present study summarizes the different neuronal niches, provides a collection of the therapeutic potential of different pro-neurogenic strategies in pre-clinical and clinical research, providing details about their possible modes of action, to guide future research and clinical practice.
Collapse
Affiliation(s)
- Mariana Vassal
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Filipa Martins
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Bruno Monteiro
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Huddinge, Sweden
| | - Ricardo Martinez-Murillo
- Neurovascular Research Group, Department of Translational Neurobiology, Cajal Institute (CSIC), Madrid, Spain
| | - Sandra Rebelo
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
7
|
Tsamou M, Roggen EL. Sex-associated microRNAs potentially implicated in sporadic Alzheimer's disease (sAD). Brain Res 2024; 1829:148791. [PMID: 38307153 DOI: 10.1016/j.brainres.2024.148791] [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: 09/22/2023] [Revised: 01/08/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND The onset and pathology of sporadic Alzheimer's disease (sAD) seem to be affected by both sex and genetic mechanisms. Evidence supports that the high prevalence of sAD in women, worldwide, may be attributed to an interplay among aging, sex, and lifestyle, influenced by genetics, metabolic changes, and hormones. Interestingly, epigenetic mechanisms such as microRNAs (miRNAs), known as master regulators of gene expression, may contribute to this observed sexual dimorphism in sAD. OBJECTIVES To investigate the potential impact of sex-associated miRNAs on processes manifesting sAD pathology, as described by the Tau-driven Adverse Outcome Pathway (AOP) leading to memory loss. METHODS Using publicly available human miRNA datasets, sex-biased miRNAs, defined as differentially expressed by sex in tissues possibly affected by sAD pathology, were collected. In addition, sex hormone-related miRNAs were also retrieved from the literature. The compiled sex-biased and sex hormone-related miRNAs were further plugged into the dysregulated processes of the Tau-driven AOP for memory loss. RESULTS Several miRNAs, previously identified as sex-associated, were implicated in dysregulated processes associated with the manifestation of sAD pathology. Importantly, the described pathology processes were not confined to a particular sex. A mechanistic-based approach utilizing miRNAs was adopted in order to elucidate the link between sex and biological processes potentially involved in the development of memory loss. CONCLUSIONS The identification of sex-associated miRNAs involved in the early processes manifesting memory loss may shed light to the complex molecular mechanisms underlying sAD pathogenesis in a sex-specific manner.
Collapse
Affiliation(s)
- Maria Tsamou
- ToxGenSolutions (TGS), Oxfordlaan 70, 6229EV Maastricht, The Netherlands.
| | - Erwin L Roggen
- ToxGenSolutions (TGS), Oxfordlaan 70, 6229EV Maastricht, The Netherlands
| |
Collapse
|
8
|
Chiang MK, Lin TC, Lin KH, Chang YC, Hsieh-Li HM, Lai DM. Hyperbaric Oxygen Therapy Attenuated the Motor Coordination and Cognitive Impairment of Polyglutamine Spinocerebellar Ataxia SCA17 Mice. CEREBELLUM (LONDON, ENGLAND) 2024; 23:401-417. [PMID: 36943575 DOI: 10.1007/s12311-023-01548-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] [Accepted: 03/14/2023] [Indexed: 03/23/2023]
Abstract
Spinocerebellar ataxias (SCAs) are a large and diverse group of autosomal-dominant neurodegenerative diseases. No drugs have been approved for these relentlessly progressive and fatal SCAs. Our previous studies indicate that oxidative stress, neuroinflammation, and neuronal apoptosis are elevated in the SCA17 mice, which are the main therapeutic targets of hyperbaric oxygen treatment (HBOT). HBOT is considered to be an alternative and less invasive therapy for SCAs. In this study, we evaluated the HBOT (2.2 ATA for 14 days) effect and the persistence for the management of SCA17 mice and their wild-type littermates. We found HBOT attenuated the motor coordination and cognitive impairment of SCA17 mice and which persisted for about 1 month after the treatment. The results of several biochemistry and liver/kidney hematoxylin and eosin staining show the HBOT condition has no obvious toxicity in the mice. Immunostaining analyses show that the neuroprotective effect of HBOT could be through the promotion of BDNF production and the amelioration of neuroinflammation. Surprisingly, HBOT executes different effects on the male and female SCA17 mice, including the reduction of neuroinflammation and activation of CaMKII and ERK. This study suggests HBOT is a potential alternative therapeutic treatment for SCA17. Accumulated findings have revealed the similarity in disease pathomechanisms and possible therapeutic strategies in polyQ diseases; therefore, HBOT could be an optional treatment as well as the other polyQ diseases.
Collapse
Affiliation(s)
- Meng-Ke Chiang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ta-Chun Lin
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | | | - Ya-Chin Chang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan.
| | - Dar-Ming Lai
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
9
|
Esperante IJ, Meyer M, Banzan C, Kruse MS, Lima A, Roig P, Guennoun R, Schumacher M, De Nicola AF, Gonzalez Deniselle MC. Testosterone Reduces Myelin Abnormalities in the Wobbler Mouse Model of Amyotrophic Lateral Sclerosis. Biomolecules 2024; 14:428. [PMID: 38672445 PMCID: PMC11048492 DOI: 10.3390/biom14040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motoneuron degenerative disease that is associated with demyelination. The Wobbler (WR) mouse exhibits motoneuron degeneration, gliosis and myelin deterioration in the cervical spinal cord. Since male WRs display low testosterone (T) levels in the nervous system, we investigated if T modified myelin-relative parameters in WRs in the absence or presence of the aromatase inhibitor, anastrozole (A). We studied myelin by using luxol-fast-blue (LFB) staining, semithin sections, electron microscopy and myelin protein expression, density of IBA1+ microglia and mRNA expression of inflammatory factors, and the glutamatergic parameters glutamine synthetase (GS) and the transporter GLT1. Controls and WR + T showed higher LFB, MBP and PLP staining, lower g-ratios and compact myelin than WRs and WR + T + A, and groups showing the rupture of myelin lamellae. WRs showed increased IBA1+ cells and mRNA for CD11b and inflammatory factors (IL-18, TLR4, TNFαR1 and P2Y12R) vs. controls or WR + T. IBA1+ cells, and CD11b were not reduced in WR + T + A, but inflammatory factors' mRNA remained low. A reduction of GS+ cells and GLT-1 immunoreactivity was observed in WRs and WR + T + A vs. controls and WR + T. Clinically, WR + T but not WR + T + A showed enhanced muscle mass, grip strength and reduced paw abnormalities. Therefore, T effects involve myelin protection, a finding of potential clinical translation.
