1
|
Marino M, Di Pietro P, D’Auria R, Lombardi M, Pastorino GMG, Troisi J, Operto FF, Carrizzo A, Vecchione C, Viggiano A, Meccariello R, Santoro A. Adult Neurogenesis Is Regulated by the Endocannabinoid and Kisspeptin Systems. Int J Mol Sci 2025; 26:3977. [PMID: 40362219 PMCID: PMC12071241 DOI: 10.3390/ijms26093977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 05/15/2025] Open
Abstract
Neurogenesis is considered the most robust form of plasticity in the adult brain. To better decipher this process, we evaluated the potential crosstalk of Kisspeptin and Endocannabinoid Systems (KPS and ECS, respectively) on hippocampal neurogenesis. Male adolescent rats were exposed to kisspeptin-10 (KP10) and the endocannabinoid anandamide (AEA) administered alone or in combination with the type 1 cannabinoid receptor (CB1R) antagonist SR141716A. The expression of Kiss1 and Kisspeptin receptor (Kiss1R) has been characterized for the first time in rat hippocampus together with the expression of the CB1R and the Transient Receptor Potential Vanilloid 1 ion channel receptor (TRPV1). Results show that both systems inhibit neurogenesis by reducing the extracellular signal-regulated kinase (ERK) signaling. Despite little differences in the expression of Kiss1R and CB1R, TRPV1 is enhanced by both KP10 and AEA treatments, suggesting TRPV1 as a common thread. KP10 administration reduces CB1R expression in the dentate gyrus, while AEA does not. KPS, unlike ECS, promotes the expression of estrogen receptor α (ER-α) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), also upregulating sirtuin 1 (SIRT1), brain-derived-neurotrophic factor (BDNF), and c-Jun. These findings suggest that the interaction between ECS and KPS could be involved in the fine-tuning of neurogenesis, highlighting a novel role for KPS.
Collapse
Affiliation(s)
- Marianna Marino
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
| | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
| | - Raffaella D’Auria
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
| | - Martina Lombardi
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
| | - Grazia Maria Giovanna Pastorino
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
- Child and Adolescent Neuropsychiatry Unit, San Giovanni di Dio Ruggi d’Aragona Hospital, 84131 Salerno, Italy;
| | - Jacopo Troisi
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
- Theoreo S.r.l. Montecorvino Pugliano, 84090 Salerno, Italy
| | - Francesca Felicia Operto
- Child and Adolescent Neuropsychiatry Unit, San Giovanni di Dio Ruggi d’Aragona Hospital, 84131 Salerno, Italy;
- Department of Health Sciences, School of Medicine, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
- Vascular Physiopathology Unit, IRCCS Neuromed Mediterranean Neurological Institute, 86077 Pozzilli, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
- Vascular Physiopathology Unit, IRCCS Neuromed Mediterranean Neurological Institute, 86077 Pozzilli, Italy
| | - Andrea Viggiano
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
| | - Rosaria Meccariello
- Department of Medical, Motor and Wellness Sciences, University of Naples Parthenope, 80133 Napoli, Italy;
| | - Antonietta Santoro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (M.M.); (P.D.P.); (R.D.); (M.L.); (G.M.G.P.); (J.T.); (A.C.); (C.V.); (A.V.)
| |
Collapse
|
2
|
Togioka BM, Harriman KA, Ye S, Berli J. Frequency and Characteristics of Postoperative Neuropathy in Individuals on Gender-Affirming Hormone Therapy Undergoing Gender Affirmation Surgery: A Retrospective Cohort Study. Cureus 2023; 15:e47988. [PMID: 38034215 PMCID: PMC10686520 DOI: 10.7759/cureus.47988] [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] [Accepted: 10/29/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Gender affirmation surgery includes procedures of the face, larynx, chest, reproductive system, external genitalia, and adipose tissue performed to ameliorate incongruence between gender identity and phenotype. The annual number of gender affirmation surgeries performed in the United States has increased significantly. There have been no investigations into the frequency of peripheral neuropathy after gender affirmation surgery, which is an important topic, given transgender individuals are at increased risk for delaying necessary medical care. After appreciating a number of cases of postoperative neuropathy in our clinical practice, we hypothesized that gender affirmation surgery is a high-risk procedure for postoperative neuropathy. Methods We conducted a one-year, monocentric, retrospective cohort study utilizing clinical data of individuals on gender-affirming hormone therapy undergoing gender-affirmation surgery under general anesthesia. The study included transgender women, assigned male at birth, receiving antiandrogen, progesterone, or estrogen therapy (target range plasma estradiol concentration 90-200 pg/ml) and transgender men, assigned female at birth, receiving antiestrogen or testosterone therapy (target range plasma testosterone concentration 320-1000 ng/dl). The primary objective was to estimate the incidence of postoperative peripheral neuropathy, defined as new numbness, paresthesia, neuropathic pain, or muscle weakness occurring in a peripheral innervation territory. Secondary objectives were to summarize the clinical presentation of neuropathy and investigate for associations between procedure characteristics and neuropathy. Results We identified nine cases of postoperative peripheral nerve injury in 232 consecutive gender affirmation procedures establishing an incidence of 3.9%. All injuries were associated with surgery longer than six hours and were characterized by sensory deficits including numbness 89% (8/9) and tingling 56% (5/9), which were diagnosed by postoperative day one. Conclusions Our results suggest that gender affirmation surgery is a high-risk procedure for postoperative neuropathy, with an incidence similar to other high-risk procedures, and an incidence that is higher than the general surgical population. However, given this has not been previously reported and our study includes a heterogenous population from a single institution, our results should be considered hypothesis generating. Additional studies that include multiple institutions are needed to confirm our findings and identify modifiable risk factors for postoperative neuropathy.