Collapse
Affiliation(s)
- Ivan J. Esperante
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Maria Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Carolina Banzan
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Maria Sol Kruse
- Laboratory of Neurobiology, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina;
| | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Paulina Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
| | - Rachida Guennoun
- U1195 INSERM and University Paris Sud: “Neuroprotective, Neuroregenerative and Remyelinating Small Molecules”, 94276 Kremlin-Bicêtre, France; (R.G.); (M.S.)
| | - Michael Schumacher
- U1195 INSERM and University Paris Sud: “Neuroprotective, Neuroregenerative and Remyelinating Small Molecules”, 94276 Kremlin-Bicêtre, France; (R.G.); (M.S.)
| | - Alejandro F. De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires 1121, Argentina
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, CONICET, Buenos Aires 1428, Argentina; (I.J.E.); (M.M.); (C.B.); (A.F.D.N.)
- Departamento de Ciencias Fisiológicas, UA1, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires 1121, Argentina
| |
Collapse
|
10
|
Zhang Y, Chen M, Chen H, Mi S, Wang C, Zuo H, Song L, Du J, Cui H, Li S. Testosterone reduces hippocampal synaptic damage in an androgen receptor-independent manner. J Endocrinol 2024; 260:e230114. [PMID: 37991884 PMCID: PMC10762536 DOI: 10.1530/joe-23-0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 11/22/2023] [Indexed: 11/24/2023]
Abstract
Aging-related reduction in androgen levels may be a possible risk factor for neurodegenerative diseases and contribute to cognitive impairment. Androgens may affect synaptic function and cognition in an androgen receptor (AR)-independent manner; however, the mechanisms connecting theses effects are unknown. Therefore, we used testicular feminization mutation (Tfm) male mice, a model with AR mutation, to test the effects of testosterone on synaptic function and cognition. Our results showed that testosterone ameliorated spatial memory deficit and neuronal damage, and increased dendritic spines density and postsynaptic density protein 95 (PSD95) and glutamate receptor 1 (GluA1) expression in the hippocampus of Tfm male mice. And these effects of testosterone were not inhibited by anastrozole, which suppressed conversion of testosterone to estradiol. Mechanistically, testosterone activated the extracellular signal-related kinase 1/2 (Erk1/2) and cyclic adenosine monophosphate response element-binding protein (CREB) in the hippocampus of Tfm male mice. Meanwhile, Erk1/2 inhibitor SCH772984 blocked the upregulation of phospho-CREB, PSD95, and GluA1 induced by testosterone in HT22 cells pretreated with flutamide, an androgen antagonist. Collectively, our data indicate that testosterone may ameliorate hippocampal synaptic damage and spatial memory deficit by activating the Erk1/2-CREB signaling pathway in an AR-independent manner.
Collapse
Affiliation(s)
- Yizhou Zhang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Meiqin Chen
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huan Chen
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shixiong Mi
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chang Wang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hongchun Zuo
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Leigang Song
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Juan Du
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huixian Cui
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sha Li
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
11
|
Bourque M, Morissette M, Di Paolo T. Neuroactive steroids and Parkinson's disease: Review of human and animal studies. Neurosci Biobehav Rev 2024; 156:105479. [PMID: 38007170 DOI: 10.1016/j.neubiorev.2023.105479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/13/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The greater prevalence and incidence of Parkinson's disease (PD) in men suggest a beneficial effect of sex hormones. Neuroactive steroids have neuroprotective activities thus offering interesting option for disease-modifying therapy for PD. Neuroactive steroids are also neuromodulators of neurotransmitter systems and may thus help to control PD symptoms and side effect of dopamine medication. Here, we review the effect on sex hormones (estrogen, androgen, progesterone and its metabolites) as well as androstenediol, pregnenolone and dehydroepiandrosterone) in human studies and in animal models of PD. The effect of neuroactive steroids is reviewed by considering sex and hormonal status to help identify specifically for women and men with PD what might be a preventive approach or a symptomatic treatment. PD is a complex disease and the pathogenesis likely involves multiple cellular processes. Thus it might be useful to target different cellular mechanisms that contribute to neuronal loss and neuroactive steroids provide therapeutics options as they have multiple mechanisms of action.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, 2705, Boulevard Laurier, Québec G1V4G2, Canada
| | - Marc Morissette
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, 2705, Boulevard Laurier, Québec G1V4G2, Canada
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, 2705, Boulevard Laurier, Québec G1V4G2, Canada; Faculté de pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec G1V 0A6, Canada.
| |
Collapse
|
12
|
Sayfullaeva J, McLoughlin J, Kwakowsky A. Hormone Replacement Therapy and Alzheimer's Disease: Current State of Knowledge and Implications for Clinical Use. J Alzheimers Dis 2024; 101:S235-S261. [PMID: 39422965 DOI: 10.3233/jad-240899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder responsible for over half of dementia cases, with two-thirds being women. Growing evidence from preclinical and clinical studies underscores the significance of sex-specific biological mechanisms in shaping AD risk. While older age is the greatest risk factor for AD, other distinct biological mechanisms increase the risk and progression of AD in women including sex hormones, brain structural differences, genetic background, immunomodulation and vascular disorders. Research indicates a correlation between declining estrogen levels during menopause and an increased risk of developing AD, highlighting a possible link with AD pathogenesis. The neuroprotective effects of estrogen vary with the age of treatment initiation, menopause stage, and type. This review assesses clinical and observational studies conducted in women, examining the influence of estrogen on cognitive function or addressing the ongoing question regarding the potential use of hormone replacement therapy (HRT) as a preventive or therapeutic option for AD. This review covers recent literature and discusses the working hypothesis, current use, controversies and challenges regarding HRT in preventing and treating age-related cognitive decline and AD. The available evidence indicates that estrogen plays a significant role in influencing dementia risk, with studies demonstrating both beneficial and detrimental effects of HRT. Recommendations regarding HRT usage should carefully consider the age when the hormonal supplementation is initiated, baseline characteristics such as genotype and cardiovascular health, and treatment duration until this approach can be more thoroughly investigated or progress in the development of alternative treatments can be made.
Collapse
Affiliation(s)
- Jessica Sayfullaeva
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - John McLoughlin
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| |
Collapse
|
13
|
Nguyen Hoai B, Hoang L, Nguyen Cao T, Pham Minh Q, A Jannini E. Testosterone and aging male, a perspective from a developing country. Aging Male 2023; 26:2223712. [PMID: 37335039 DOI: 10.1080/13685538.2023.2223712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023] Open
Abstract
PURPOSE Hypogonadism is associated with a wide range of physical and psychological symptoms that can affect the overall health of men. However, in a developing country, there are several imposing challenges in the diagnosis and treatment of hypogonadism, including a lack of awareness and understanding of the condition among healthcare providers and patients, limited resources and the high cost of treatment. This review aimed to examine the potential benefits and risks of testosterone replacement therapy (TRT) and provides a perspective of a developing country on the topic. MATERIALS AND METHODS A comprehensive literature review was conducted to gather relevant information on the impact of testosterone deficiency on ageing males and the effectiveness of TRT for treating hypogonadism. Published peer-reviewed articles were analyzed to evaluate the benefits and risks of TRT. Additionally, the unique challenges faced in the diagnosis and treatment of hypogonadism in a developing country were considered. RESULTS Testosterone replacement therapy has been shown to be an effective treatment for hypogonadism, particularly in symptomatic men with low testosterone levels. It offers potential benefits such as improvements in symptoms and overall quality of life. However, there are associated risks and side effects that need to be considered. In a developing country, challenges such as limited awareness and understanding of hypogonadism, resource constraints, and high treatment costs pose additional barriers to accessing TRT and comprehensive care. CONCLUSION In conclusion, TRT holds promise as a treatment for hypogonadism, but its implementation and accessibility face significant challenges in a developing country. Addressing these challenges, including raising awareness, allocating resources, and finding cost-effective solutions, is crucial for ensuring that men with hypogonadism in such settings receive appropriate diagnosis and treatment. Further research and efforts are needed to improve the management of hypogonadism in developing countries and optimize the potential benefits of TRT for affected individuals.
Collapse
Affiliation(s)
- Bac Nguyen Hoai
- Department of Andrology and Sexual Medicine, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Long Hoang
- Department of Urology, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Thang Nguyen Cao
- Department of Andrology and Sexual Medicine, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Quan Pham Minh
- Department of Andrology and Sexual Medicine, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Emmanuele A Jannini
- Chair of Endocrinology and Sexual Medicine (ENDOSEX), University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
14
|
Tan TH, Li SW, Chang CW, Chen YC, Liu YH, Ma JT, Chang CP, Liao PC. Rat Hair Metabolomics Analysis Reveals Perturbations of Unsaturated Fatty Acid Biosynthesis, Phenylalanine, and Arachidonic Acid Metabolism Pathways Are Associated with Amyloid-β-Induced Cognitive Deficits. Mol Neurobiol 2023; 60:4373-4395. [PMID: 37095368 PMCID: PMC10293421 DOI: 10.1007/s12035-023-03343-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 04/04/2023] [Indexed: 04/26/2023]
Abstract
Hair is a noninvasive valuable biospecimen for the long-term assessment of endogenous metabolic disturbance. Whether the hair is suitable for identifying biomarkers of the Alzheimer's disease (AD) process remains unknown. We aim to investigate the metabolism changes in hair after β-amyloid (Aβ1-42) exposure in rats using ultra-high-performance liquid chromatography-high-resolution mass spectrometry-based untargeted and targeted methods. Thirty-five days after Aβ1-42 induction, rats displayed significant cognitive deficits, and forty metabolites were changed, of which twenty belonged to three perturbed pathways: (1) phenylalanine metabolism and phenylalanine, tyrosine, and tryptophan biosynthesis-L-phenylalanine, phenylpyruvate, ortho-hydroxyphenylacetic acid, and phenyllactic acid are up-regulated; (2) arachidonic acid (ARA) metabolism-leukotriene B4 (LTB4), arachidonyl carnitine, and 5(S)-HPETE are upregulation, but ARA, 14,15-DiHETrE, 5(S)-HETE, and PGB2 are opposite; and (3) unsaturated fatty acid biosynthesis- eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), FA 18:3 + 1O, and FA 18:3 + 2O are downregulated. Linoleic acid metabolism belonging to the biosynthesis of unsaturated fatty acid includes the upregulation of 8-hydroxy-9,10-epoxystearic acid, 13-oxoODE, and FA 18:2 + 4O, and downregulation of 9(S)-HPODE and dihomo-γ-linolenic acid. In addition, cortisone and dehydroepiandrosterone belonging to steroid hormone biosynthesis are upregulated. These three perturbed metabolic pathways also correlate with cognitive impairment after Aβ1-42 stimulation. Furthermore, ARA, DHA, EPA, L-phenylalanine, and cortisone have been previously implicated in the cerebrospinal fluid of AD patients and show a similar changing trend in Aβ1-42 rats' hair. These data suggest hair can be a useful biospecimen that well reflects the expression of non-polar molecules under Aβ1-42 stimulation, and the five metabolites have the potential to serve as novel AD biomarkers.
Collapse
Affiliation(s)
- Tian-Hoe Tan
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, 710, Taiwan
- Department of Senior Services, Southern Taiwan University of Science and Technology, No.1, Nantai St., Yungkang Dist., Tainan, 710, Taiwan
| | - Shih-Wen Li
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Chih-Wei Chang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Yuan-Chih Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Yu-Hsuan Liu
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Jui-Ti Ma
- Department of Medical Research, Chi Mei Medical Center, No. 901, Zhonghua Rd., Yongkang Dist., Tainan, 710, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, No. 901, Zhonghua Rd., Yongkang Dist., Tainan, 710, Taiwan.
| | - Pao-Chi Liao
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan.
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
15
|
Kang J, Yan J, Yan W. Testosterone ameliorated the behavioural deficits of gonadectomised rats and counteracted free radicals in a dosage-dependent manner. Behav Brain Res 2023; 450:114501. [PMID: 37207980 DOI: 10.1016/j.bbr.2023.114501] [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: 01/27/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 05/21/2023]
Abstract
Testosterone deficiency may induce behavioural changes in individuals. Oxidative stress resulting from a redox imbalance may be implicated in the initiation and progression of neurobehavioural disorders. However, whether exogenous testosterone intervention in male gonadectomised (GDX) rats ameliorates oxidative stress and plays a neuroprotective role remains unknown. Therefore, we examined this hypothesis by performing sham or gonadectomy surgeries on Sprague-Dawley rats with or without supplementation with different doses of testosterone propionate (TP). Open field and Morris water maze tests were performed, the serum and brain testosterone levels, and oxidative stress markers were analysed. GDX and lower TP doses (0.5mg/kg) induced reduced exploratory and motor behaviours, but impaired spatial learning and memory compared to Sham rats. Administration of physiological TP levels (0.75-1.25mg/kg) to the GDX rats restored the behaviour observed in the intact rats. However, higher TP doses (1.5-3.0mg/kg) induced increased exploratory and motor behaviours but impaired spatial learning and memory. These behavioural impairments were accompanied by a marked decrease in levels of antioxidant enzymes (superoxide dismutase and catalase) and an increase in lipid peroxidation levels in the substantia nigra and hippocampus. These findings indicate that TP administration can alter behavioural performance and induce memory and learning impairment, which may result from changes in redox homeostasis in male GDX animals.