Collapse
Affiliation(s)
- Brandon M Togioka
- Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, USA
| | | | - Shangyuan Ye
- Biostatistics, Oregon Health & Science University, Portland, USA
| | - Jens Berli
- Plastic Surgery, Oregon Health & Science University, Portland, USA
| |
Collapse
|
3
|
Boueid MJ, El-Hage O, Schumacher M, Degerny C, Tawk M. Zebrafish as an emerging model to study estrogen receptors in neural development. Front Endocrinol (Lausanne) 2023; 14:1240018. [PMID: 37664862 PMCID: PMC10469878 DOI: 10.3389/fendo.2023.1240018] [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: 06/14/2023] [Accepted: 07/27/2023] [Indexed: 09/05/2023] Open
Abstract
Estrogens induce several regulatory signals in the nervous system that are mainly mediated through estrogen receptors (ERs). ERs are largely expressed in the nervous system, yet the importance of ERs to neural development has only been elucidated over the last decades. Accumulating evidence shows a fundamental role for estrogens in the development of the central and peripheral nervous systems, hence, the contribution of ERs to neural function is now a growing area of research. The conservation of the structure of the ERs and their response to estrogens make the zebrafish an interesting model to dissect the role of estrogens in the nervous system. In this review, we highlight major findings of ER signaling in embryonic zebrafish neural development and compare the similarities and differences to research in rodents. We also discuss how the recent generation of zebrafish ER mutants, coupled with the availability of several transgenic reporter lines, its amenability to pharmacological studies and in vivo live imaging, could help us explore ER function in embryonic neural development.
Collapse
Affiliation(s)
| | | | | | | | - Marcel Tawk
- *Correspondence: Cindy Degerny, ; Marcel Tawk,
| |
Collapse
|
4
|
Abdullah A, Talwar P, d'Hellencourt CL, Ravanan P. IRE1α is critical for Kaempferol-induced neuroblastoma differentiation. FEBS J 2019; 286:1375-1392. [PMID: 30719816 DOI: 10.1111/febs.14776] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/13/2019] [Accepted: 01/31/2019] [Indexed: 12/21/2022]
Abstract
Neuroblastoma is an embryonic malignancy that arises out of the neural crest cells of the sympathetic nervous system. It is the most common childhood tumor known for its spontaneous regression via the process of differentiation. The induction of differentiation using small molecules such as retinoic acid is one of the therapeutic strategies to treat the residual disease. In this study, we have reported the effect of kaempferol (KFL) in inducing differentiation of neuroblastoma cells in vitro. Treatment of neuroblastoma cells with KFL reduced the proliferation and enhanced apoptosis along with the induction of neuritogenesis. Analysis of the expression of neuron-specific markers such as β-III tubulin, neuron-specific enolase, and N-myc downregulated gene 1 revealed the process of differentiation accompanying KFL-induced apoptosis. Further analysis to understand the molecular mechanism of action showed that the effect of KFL is mediated by the activation of the endoribonuclease activity of inositol-requiring enzyme 1 alpha (IRE1α), an endoplasmic reticulum-resident transmembrane protein. In silico docking analysis and biochemical assays using recombinant human IRE1α confirm the binding of KFL to the ATP-binding site of IRE1α, which thereby activates IRE1α ribonuclease activity. Treatment of cells with the small molecule STF083010, which specifically targets and inhibits the endoribonuclease activity of IRE1α, showed reduced expression of neuron-specific markers and curtailed neuritogenesis. The knockdown of IRE1α using plasmid-based shRNA lentiviral particles also showed diminished changes in the morphology of the cells upon KFL treatment. Thus, our study suggests that KFL induces differentiation of neuroblastoma cells via the IRE1α -XBP1 pathway.
Collapse
Affiliation(s)
- Ahmad Abdullah
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, India
| | - Priti Talwar
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, India
| | - Christian Lefebvre d'Hellencourt
- Institut National de la Santé et de la Recherche Médicale, UMR Diabète Athérothombose Thérapies Réunion Océan Indien, Université de La Réunion, Saint-Denis de La Réunion, France
| | - Palaniyandi Ravanan
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, India
| |
Collapse
|
5
|
Lephart ED. Resveratrol, 4' Acetoxy Resveratrol, R-equol, Racemic Equol or S-equol as Cosmeceuticals to Improve Dermal Health. Int J Mol Sci 2017; 18:ijms18061193. [PMID: 28587197 PMCID: PMC5486016 DOI: 10.3390/ijms18061193] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 12/22/2022] Open
Abstract
Phytochemicals are botanical compounds used in dermatology applications as cosmeceuticals to improve skin health. Resveratrol and equol are two of the best-known polyphenolic or phytoestrogens having similar chemical structures and some overlapping biological functions to 17β-estradiol. Human skin gene expression was reviewed for 28 different biomarkers when resveratrol, 4′ acetoxy resveratrol (4AR), R-equol, racemic equol or S-equol were tested. Sirtuin 1 activator (SIRT 1) was stimulated by resveratrol and 4AR only. Resveratrol, R-equol and racemic equol were effective on the aging biomarkers proliferating cell nuclear factor (PCNA), nerve growth factor (NGF), 5α-reductase and the calcium binding proteins S100 A8 and A9. Racemic equol and 4AR displayed among the highest levels for the collagens, elastin and tissue inhibitor of the matrix metalloproteinase 1 (TIMP 1). S-equol displayed the lowest level of effectiveness compared to the other compounds. The 4AR analog was more effective compared to resveratrol by 1.6-fold. R-equol and racemic equol were almost equal in potency displaying greater inhibition vs. resveratrol or its 4′ analog for the matrix metalloproteinases (MMPs), but among the inflammatory biomarkers, resveratrol, 4AR, R-equol and racemic equol displayed high inhibition. Thus, these cosmeceuticals display promise to improve dermal health; however, further study is warranted to understand how phytochemicals protect/enhance the skin.