Collapse
Affiliation(s)
- Jie Kang
- Affiliated Yiling Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Jixing Yan
- College of Rehabilitation Sciences, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Wensheng Yan
- Department of Sports Medicine, Hebei Sport University, Shijiazhuang, PR China.
| |
Collapse
|
16
|
Duarte-Pereira S, Matos S, Oliveira JL, Silva RM. Study of NAD-interacting proteins highlights the extent of NAD regulatory roles in the cell and its potential as a therapeutic target. J Integr Bioinform 2023:jib-2022-0049. [PMID: 36880517 PMCID: PMC10389049 DOI: 10.1515/jib-2022-0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) levels are essential for the normal physiology of the cell and are strictly regulated to prevent pathological conditions. NAD functions as a coenzyme in redox reactions, as a substrate of regulatory proteins, and as a mediator of protein-protein interactions. The main objectives of this study were to identify the NAD-binding and NAD-interacting proteins, and to uncover novel proteins and functions that could be regulated by this metabolite. It was considered if cancer-associated proteins were potential therapeutic targets. Using multiple experimental databases, we defined datasets of proteins that directly interact with NAD - the NAD-binding proteins (NADBPs) dataset - and of proteins that interact with NADBPs - the NAD-protein-protein interactions (NAD-PPIs) dataset. Pathway enrichment analysis revealed that NADBPs participate in several metabolic pathways, while NAD-PPIs are mostly involved in signalling pathways. These include disease-related pathways, namely, three major neurodegenerative disorders: Alzheimer's disease, Huntington's disease, and Parkinson's disease. Then, the complete human proteome was further analysed to select potential NADBPs. TRPC3 and isoforms of diacylglycerol (DAG) kinases, which are involved in calcium signalling, were identified as new NADBPs. Potential therapeutic targets that interact with NAD were identified, that have regulatory and signalling functions in cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sara Duarte-Pereira
- IEETA/DETI, University of Aveiro, Aveiro, Portugal.,Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Sérgio Matos
- IEETA/DETI, University of Aveiro, Aveiro, Portugal.,LASI - Intelligent Systems Associate Laboratory, Guimarães, Portugal
| | - José Luís Oliveira
- IEETA/DETI, University of Aveiro, Aveiro, Portugal.,LASI - Intelligent Systems Associate Laboratory, Guimarães, Portugal
| | - Raquel M Silva
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Aveiro, Portugal.,Universidade Católica Portuguesa, Faculty of Dental Medicine, Center for Interdisciplinary Research in Health (CIIS), Viseu, Portugal
| |
Collapse
|
17
|
Fucic A, Mantovani A, Vena J, Bloom MS, Sincic N, Vazquez M, Aguado-Sierra J. Impact of endocrine disruptors from mother's diet on immuno-hormonal orchestration of brain development and introduction of the virtual human twin tool. Reprod Toxicol 2023; 117:108357. [PMID: 36863570 DOI: 10.1016/j.reprotox.2023.108357] [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: 12/21/2022] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023]
Abstract
Diet has long been known to modify physiology during development and adulthood. However, due to a growing number of manufactured contaminants and additives over the last few decades, diet has increasingly become a source of exposure to chemicals that has been associated with adverse health risks. Sources of food contaminants include the environment, crops treated with agrochemicals, inappropriate storage (e.g., mycotoxins) and migration of xenobiotics from food packaging and food production equipment. Hence, consumers are exposed to a mixture of xenobiotics, some of which are endocrine disruptors (EDs). The complex interactions between immune function and brain development and their orchestration by steroid hormones are insufficiently understood in human populations, and little is known about the impact on immune-brain interactions by transplacental fetal exposure to EDs via maternal diet. To help to identify the key data gaps, this paper aims to present (a) how transplacental EDs modify immune system and brain development, and (b) how these mechanisms may correlate with diseases such as autism and disturbances of lateral brain development. Attention is given to disturbances of the subplate, a transient structure of crucial significance in brain development. Additionally, we describe cutting edge approaches to investigate the developmental neurotoxicity of EDs, such as the application of artificial intelligence and comprehensive modelling. In the future, highly complex investigations will be performed using virtual brain models constructed using sophisticated multi-physics/multi-scale modelling strategies based on patient and synthetic data, which will enable a greater understanding of healthy or disturbed brain development.
Collapse
Affiliation(s)
- A Fucic
- Institute for Medical Research and Occupational Health, Ksaverska C 2, Zagreb, Croatia.
| | - A Mantovani
- Istituto Superiore di Sanità, Department of Food Safety, Nutrition and Veterinary Public Health, Rome, Italy
| | - J Vena
- Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - M S Bloom
- Global and Community Health, George Mason University, 4400 University Dr., Fairfax, VA, USA
| | - N Sincic
- Medical School, University of Zagreb, Salata 3, Croatia
| | - M Vazquez
- Barcelona Supercomputing Center, Plaça Eusebi Güell, 1-3, Barcelona 08034, Spain
| | - J Aguado-Sierra
- Barcelona Supercomputing Center, Plaça Eusebi Güell, 1-3, Barcelona 08034, Spain
| |
Collapse
|
18
|
Androgens and NGF Mediate the Neurite-Outgrowth through Inactivation of RhoA. Cells 2023; 12:cells12030373. [PMID: 36766714 PMCID: PMC9913450 DOI: 10.3390/cells12030373] [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: 12/01/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Steroid hormones and growth factors control neuritogenesis through their cognate receptors under physiological and pathological conditions. We have already shown that nerve growth factor and androgens induce neurite outgrowth of PC12 cells through a reciprocal crosstalk between the NGF receptor, TrkA and the androgen receptor. Here, we report that androgens or NGF induce neuritogenesis in PC12 cells through inactivation of RhoA. Ectopic expression of the dominant negative RhoA N19 promotes, indeed, the neurite-elongation of unchallenged and androgen- or NGF-challenged PC12 cells and the increase in the expression levels of βIII tubulin, a specific neuronal marker. Pharmacological inhibition of the Ser/Thr kinase ROCK, an RhoA effector, induces neuritogenesis in unchallenged PC12 cells, and potentiates the effect of androgens and NGF, confirming the role of RhoA/ROCK axis in the neuritogenesis induced by androgen and NGF, through the phosphorylation of Akt. These findings suggest that therapies based on new selective androgen receptor modulators and/or RhoA/ROCK inhibitors might exert beneficial effects in the treatment of neuro-disorders, neurological diseases and ageing-related processes.
Collapse
|
19
|
Verma S, Kushwaha PP, Shankar E, Ponsky LE, Gupta S. Increased cytokine gene expression and cognition risk associated with androgen deprivation therapy. Prostate 2022; 82:1389-1399. [PMID: 35821621 PMCID: PMC9544768 DOI: 10.1002/pros.24411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/25/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) is a standard treatment modality for locally advanced, high-risk, and metastatic hormone-sensitive prostate cancer. Long-term ADT treatment likely develops side-effects that include changes in cognition or onset of dementia. However, the molecular understanding of this effect remains elusive. We attempt to establish a link between ADT and changes in cognitive function using patient databases and bioinformatics analyses. METHODS Gene expression profiling was performed using RNA sequencing data from Alzheimer patient cohort and compared with the data from advanced-stage prostate cancer patients receiving neoadjuvant antiandrogen therapy. Differentially expressed genes (DEGs) were analyzed using the Ingenuity knowledge database. RESULTS A total of 1952 DEGs in the Alzheimer patient cohort and 101 DEGs were identified in ADT treated prostate cancer patients. Comparing both data sets provided a subset of 33 commonly expressed genes involving cytokine-cytokine signaling with an over representation of cytokine-cytokine receptor interaction, inflammatory cytokines, signaling by interleukins together with alterations in the circulating lymphocyte repertoire, adaptive immune responses, regulation of cytokine production, and changes in T-cell subsets. Additionally, lipopolysaccharide, tumor necrosis factor, and toll-like receptors were identified as upstream transcriptional regulators of these pathways. The most commonly expressed genes viz. IL-17A, CCL2, IL-10, IL-6, IL-1RN, LIF/LIFR were further validated by quantitative RT-PCR exhibited higher expression in antiandrogen treated neuronal, glial, and androgen-responsive prostate cancer cells, compared to no-androgen antagonist treatment. CONCLUSIONS Our findings suggest that changes in cytokine signaling under the influence of ADT in prostate cancer patients may be linked with cognitive impairment presenting new avenues for diagnostic and therapeutic development in combating brain deficits.
Collapse
Affiliation(s)
- Shiv Verma
- Department of Urology, School of MedicineCase Western Reserve UniversityClevelandOhioUSA
- The Urology InstituteUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
| | - Prem Prakash Kushwaha
- Department of Urology, School of MedicineCase Western Reserve UniversityClevelandOhioUSA
- The Urology InstituteUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
| | - Eswar Shankar
- Department of Urology, School of MedicineCase Western Reserve UniversityClevelandOhioUSA
- The Urology InstituteUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Present address:
Division of Medical OncologyThe Ohio State UniversityColumbus43210OhioUSA
| | - Lee E. Ponsky
- Department of Urology, School of MedicineCase Western Reserve UniversityClevelandOhioUSA
- The Urology InstituteUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
| | - Sanjay Gupta
- Department of Urology, School of MedicineCase Western Reserve UniversityClevelandOhioUSA
- The Urology InstituteUniversity Hospitals Cleveland Medical CenterClevelandOhioUSA
- Department of PathologyCase Western Reserve UniversityClevelandOhioUSA
- Department of PharmacologyCase Western Reserve UniversityClevelandOhioUSA
- Department of NutritionCase Western Reserve UniversityClevelandOhioUSA
- Division of General Medical SciencesCase Comprehensive Cancer CenterClevelandOhioUSA
- Department of UrologyLouis Stokes Cleveland Veterans Affairs Medical CenterClevelandOhioUSA
| |
Collapse
|
20
|
Marriott RJ, Murray K, Flicker L, Hankey GJ, Matsumoto AM, Dwivedi G, Antonio L, Almeida OP, Bhasin S, Dobs AS, Handelsman DJ, Haring R, O'Neill TW, Ohlsson C, Orwoll ES, Vanderschueren D, Wittert GA, Wu FCW, Yeap BB. Lower serum testosterone concentrations are associated with a higher incidence of dementia in men: The UK Biobank prospective cohort study. Alzheimers Dement 2022; 18:1907-1918. [PMID: 34978125 DOI: 10.1002/alz.12529] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 08/04/2021] [Accepted: 10/14/2021] [Indexed: 01/28/2023]
Abstract
INTRODUCTION The association of testosterone concentrations with dementia risk remains uncertain. We examined associations of serum testosterone and sex hormone-binding globulin (SHBG) with incidence of dementia and Alzheimer's disease. METHODS Serum total testosterone and SHBG were measured by immunoassay. The incidence of dementia and Alzheimer's disease (AD) was recorded. Cox proportional hazards regression was adjusted for age and other variables. RESULTS In 159,411 community-dwelling men (median age 61, followed for 7 years), 826 developed dementia, including 288 from AD. Lower total testosterone was associated with a higher incidence of dementia (overall trend: P = .001, lowest vs highest quintile: hazard ratio [HR] = 1.43, 95% confidence interval [CI] = 1.13-1.81), and AD (P = .017, HR = 1.80, CI = 1.21-2.66). Lower SHBG was associated with a lower incidence of dementia (P < .001, HR = 0.66, CI = 0.51-0.85) and AD (P = .012, HR = 0.53, CI = 0.34-0.84). DISCUSSION Lower total testosterone and higher SHBG are independently associated with incident dementia and AD in older men. Additional research is needed to determine causality.
Collapse
Affiliation(s)
- Ross J Marriott
- School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Kevin Murray
- School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Leon Flicker
- Medical School, University of Western Australia, Perth, Australia.,Western Australian Centre for Healthy Ageing, University of Western Australia, Perth, Australia
| | - Graeme J Hankey
- Medical School, University of Western Australia, Perth, Australia
| | - Alvin M Matsumoto
- Department of Medicine, University of Washington School of Medicine, Seattle, USA.,Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, USA
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, Australia.,Harry Perkins Institute of Medical Research, Fiona Stanley Hospital, Perth, Australia
| | - Leen Antonio
- Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Osvaldo P Almeida
- Medical School, University of Western Australia, Perth, Australia.,Western Australian Centre for Healthy Ageing, University of Western Australia, Perth, Australia
| | - Shalender Bhasin
- Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Adrian S Dobs
- Division of Endocrinology, Johns Hopkins University School of Medicine, Baltimore, USA
| | | | - Robin Haring
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.,European University of Applied Sciences, Faculty of Applied Public Health, Rostock, Germany
| | - Terence W O'Neill
- Centre for Epidemiology Versus Arthritis, University of Manchester and NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Vastra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Dirk Vanderschueren
- Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Gary A Wittert
- Freemasons Centre for Men's Health and Wellbeing, School of Medicine, University of Adelaide, Adelaide, Australia
| | - Frederick C W Wu
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Bu B Yeap
- Medical School, University of Western Australia, Perth, Australia.,Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Australia
| |
Collapse
|
21
|
Proaño B, Casani-Cubel J, Benlloch M, Rodriguez-Mateos A, Navarro-Illana E, Lajara-Romance JM, de la Rubia Ortí JE. Is Dutasteride a Therapeutic Alternative for Amyotrophic Lateral Sclerosis? Biomedicines 2022; 10:biomedicines10092084. [PMID: 36140184 PMCID: PMC9495995 DOI: 10.3390/biomedicines10092084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that is characterized by the loss of upper and lower motor neurons (MNs) in the cerebral cortex, brainstem and spinal cord, with consequent weakness, atrophy and the progressive paralysis of all muscles. There is currently no medical cure, and riluzole and edaravone are the only two known approved drugs for treating this condition. However, they have limited efficacy, and hence there is a need to find new molecules. Dutasteride, a dual inhibitor of type 1 and type 2 5α-reductase (5AR) enzymes, the therapeutic purposes of which, to date, are the treatment of benign prostatic hyperplasia and androgenic alopecia, shows great anti-ALS properties by the molecular-topology methodology. Based on this evidence, this review aims to assess the effects of dutasteride on testosterone (T), progesterone (PROG) and 17β-estradiol (17BE) as a therapeutic alternative for the clinical improvement of ALS, based on the hormonal, metabolic and molecular pathways related to the pathogenesis of the disease. According to the evidence found, dutasteride shows great neuroprotective, antioxidant and anti-inflammatory effects. It also appears effective against glutamate toxicity, and it is capable of restoring altered dopamine activity (DA). These effects are achieved both directly and through steroid hormones. Therefore, dutasteride seems to be a promising molecule for the treatment of ALS, although clinical studies are required for confirmation.