Collapse
Affiliation(s)
- Edwin D Lephart
- Department of Physiology and Developmental Biology and The Neuroscience Center, LS 4005, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
6
|
Konar A, Singh P, Thakur MK. Age-associated Cognitive Decline: Insights into Molecular Switches and Recovery Avenues. Aging Dis 2016; 7:121-9. [PMID: 27114845 PMCID: PMC4809604 DOI: 10.14336/ad.2015.1004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/04/2015] [Indexed: 12/21/2022] Open
Abstract
Age-associated cognitive decline is an inevitable phenomenon that predisposes individuals for neurological and psychiatric disorders eventually affecting the quality of life. Scientists have endeavored to identify the key molecular switches that drive cognitive decline with advancing age. These newly identified molecules are then targeted as recovery of cognitive aging and related disorders. Cognitive decline during aging is multi-factorial and amongst several factors influencing this trajectory, gene expression changes are pivotal. Identifying these genes would elucidate the neurobiological underpinnings as well as offer clues that make certain individuals resilient to withstand the inevitable age-related deteriorations. Our laboratory has focused on this aspect and investigated a wide spectrum of genes involved in crucial brain functions that attribute to senescence induced cognitive deficits. We have recently identified master switches in the epigenome regulating gene expression alteration during brain aging. Interestingly, these factors when manipulated by chemical or genetic strategies successfully reverse the age-related cognitive impairments. In the present article, we review findings from our laboratory and others combined with supporting literary evidences on molecular switches of brain aging and their potential as recovery targets.
Collapse
Affiliation(s)
- Arpita Konar
- Department of Zoology, Banaras Hindu University, Varanasi 221005, India; CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
| | - Padmanabh Singh
- Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Mahendra K Thakur
- Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
7
|
Zakir Hossain S. Enzyme-luminescence method: Tool for real-time monitoring of natural neurotoxins in vitro and l-glutamate release from primary cortical neurons. ACTA ACUST UNITED AC 2016; 9:57-65. [PMID: 28352593 PMCID: PMC5360985 DOI: 10.1016/j.btre.2015.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/06/2015] [Accepted: 12/14/2015] [Indexed: 02/05/2023]
Abstract
We describe the applicability of our previously developed novel enzyme-luminescence method for rapid and sensitive detection of natural neurotoxins (e.g., shellfish and mushroom toxins) using brain model cells (C6 glioma cells) in vitro. A racemic mixtures of the gonyautoxins (GTX), including GTX2,3 and GTX1,4 were used for evaluating the inhibition effects of these toxins on glutamate release from the C6 glioma cells. The potency was compared based on IC50 values. The activation effect of ibotenic acid (a mushroom toxin) on glutamate release from C6 cells was also evaluated. The potency was compared based on EC50 values. We also tested the applicability of our system for real-time detection of glutamate release from primary rat cortical neurons instead of model cells. This novel detection technique may be also applicable in determining neuronal differentiation ratio as well finding glutamatergic neurons without immunostaining in situ. This sensing tool may also has a great potential for the investigation of the effects of various growth factors and chemicals on neuronal differentiation, neurotransmitter dynamics, neurodegeneration, and synaptogenesis.
Novel enzyme-luminescence method is used for the rapid and sensitive in vitro detection of natural neurotoxins (e.g., shellfish and mushroom toxins) using model brain cells. Paralytic shellfish poisons gonyautoxins (e.g., GTX2,3 and GTX1,4) were detected at 1 nM level by their inhibition of glutamate release from C6 glioma cells upon drug stimulation (IC50: GTX2,3 = 30 nM and GTX1,4 = 8 nM). Activation of glutamate release from C6 cells by ibotenic acid (a mushroom toxin) was also evaluated (EC50 = 10 nM). The method was tested for real-time detection of glutamate release from primary rat cortical neurons. Dose-dependent effects of KCl (0–200 mM) and NMDA on glutamate release from primary cortical neurons were studied. The effects of different culture conditions on K+-depolarization-induced glutamate release were also investigated. The method may be applicable to screening of drugs and toxins, and finding glutamatergic neurons in brain slices without in situ staining.