Collapse
Affiliation(s)
- Belén Proaño
- Doctoral Degree School, Health Sciences, Catholic University of Valencia San Vicente Mártir, 46001 Valencia, Spain
| | - Julia Casani-Cubel
- School of Medicine and Health Sciences, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - María Benlloch
- Department Nursing, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, UK
| | | | | | | |
Collapse
|
22
|
Branigan GL, Torrandell‐Haro G, Soto M, Gelmann EP, Vitali F, Rodgers KE, Brinton RD. Androgen-targeting therapeutics mitigate the adverse effect of GnRH agonist on the risk of neurodegenerative disease in men treated for prostate cancer. Cancer Med 2022; 11:2687-2698. [PMID: 35293700 PMCID: PMC9249980 DOI: 10.1002/cam4.4650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/18/2022] [Accepted: 02/23/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Prostate cancer and multiple neurodegenerative diseases (NDD) share an age-associated pattern of onset. Therapy of prostate cancer is known to impact cognitive function. The objective of this study was to determine the impact of multiple classes of androgen-targeting therapeutics (ATT) on the risk of NDD. METHODS A retrospective cohort study of men aged 45 and older with prostate within the US-based Mariner claims data set between January 1 and 27, 2021. A propensity score approach was used to minimize measured and unmeasured selection bias. Disease risk was determined using Kaplan-Meier survival analyses. RESULTS Of the 1,798,648 men with prostate cancer, 209,722 met inclusion criteria. Mean (SD) follow-up was 6.4 (1.8) years. In the propensity score-matched population, exposure to ATT was associated with a minimal increase in NDD incidence (relative risk [RR], 1.07; 95% CI, 1.05-1.10; p < 0.001). However, GnRH agonists alone were associated with significantly increased NDD risk (RR, 1.47; 95% CI, 1.30-1.66; p <0.001). Abiraterone, commonly administered with GnRH agonists and low-dose prednisone, was associated with a significantly decreased risk (RR, 0.77; 95% CI, 0.68-0.87; p < 0.001) of any NDD. CONCLUSIONS Among patients with prostate cancer, GnRH agonist exposure was associated with an increased NDD risk. Abiraterone acetate reduced the risks of Alzheimer's disease and Parkinson's disease conferred by GnRH agonists, whereas the risk for ALS was reduced by androgen receptor inhibitors. Outcomes of these analyses contribute to addressing controversies in the field and indicate that GnRH agonism may be a predictable instigator of risk for NDD with opportunities for risk mitigation in combination with another ATT.
Collapse
Affiliation(s)
- Gregory L. Branigan
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of PharmacologyUniversity of Arizona College of MedicineTucsonArizonaUSA
- Medical Scientist Training ProgramUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Georgina Torrandell‐Haro
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of PharmacologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Maira Soto
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| | - Edward P. Gelmann
- Department of Medicine, Division of Hematology and OncologyUniversity of Arizona College of Medicine and University of Arizona Cancer CenterTucsonArizonaUSA
| | - Francesca Vitali
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of NeurologyUniversity of Arizona College of MedicineTucsonArizonaUSA
- Center for Biomedical Informatics and BiostatisticsUniversity of ArizonaTucsonArizonaUSA
| | - Kathleen E. Rodgers
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of PharmacologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of PharmacologyUniversity of Arizona College of MedicineTucsonArizonaUSA
- Department of NeurologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| |
Collapse
|
23
|
Effects of an Exercise Program Combining Aerobic and Resistance Training on Protein Expressions of Neurotrophic Factors in Obese Rats Injected with Beta-Amyloid. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19137921. [PMID: 35805580 PMCID: PMC9266049 DOI: 10.3390/ijerph19137921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022]
Abstract
In this study, the effects of a 12-week exercise program combining aerobic and resistance training on high-fat diet-induced obese Sprague Dawley (SD) rats after the injection of beta-amyloid into the cerebral ventricle were investigated. Changes in physical fitness, cognitive function, blood levels of beta-amyloid and metabolic factors, and protein expressions of neurotrophic factors related to brain function such as BDNF (brain-derived neurotrophic factor) in the quadriceps femoris, hippocampus, and cerebral cortex were analyzed. The subjects were thirty-two 10-week-old SD rats (DBL Co., Ltd., Seoul, Korea). The rats were randomized into four groups: β-Non-Ex group (n = 8) with induced obesity and βA25-35 injection into the cerebral ventricle through stereotactic biopsy; β-Ex group (n = 8) with induced obesity, βA25-35 injection, and exercise; S-Non-Ex group (n = 8) with an injection of saline in lieu of βA25-35 as the control; and S-Ex group (n = 8) with saline injection and exercise. The 12-week exercise program combined aerobic training and resistance training. As for protein expressions of the factors related to brain function, the combined exercise program was shown to have a clear effect on activating the following factors: PGC-1α (peroxisome proliferator-activated receptor gamma coactivator 1-alpha), FNDC5 (fibronectin type III domain-containing protein 5), and BDNF in the quadriceps femoris; TrkB (Tropomyosin receptor kinase B), FNDC5, and BDNF in the hippocampus; PGC-1α, FNDC5, and BDNF in the cerebral cortex. The protein expression of β-amyloid in the cerebral cortex was significantly lower in the β-Ex group than in the β-Non-Ex group (p < 0.05). The 12-week intervention with the combined exercise program of aerobic and resistance training was shown to improve cardiopulmonary function, muscular endurance, and short-term memory. The results demonstrate a set of positive effects of the combined exercise program, which were presumed to have arisen mainly due to its alleviating effect on β-amyloid plaques, the main cause of reduced brain function, as well as the promotion of protein expressions of PGC-1α, FNDC5, and BDNF in the quadriceps femoris, hippocampus, and cerebral cortex.
Collapse
|
24
|
Strogulski NR, Kopczynski A, de Oliveira VG, Carteri RB, Hansel G, Venturin GT, Greggio S, DaCosta JC, De Bastiani MA, Rodolphi MS, Portela LV. Nandrolone Supplementation Promotes AMPK Activation and Divergent 18[FDG] PET Brain Connectivity in Adult and Aged Mice. Neurochem Res 2022; 47:2032-2042. [PMID: 35415802 DOI: 10.1007/s11064-022-03592-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022]
Abstract
Decreased anabolic androgen levels are followed by impaired brain energy support and sensing with loss of neural connectivity during physiological aging, providing a neurobiological basis for hormone supplementation. Here, we investigated whether nandrolone decanoate (ND) administration mediates hypothalamic AMPK activation and glucose metabolism, thus affecting metabolic connectivity in brain areas of adult and aged mice. Metabolic interconnected brain areas of rodents can be detected by positron emission tomography using 18FDG-mPET. Albino CF1 mice at 3 and 18 months of age were separated into 4 groups that received daily subcutaneous injections of either ND (15 mg/kg) or vehicle for 15 days. At the in vivo baseline and on the 14th day, brain 18FDG-microPET scans were performed. Hypothalamic pAMPKT172/AMPK protein levels were assessed, and basal mitochondrial respiratory states were evaluated in synaptosomes. A metabolic connectivity network between brain areas was estimated based on 18FDG uptake. We found that ND increased the pAMPKT172/AMPK ratio in both adult and aged mice but increased 18FDG uptake and mitochondrial basal respiration only in adult mice. Furthermore, ND triggered rearrangement in the metabolic connectivity of adult mice and aged mice compared to age-matched controls. Altogether, our findings suggest that ND promotes hypothalamic AMPK activation, and distinct glucose metabolism and metabolic connectivity rearrangements in the brains of adult and aged mice.
Collapse
Affiliation(s)
- N R Strogulski
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - A Kopczynski
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - V G de Oliveira
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - R B Carteri
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - G Hansel
- Neuroinflammation and Neuroimmunology Laboratory, Brain Institute of Rio Grande Do Sul, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - G T Venturin
- Brain Institute of Rio Grande Do Sul (BraIns), Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - S Greggio
- Brain Institute of Rio Grande Do Sul (BraIns), Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - J C DaCosta
- Brain Institute of Rio Grande Do Sul (BraIns), Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - M A De Bastiani
- Zimmer Neuroimaging Lab, Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - M S Rodolphi
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - L V Portela
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
| |
Collapse
|
25
|
Payne A, Nahashon S, Taka E, Adinew GM, Soliman KFA. Epigallocatechin-3-Gallate (EGCG): New Therapeutic Perspectives for Neuroprotection, Aging, and Neuroinflammation for the Modern Age. Biomolecules 2022; 12:biom12030371. [PMID: 35327563 PMCID: PMC8945730 DOI: 10.3390/biom12030371] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/28/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s and Parkinson’s diseases are the two most common forms of neurodegenerative diseases. The exact etiology of these disorders is not well known; however, environmental, molecular, and genetic influences play a major role in the pathogenesis of these diseases. Using Alzheimer’s disease (AD) as the archetype, the pathological findings include the aggregation of Amyloid Beta (Aβ) peptides, mitochondrial dysfunction, synaptic degradation caused by inflammation, elevated reactive oxygen species (ROS), and cerebrovascular dysregulation. This review highlights the neuroinflammatory and neuroprotective role of epigallocatechin-3-gallate (EGCG): the medicinal component of green tea, a known nutraceutical that has shown promise in modulating AD progression due to its antioxidant, anti-inflammatory, and anti-aging abilities. This report also re-examines the current literature and provides innovative approaches for EGCG to be used as a preventive measure to alleviate AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Ashley Payne
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (A.P.); (E.T.); (G.M.A.)
| | - Samuel Nahashon
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN 37209, USA;
| | - Equar Taka
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (A.P.); (E.T.); (G.M.A.)
| | - Getinet M. Adinew
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (A.P.); (E.T.); (G.M.A.)
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (A.P.); (E.T.); (G.M.A.)
- Correspondence: ; Tel.: +1850-322-8788
| |
Collapse
|
26
|
Vitku J, Hill M, Kolatorova L, Kubala Havrdova E, Kancheva R. Steroid Sulfation in Neurodegenerative Diseases. Front Mol Biosci 2022; 9:839887. [PMID: 35281259 PMCID: PMC8904904 DOI: 10.3389/fmolb.2022.839887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Steroid sulfation and desulfation participates in the regulation of steroid bioactivity, metabolism and transport. The authors focused on sulfation and desulfation balance in three neurodegenerative diseases: Alzheimer´s disease (AD), Parkinson´s disease (PD), and multiple sclerosis (MS). Circulating steroid conjugates dominate their unconjugated counterparts, but unconjugated steroids outweigh their conjugated counterparts in the brain. Apart from the neurosteroid synthesis in the central nervous system (CNS), most brain steroids cross the blood-brain barrier (BBB) from the periphery and then may be further metabolized. Therefore, steroid levels in the periphery partly reflect the situation in the brain. The CNS steroids subsequently influence the neuronal excitability and have neuroprotective, neuroexcitatory, antidepressant and memory enhancing effects. They also exert anti-inflammatory and immunoprotective actions. Like the unconjugated steroids, the sulfated ones modulate various ligand-gated ion channels. Conjugation by sulfotransferases increases steroid water solubility and facilitates steroid transport. Steroid sulfates, having greater half-lives than their unconjugated counterparts, also serve as a steroid stock pool. Sulfotransferases are ubiquitous enzymes providing massive steroid sulfation in adrenal zona reticularis and zona fasciculata.. Steroid sulfatase hydrolyzing the steroid conjugates is exceedingly expressed in placenta but is ubiquitous in low amounts including brain capillaries of BBB which can rapidly hydrolyze the steroid sulfates coming across the BBB from the periphery. Lower dehydroepiandrosterone sulfate (DHEAS) plasma levels and reduced sulfotransferase activity are considered as risk factors in AD patients. The shifted balance towards unconjugated steroids can participate in the pathophysiology of PD and anti-inflammatory effects of DHEAS may counteract the MS.