Collapse
|
8
|
Bi RY, Ding Y, Gan YH. A new hypothesis of sex-differences in temporomandibular disorders: estrogen enhances hyperalgesia of inflamed TMJ through modulating voltage-gated sodium channel 1.7 in trigeminal ganglion? Med Hypotheses 2014; 84:100-3. [PMID: 25561322 DOI: 10.1016/j.mehy.2014.12.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/14/2014] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Temporomandibular disorders (TMD) are an assorted set of clinical conditions characterized mainly by pain in the temporomandibular joint (TMJ). TMJ inflammation or synovitis is frequently observed in TMD patients and is the major reason for TMD pain. TMD is prevalent in women of childbearing age, at least twice than in men, implying that estrogen may be involved in TMD pain processing. Estrogen affects a cell mainly through the estrogen receptors (ER). The estrogen-ER complex binds to estrogen response element sequences (ERE) in the promoter region of specific genes and then exerts its regulatory potential. The voltage-gated sodium channel 1.7 (Nav1.7), whose single disruption leads to a complete loss of pain, amplifies weak stimuli in the neurons and acts as the threshold channel for firing action potentials and plays a prominent role in pain perception, including inflammatory pain. Furthermore, our previous study showed that trigeminal ganglionic Nav1.7 was involved in the hyperalgesia of the inflamed TMJ. We propose that estrogen may enhance hyperalgesia of inflamed TMJ through decrease nociceptive threshold of TMJ or inflamed TMJ by modulating both expression and channel threshold of Nav1.7 in trigeminal ganglion.
Collapse
Affiliation(s)
- Rui-Yun Bi
- The Third Dental Center, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yun Ding
- The Third Dental Center, Peking University School and Hospital of Stomatology, Beijing, China.
| | - Ye-Hua Gan
- Central Laboratory and Center for Temporomandibular Disorders & Orofacial Pain, Peking University School and Hospital of Stomatology, Beijing, China.
| |
Collapse
|
9
|
Habauzit D, Ferrière F, Botherel N, Flouriot G, Pakdel F, Saligaut C. Differentiation of PC12 cells expressing estrogen receptor alpha: a new bioassay for endocrine-disrupting chemicals evaluation. CHEMOSPHERE 2014; 112:240-247. [PMID: 25048912 DOI: 10.1016/j.chemosphere.2014.03.101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 03/21/2014] [Accepted: 03/27/2014] [Indexed: 06/03/2023]
Abstract
Xeno-estrogens, a class of endocrine disrupting chemicals (EDCs), can disturb estrogen receptor-dependent pathways involved in differentiation, proliferation or protection. Multiple methods have been developed to characterize the disturbances induced by EDCs in different cells or organs. In this study we have developed a new tool for the assessment of estrogenic compounds on differentiation. For this purpose we used the global model of NGF-induced neurite outgrowth of a pseudoneuronal PC12 cell line stably transfected with estrogen receptor alpha (PC12 ER). This new test evidences a new selectivity in which estradiol, genistein and 4-hydroxytamoxifen increased the NGF-induced neurite outgrowth of PC12 ER cells in a dose-dependent manner. In contrast, the strong estrogen agonist 17α-ethynylestradiol, the strong antagonist raloxifene and the agonist bisphenol A were unable to modify the neuritogenesis of PC12 ER cells. Therefore, the analysis of neuritogenesis in PC12 ER cells constitutes a complementary tool for the characterization of xeno-estrogen activity and also serves as a basis for further studies focusing on the mechanisms of EDCs in a neuronal context. Moreover, this test constitutes an alternative to animal testing.
Collapse
Affiliation(s)
- Denis Habauzit
- Institut de Recherche en Santé Environnement et Travail (IRSET), INSERM U1085, Equipe TREC, Université de Rennes 1, SFR Biosit, Rennes, France.
| | - François Ferrière
- Institut de Recherche en Santé Environnement et Travail (IRSET), INSERM U1085, Equipe TREC, Université de Rennes 1, SFR Biosit, Rennes, France
| | - Nadine Botherel
- Institut de Recherche en Santé Environnement et Travail (IRSET), INSERM U1085, Equipe TREC, Université de Rennes 1, SFR Biosit, Rennes, France
| | - Gilles Flouriot
- Institut de Recherche en Santé Environnement et Travail (IRSET), INSERM U1085, Equipe TREC, Université de Rennes 1, SFR Biosit, Rennes, France
| | - Farzad Pakdel
- Institut de Recherche en Santé Environnement et Travail (IRSET), INSERM U1085, Equipe TREC, Université de Rennes 1, SFR Biosit, Rennes, France.