Collapse
Affiliation(s)
- Jana Vitku
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
- *Correspondence: Jana Vitku,
| | - Martin Hill
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
| | - Lucie Kolatorova
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
| | - Eva Kubala Havrdova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Radmila Kancheva
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
| |
Collapse
|
27
|
Bianchi VE. Impact of Testosterone on Alzheimer's Disease. World J Mens Health 2022; 40:243-256. [PMID: 35021306 PMCID: PMC8987133 DOI: 10.5534/wjmh.210175] [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: 08/31/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 11/15/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease responsible for almost half of all dementia cases in the world and progressively increasing. The etiopathology includes heritability, genetic factors, aging, nutrition, but sex hormones play a relevant role. Animal models demonstrated that testosterone (T) exerted a neuroprotective effect reducing the production of amyloid-beta (Aβ), improving synaptic signaling, and counteracting neuronal death. This study aims to evaluate the impact of T deprivation and T administration in humans on the onset of dementia and AD. A search was conducted on MEDLINE and Scopus for the “androgen deprivation therapy” and “testosterone therapy” with “dementia” and “Alzheimer’s.” Studies lasting twenty years with low risk of bias, randomized clinical trial, and case-controlled studies were considered. Twelve articles on the effect of androgen deprivation therapy (ADT) and AD and seventeen on T therapy and AD were retrieved. Men with prostate cancer under ADT showed a higher incidence of dementia and AD. The effect of T administration in hypogonadal men with AD and cognitive impairment has evidenced some positive results. The majority of studies showed the T administration improved memory and cognition in AD while others did not find any benefit. Although some biases in the studies are evident, T therapy for AD patients may represent an essential clinical therapy to reduce dementia incidence and AD progression. However, more specific case-controlled trials on the effect of androgens therapy in men and women to reducing the onset of AD are necessary.
Collapse
Affiliation(s)
- Vittorio Emanuele Bianchi
- Department of Endocrinology and Metabolism, Clinical Research Center Stella Maris, Falciano, San Marino, Italy.
| |
Collapse
|
28
|
Ysrraelit MC, Correale J. Impact of Andropause on Multiple Sclerosis. Front Neurol 2021; 12:766308. [PMID: 34803897 PMCID: PMC8602357 DOI: 10.3389/fneur.2021.766308] [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: 08/28/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022] Open
Abstract
Andropause results from the natural decrease in testosterone levels that occurs with age. In contrast to menopause, which is a universal, well-characterized process associated with absolute gonadal failure, andropause ensues after gradual decline of both hypothalamic-pituitary-gonadal axis activity, as well as of testicular function, a process which usually develops over a period of many years. Increasing evidence on greater risk of Multiple sclerosis (MS) associated with lower testosterone levels is being reported. Likewise, epidemiological studies have shown a later age of onset of MS in men, relative to women, which could perhaps respond to the decline in protective testosterone levels. In this review, we will discuss the role of androgens in the development and function of the innate and adaptive immune response, as well as in neuroprotective mechanisms relevant to MS. Testosterone effects observed in different animal models and in epidemiological studies in humans will be discussed, as well as their correlation with physical disability and cognitive function levels. Finally, published and ongoing clinical trials exploring the role of androgens, particularly at key stages of sexual maturation, will be reviewed.
Collapse
Affiliation(s)
- Maria C Ysrraelit
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Jorge Correale
- Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| |
Collapse
|
29
|
Özen İ, Wang X. Biomedicine: electrospun nanofibrous hormonal therapies through skin/tissue—a review. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2021.1985493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- İlhan Özen
- Textile Engineering Department, Erciyes University, Melikgazi, Kayseri, Turkey
| | - Xungai Wang
- Institute for Frontier Materials, Deakin University, Geelong, Australia
| |
Collapse
|
30
|
Chen YH, Chen YC, Hwang LL, Yang LY, Lu DY. Deficiency in Androgen Receptor Aggravates Traumatic Brain Injury-Induced Pathophysiology and Motor Deficits in Mice. Molecules 2021; 26:6250. [PMID: 34684832 PMCID: PMC8537172 DOI: 10.3390/molecules26206250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Androgens have been shown to have a beneficial effect on brain injury and lower reactive astrocyte expression after TBI. Androgen receptors (ARs) are known to mediate the neuroprotective effects of androgens. However, whether ARs play a crucial role in TBI remains unknown. In this study, we investigated the role of ARs in TBI pathophysiology, using AR knockout (ARKO) mice. We used the controlled cortical impact model to produce primary and mechanical brain injuries and assessed motor function and brain-lesion volume. In addition, the AR knockout effects on necrosis and autophagy were evaluated after TBI. AR knockout significantly increased TBI-induced expression of the necrosis marker alpha-II-spectrin breakdown product 150 and astrogliosis marker glial fibrillary acidic protein. In addition, the TBI-induced astrogliosis increase in ARKO mice lasted for three weeks after a TBI. The autophagy marker Beclin-1 was also enhanced in ARKO mice compared with wild-type mice after TBI. Our results also indicated that ARKO mice showed a more unsatisfactory performance than wild-type mice in a motor function test following TBI. Further, they were observed to have more severe lesions than wild-type mice after injury. These findings strongly suggest that ARs play a role in TBI.
Collapse
Affiliation(s)
- Yu-Hsin Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Yen-Chou Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Ling-Ling Hwang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Laboratory of Neural Repair, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 404333, Taiwan
- Department of Photonics and Communication Engineering, Asia University, Taichung 404333, Taiwan
| |
Collapse
|
31
|
Bianchi VE, Bresciani E, Meanti R, Rizzi L, Omeljaniuk RJ, Torsello A. The role of androgens in women's health and wellbeing. Pharmacol Res 2021; 171:105758. [PMID: 34242799 DOI: 10.1016/j.phrs.2021.105758] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/29/2022]
Abstract
Androgens in women, as well as in men, are intrinsic to maintenance of (i) reproductive competency, (ii) cardiac health, (iii) appropriate bone remodeling and mass retention, (iii) muscle tone and mass, and (iv) brain function, in part, through their mitigation of neurodegenerative disease effects. In recognition of the pluripotency of endogenous androgens, exogenous androgens, and selected congeners, have been prescribed off-label for several decades to treat low libido and sexual dysfunction in menopausal women, as well as, to improve physical performance. However, long-term safety and efficacy of androgen administration has yet to be fully elucidated. Side effects often observed include (i) hirsutism, (ii) acne, (iii) deepening of the voice, and (iv) weight gain but are associated most frequently with supra-physiological doses. By contrast, short-term clinical trials suggest that the use of low-dose testosterone therapy in women appears to be effective, safe and economical. There are, however, few clinical studies, which have focused on effects of androgen therapy on pre- and post-menopausal women; moreover, androgen mechanisms of action have not yet been thoroughly explained in these subjects. This review considers clinical effects of androgens on women's health in order to prevent chronic diseases and reduce cancer risk in gynecological tissues.
Collapse
Affiliation(s)
- Vittorio E Bianchi
- Endocrinology and Metabolism, Clinical Center Stella Maris, Strada Rovereta 42, Falciano 47891, San Marino.
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Robert J Omeljaniuk
- Department of Biology, Lakehead University, 955 Oliver Rd, Thunder Bay, Ontario P7B 5E1, Canada.
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| |
Collapse
|