| | - Christian Saligaut
- Institut de Recherche en Santé Environnement et Travail (IRSET), INSERM U1085, Equipe TREC, Université de Rennes 1, SFR Biosit, Rennes, France
| |
Collapse
|
10
|
Synergistic Action of Flavonoids, Baicalein, and Daidzein in Estrogenic and Neuroprotective Effects: A Development of Potential Health Products and Therapeutic Drugs against Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:635694. [PMID: 24058373 PMCID: PMC3766606 DOI: 10.1155/2013/635694] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 05/15/2013] [Accepted: 06/10/2013] [Indexed: 01/13/2023]
Abstract
Despite the classical hormonal effect, estrogen has been reported to mediate neuroprotection in the brain, which leads to the searching of estrogen-like substances for treating neurodegenerative diseases. Flavonoids, a group of natural compounds, are well known to possess estrogenic effects and used to substitute estrogen, that is, phytoestrogen. Flavonoid serves as one of the potential targets for the development of natural supplements and therapeutic drugs against different diseases. The neuroprotection activity of flavonoids was chosen for a possible development of anti-Alzheimer's drugs or food supplements. The estrogenic activity of two flavonoids, baicalein and daidzein, were demonstrated by their strong abilities in stimulating estrogen receptor phosphorylation and transcriptional activation of estrogen responsive element in MCF-7 breast cells. The neuroprotection effects of flavonoids against β-amyloid (Aβ) were revealed by their inhibition effects on in vitro Aβ aggregation and Aβ-induced cytotoxicity in PC12 neuronal cells. More importantly, the estrogenic and neuroprotective activities of individual flavonoid could be further enhanced by the cotreatment in the cultures. Taken together, this synergistic effect of baicalein and daidzein might serve as a method to improve the therapeutic efficacy of different flavonoids against Aβ, which might be crucial in developing those flavonoidsin treating Alzheimer's disease in the future.
Collapse
|
11
|
Ferriere F, Habauzit D, Pakdel F, Saligaut C, Flouriot G. Unliganded estrogen receptor alpha promotes PC12 survival during serum starvation. PLoS One 2013; 8:e69081. [PMID: 23825704 PMCID: PMC3692477 DOI: 10.1371/journal.pone.0069081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 06/10/2013] [Indexed: 12/24/2022] Open
Abstract
Many studies have reported proliferative, differentiating or protective effects of estradiol, notably through estrogen receptor alpha (ERα). On the contrary, the ligand-independent action of ERα is currently poorly documented notably in cell protection. The stable transfection of wild type, substituted or truncated form of ERα in PC12 cells (ERα negative cell line) lead the specific study of its ligand-independent action. Hence, we demonstrate here that, in the absence of E2, the expression of ERα prevents cells from apoptosis induced by serum deprivation. This protection is not due to an ERE-mediated transcription and does not require either AF-1 or AF-2 transactivation functions. It is afforded to the Y537 residue of ERα and activation of c-Src/Stat3 signaling pathway.
Collapse
Affiliation(s)
- François Ferriere
- Transcription, Environment and Cancer Group, Institut de Recherche sur la Santé, Environnement et Travail (IRSET), Institut National de la Santé et de la Recherche Médicale (INSERM) U1085, Université de Rennes 1, Rennes, France.
| | | | | | | | | |
Collapse
|
12
|
Varea O, Escoll M, Diez H, Garrido J, Wandosell F. Oestradiol signalling through the Akt–mTORC1–S6K1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1052-64. [DOI: 10.1016/j.bbamcr.2012.12.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 12/11/2012] [Accepted: 12/26/2012] [Indexed: 12/12/2022]
|
13
|
Abstract
In the present report, we studied if an isoflavone, genistein, enhances the nerve growth factor (NGF)-induced neurite outgrowth of PC12 cells. Application of genistein enhanced the NGF-induced neurite outgrowth. Knockdown of Na+/K+/2Cl- cotransporter isoform 1 (NKCC1) abolished the stimulatory effect of genistein on the neurite outgrowth. These observations indicate that NKCC1 is essential for genistein to stimulate the NGF-induced neurite outgrowth, although genistein had no effect on the protein expression of NKCC1. On the other hand, genistein activates NKCC1 as shown in our previous study. Taken together, these observations indicate that genistein enhanced the NGF-induced neurite outgrowth in PC12 cells via activation of NKCC1.
Collapse
Affiliation(s)
- Ken-Ichi Nakajima
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | |
Collapse
|
14
|
Simpkins JW, Perez E, Wang X, Yang S, Wen Y, Singh M. The potential for estrogens in preventing Alzheimer's disease and vascular dementia. Ther Adv Neurol Disord 2011; 2:31-49. [PMID: 19890493 DOI: 10.1177/1756285608100427] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Estrogens are the best-studied class of drugs for potential use in the prevention of Alzheimer's disease (AD). These steroids have been shown to be potent neuroprotectants both in vitro and in vivo, and to exert effects that are consistent with their potential use in prevention of AD. These include the prevention of the processing of amyloid precursor protein (APP) into beta-amyloid (Aß), the reduction in tau hyperphosphorylation, and the elimination of catastrophic attempts at neuronal mitosis. Further, epidemiological data support the efficacy of early postmenopausal use of estrogens for the delay or prevention of AD. Collectively, this evidence supports the further development of estrogen-like compounds for prevention of AD. Several approaches to enhance brain specificity of estrogen action are now underway in an attempt to reduce the side effects of chronic estrogen therapy in AD.
Collapse
Affiliation(s)
- James W Simpkins
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, Center FOR HER (Focused On Resources for her Health, Education and Research), University of North Texas Health Science Center, Fort Worth, TX, USA
| | | | | | | | | | | |
Collapse
|
15
|
Li XF, Lui CNP, Jiang ZH, Ken YKL. Neuroprotective effects of ginsenosides Rh1 and Rg2 on neuronal cells. Chin Med 2011; 6:19. [PMID: 21592408 PMCID: PMC3121663 DOI: 10.1186/1749-8546-6-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 05/19/2011] [Indexed: 12/26/2022] Open
Abstract
Background The present study investigates the effects of ginsenosides Rh1 and Rg2 against 6-hydroxydopamine (6-OHDA), a neurotoxin on SH-SY5Y cells and PC-12 cells. The effects of these two ginsenosides on neuronal differentiation are also examined. Methods LDH assay was used to measure cell viability after exposure to 6-OHDA and ginsenosides. Neuronal differentiation was evaluated by changes in cell morphology and density of neurite outgrowths. Western blotting was used to determine the ginsenosides' effects on activation of extracellular signal-regulated protein kinases (ERKs). Results Rh1 and Rg2 attenuated 6-OHDA toxicity in SH-SY5Y cells and induced neurite outgrowths in PC-12 cells. 6-OHDA-induced ERK phosphorylation was decreased by Rh1 and Rg2. 20(R)-form and 20(S)-form of the ginsenosides exerted similar effects in inducing neurite outgrowths in PC-12 cells. Conclusion The present study demonstrates neuroprotective effects of ginsenosides Rh1 and Rg2 on neuronal cell lines. These results suggest potential Chinese medicine treatment for neurodegenerative disorders (eg Parkinson's disease).
Collapse
Affiliation(s)
- Xiao-Fan Li
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China.
| | | | | | | |
Collapse
|
16
|
Zhou FC, Balaraman Y, Teng M, Liu Y, Singh RP, Nephew KP. Alcohol alters DNA methylation patterns and inhibits neural stem cell differentiation. Alcohol Clin Exp Res 2011; 35:735-46. [PMID: 21223309 DOI: 10.1111/j.1530-0277.2010.01391.x] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Potential epigenetic mechanisms underlying fetal alcohol syndrome (FAS) include alcohol-induced alterations of methyl metabolism, resulting in aberrant patterns of DNA methylation and gene expression during development. Having previously demonstrated an essential role for epigenetics in neural stem cell (NSC) development and that inhibiting DNA methylation prevents NSC differentiation, here we investigated the effect of alcohol exposure on genome-wide DNA methylation patterns and NSC differentiation. METHODS Neural stem cells in culture were treated with or without a 6-hour 88 mM ("binge-like") alcohol exposure and examined at 48 hours, for migration, growth, and genome-wide DNA methylation. The DNA methylation was examined using DNA-methylation immunoprecipitation followed by microarray analysis. Further validation was performed using Independent Sequenom analysis. RESULTS Neural stem cell differentiated in 24 to 48 hours with migration, neuronal expression, and morphological transformation. Alcohol exposure retarded the migration, neuronal formation, and growth processes of NSC, similar to treatment with the methylation inhibitor 5-aza-cytidine. When NSC departed from the quiescent state, a genome-wide diversification of DNA methylation was observed-that is, many moderately methylated genes altered methylation levels and became hyper- and hypomethylated. Alcohol prevented many genes from such diversification, including genes related to neural development, neuronal receptors, and olfaction, while retarding differentiation. Validation of specific genes by Sequenom analysis demonstrated that alcohol exposure prevented methylation of specific genes associated with neural development [cut-like 2 (cutl2), insulin-like growth factor 1 (Igf1), epidermal growth factor-containing fibulin-like extracellular matrix protein 1 (Efemp1), and SRY-box-containing gene 7 (Sox 7)]; eye development, lens intrinsic membrane protein 2 (Lim 2); the epigenetic mark Smarca2 (SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 2); and developmental disorder [DiGeorge syndrome critical region gene 2 (Dgcr2)]. Specific sites altered by DNA methylation also correlated with transcription factor binding sites known to be critical for regulating neural development. CONCLUSION The data indicate that alcohol prevents normal DNA methylation programming of key neural stem cell genes and retards NSC differentiation. Thus, the role of DNA methylation in FAS warrants further investigation.
Collapse
Affiliation(s)
- Feng C Zhou
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS 508, Indianapolis, Indiana 46202, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Habauzit D, Flouriot G, Pakdel F, Saligaut C. Effects of estrogens and endocrine-disrupting chemicals on cell differentiation-survival-proliferation in brain: contributions of neuronal cell lines. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2011; 14:300-327. [PMID: 21790314 DOI: 10.1080/10937404.2011.578554] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Estrogens and estrogen receptors (ER) are key actors in the control of differentiation and survival and act on extrareproductive tissues such as brain. Thus, estrogens may display neuritogenic effects during development and neuroprotective effects in the pathophysiological context of brain ischemia and neurodegenerative pathologies like Alzheimer's disease or Parkinson's disease. Some of these effects require classical transcriptional "genomic" mechanisms through ER, whereas other effects appear to rely clearly on "membrane-initiated mechanisms" through cytoplasmic signal transduction pathways. Disturbances of these mechanisms by endocrine-disrupting chemicals (EDC) may exert adverse effects on brain. Some EDC may act via ER-independent mechanisms but might cross-react with endogenous estrogen. Other EDC may act through ER-dependent mechanisms and display agonistic/antagonistic estrogenic properties. Because of these potential effects of EDC, it is necessary to establish sensitive cell-based assays to determine EDC effects on brain. In the present review, some effects of estrogens and EDC are described with focus on ER-mediated effects in neuronal cells. Particular attention is given to PC12 cells, an interesting model to study the mechanisms underlying ER-mediated differentiating and neuroprotective effects of estrogens.
Collapse
Affiliation(s)
- Denis Habauzit
- UMR CNRS 6026 (Interactions Cellulaires et Moléculaires, Equipe RED), Université de Rennes 1, Rennes, France
| | | | | | | |
Collapse
|
18
|
Diotel N, Le Page Y, Mouriec K, Tong SK, Pellegrini E, Vaillant C, Anglade I, Brion F, Pakdel F, Chung BC, Kah O. Aromatase in the brain of teleost fish: expression, regulation and putative functions. Front Neuroendocrinol 2010; 31:172-92. [PMID: 20116395 DOI: 10.1016/j.yfrne.2010.01.003] [Citation(s) in RCA: 234] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 01/20/2010] [Accepted: 01/24/2010] [Indexed: 12/25/2022]
Abstract
Unlike that of mammals, the brain of teleost fish exhibits an intense aromatase activity due to the strong expression of one of two aromatase genes (aromatase A or cyp19a1a and aromatase B or cyp19a1b) that arose from a gene duplication event. In situ hybridization, immunohistochemistry and expression of GFP (green fluorescent protein) in transgenic tg(cyp19a1b-GFP) fish demonstrate that aromatase B is only expressed in radial glial cells (RGC) of adult fish. These cells persist throughout life and act as progenitors in the brain of both developing and adult fish. Although aromatase B-positive radial glial cells are most abundant in the preoptic area and the hypothalamus, they are observed throughout the entire central nervous system and spinal cord. In agreement with the fact that brain aromatase activity is correlated to sex steroid levels, the high expression of cyp19a1b is due to an auto-regulatory loop through which estrogens and aromatizable androgens up-regulate aromatase expression. This mechanism involves estrogen receptor binding on an estrogen response element located on the cyp19a1b promoter. Cell specificity is achieved by a mandatory cooperation between estrogen receptors and unidentified glial factors. Given the emerging roles of estrogens in neurogenesis, the unique feature of the adult fish brain suggests that, in addition to classical functions on brain sexual differentiation and sexual behaviour, aromatase expression in radial glial cells could be part of the mechanisms authorizing the maintenance of a high proliferative activity in the brain of fish.
Collapse
Affiliation(s)
- Nicolas Diotel
- Neurogenesis And OEstrogens, UMR CNRS 6026, IFR 140, Université de Rennes 1, Rennes, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ghosh S, Thakur MK. Interaction of estrogen receptor-alpha ligand binding domain with nuclear proteins of aging mouse brain. J Neurosci Res 2009; 87:2591-600. [PMID: 19326447 DOI: 10.1002/jnr.22068] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
After the interaction of estrogen with the ligand binding domain (LBD) of mouse estrogen receptor-alpha (mERalpha) and hormone-responsive elements of target genes, many nuclear proteins are recruited to regulate the expression of specific genes. Because it is not known which brain proteins interact with LBD or whether these proteins vary with age and sex, we used pull-down assay and far Western blotting to detect five nuclear proteins of 160, 140, 87, 60, and 46 kD in the mouse brain. These interacting proteins were identified as PELP1, RIP140, PGC1alpha, BAF60, and ADA3, respectively. The level of PELP1, RIP140, PGC1alpha, and BAF60 decreased drastically in old compared with adult male mice, whereas the ADA3 level showed no significant change. PELP1, PGC1alpha, and BAF60 levels were lower in old male compared with female mice. Thus we report the identification and interaction of five nuclear proteins with mERalpha-LBD, indicating their role in estrogen signaling and brain functions during aging.
Collapse
Affiliation(s)
- Swati Ghosh
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
20
|
Ghosh S, Thakur MK. Interaction of estrogen receptor-alpha transactivation domain with nuclear proteins of mouse brain: p68 RNA helicase shows age- and sex-specific change. J Neurosci Res 2009; 87:1323-8. [PMID: 19025768 DOI: 10.1002/jnr.21948] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Estrogen receptor (ER)-alpha interacts with nuclear proteins to mediate its multiple functions in the brain. However, it is not known which proteins interact with the ERalpha-transactivation domain (TAD) in mouse brain and whether they change with age and sex. Therefore, we have used affinity-purified GST-tagged mouse ERalpha-TAD fusion protein for interaction with nuclear proteins from the mouse brain. The pull-down assay and far-Western blotting detected four nuclear proteins of 100, 80, 68, and 50 kD. We have recently identified the 80-kD protein as MTA1 and demonstrated its decrease in old age. Here we report alteration in the interaction and expression of the 68-kD protein of adult and old mice of both sexes. This protein was identified as p68 RNA helicase through NCBI database search, immunoprecipitation, and immunoblotting. Further analysis showed that the extent of its interaction was relatively lower in old mice of both sexes and in male mice of both ages compared with their counterparts. However, the expression of p68 was significantly lower in old males compared with adult males, although other groups did not show significant changes. Such age- and sex-specific interaction of p68 suggests its implication in ERalpha-mediated brain functions during aging.
Collapse
Affiliation(s)
- Swati Ghosh
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
21
|
Mérot Y, Ferrière F, Gailhouste L, Huet G, Percevault F, Saligaut C, Flouriot G. Different outcomes of unliganded and liganded estrogen receptor-alpha on neurite outgrowth in PC12 cells. Endocrinology 2009; 150:200-11. [PMID: 18772239 DOI: 10.1210/en.2008-0449] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A precise description of the mechanisms by which estrogen receptor-alpha (ERalpha) exerts its influences on cellular growth and differentiation is still pending. Here, we report that the differentiation of PC12 cells is profoundly affected by ERalpha. Importantly, depending upon its binding to 17beta-estradiol (17betaE2), ERalpha is found to exert different effects on pathways involved in nerve growth factor (NGF) signaling. Indeed, upon its stable expression in PC12 cells, unliganded ERalpha is able to partially inhibit the neurite outgrowth induced by NGF. This process involves a repression of MAPK and phosphatidylinositol 3-kinase/Akt signaling pathways, which leads to a negative regulation of markers of neuronal differentiation such as VGF and NFLc. This repressive action of unliganded ERalpha is mediated by its D domain and does not involve its transactivation and DNA-binding domains, thereby suggesting that direct transcriptional activity of ERalpha is not required. In contrast with this repressive action occurring in the absence of 17betaE2, the expression of ERalpha in PC12 cells allows 17betaE2 to potentiate the NGF-induced neurite outgrowth. Importantly, 17betaE2 has no impact on NGF-induced activity of MAPK and Akt signaling pathways. The mechanisms engaged by liganded ERalpha are thus unlikely to rely on an antagonism of the inhibition mediated by the unliganded ERalpha. Furthermore, 17betaE2 enhances NGF-induced response of VGF and NFLc neuronal markers in PC12 clones expressing ERalpha. This stimulatory effect of 17betaE2 requires the transactivation functions of ERalpha and its D domain, suggesting that an estrogen-responsive element-independent transcriptional mechanism is potentially relevant for the neuritogenic properties of 17betaE2 in ERalpha-expressing PC12 cells.
Collapse
Affiliation(s)
- Yohann Mérot
- Université de Rennes 1, Centre National de la Recherche Scientifique, Unité Mixte 6026, Equipe Récepteur des oestrogènes et destinée cellulaire, 35042 Rennes, France
| | | | | | | | | | | | | |
Collapse
|
22
|
Ishunina TA, Swaab DF. Age-dependent ERα MB1 splice variant expression in discrete areas of the human brain. Neurobiol Aging 2008; 29:1177-89. [PMID: 17368651 DOI: 10.1016/j.neurobiolaging.2007.02.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 01/08/2007] [Accepted: 02/10/2007] [Indexed: 11/29/2022]
Abstract
A role of estrogens in brain aging and Alzheimer's disease (AD) is a hot topic of research. We show in material of 71 patients that the estrogen receptor alpha (ERalpha) splice variant MB1 is expressed at the protein and mRNA level in the human brain. MB1 is mainly confined to astrocytes, membranes and cytoplasm of projecting neurons and endothelial cells. It was consistently observed in the thalamus, colliculus inferior, pontine nuclei, dorsal motor nucleus of vagus, some motor neurons in the anterior and lateral horns of the spinal cord and rarely in pyramidal neurons of the cerebral cortex. The highest level of MB1 immunoreactivity (MB1-ir) was noted in the caudal hypothalamus, in particular in the tuberomamillary nucleus (TMN). MB1-ir in the TMN increased during aging in women. MB1-ir was higher in young (<50 years of age) men than in premenopausal women. No significant changes of this variant were observed in the TMN of AD cases. In conclusion, MB1 may function as a dominant negative isoform in the human brain.
Collapse
Affiliation(s)
- Tatjana A Ishunina
- Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | | |
Collapse
|
23
|
Zhu JTT, Choi RCY, Chu GKY, Cheung AWH, Gao QT, Li J, Jiang ZY, Dong TTX, Tsim KWK. Flavonoids possess neuroprotective effects on cultured pheochromocytoma PC12 cells: a comparison of different flavonoids in activating estrogenic effect and in preventing beta-amyloid-induced cell death. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2007; 55:2438-45. [PMID: 17323972 DOI: 10.1021/jf063299z] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Despite the classical hormonal effect, estrogen possesses a neuroprotective effect in the brain, which has led many to search for novel treatments for neurodegenerative diseases. Flavonoids, a group of compounds mainly derived from vegetables, share a resemblance, chemically, to estrogen, and indeed, some have been used as estrogen substitutes. To search for potential therapeutic agents against neurodegenerative diseases, different subclasses of flavonoids were analyzed and compared with estrogen. First, the estrogenic activities of these flavonoids were determined by activating the estrogen-responsive elements in cultured MCF-7 breast cancer cells. Second, the neuroprotective effects of flavonoids were revealed by measuring its inhibition effects on the formation of reactive oxygen species, the aggregation of beta-amyloid, and the induction of cell death by beta-amyloid in cultured neuronal PC12 cells. Among these flavonoids, baicalein, scutellarin, hibifolin, and quercetin-3'-glucoside possessed the strongest effect in neuroprotection; however, the neuroprotective activity did not directly correlate with the estrogenic activity of the flavonoids. Identification of these flavonoids could be very useful in finding potential drugs, or food supplements, for treating Alzheimer's disease.
Collapse
Affiliation(s)
- Judy T T Zhu
- Department of Biology and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|