1
|
Anderson KJ, Thorolfsdottir ET, Nodelman IM, Halldorsdottir ST, Benonisdottir S, Alghamdi M, Almontashiri N, Barry BJ, Begemann M, Britton JF, Burke S, Cogne B, Cohen AS, de Diego Boguñá C, Eichler EE, Engle EC, Fahrner JA, Faivre L, Fradin M, Fuhrmann N, Gao CW, Garg G, Grečmalová D, Grippa M, Harris JR, Hoekzema K, Hershkovitz T, Hubbard S, Janssens K, Jurgens JA, Kmoch S, Knopp C, Koptagel MA, Ladha FA, Lapunzina P, Lindau T, Meuwissen M, Minicucci A, Neuhaus E, Nizon M, Nosková L, Park K, Patel C, Pfundt R, Prasun P, Rahner N, Robin NH, Ronspies C, Roohi J, Rosenfeld J, Saenz M, Saunders C, Stark Z, Thiffault I, Thull S, Velasco D, Velmans C, Verseput J, Vitobello A, Wang T, Weiss K, Wentzensen IM, Pilarowski G, Eysteinsson T, Gillentine M, Stefánsson K, Helgason A, Bowman GD, Bjornsson HT. Androgens mediate sexual dimorphism in Pilarowski-Bjornsson Syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.06.25326635. [PMID: 40385454 PMCID: PMC12083630 DOI: 10.1101/2025.05.06.25326635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Sex-specific penetrance in autosomal dominant Mendelian conditions is largely understudied. The neurodevelopmental disorder Pilarowski-Bjornsson syndrome (PILBOS) was initially described in females. Here, we describe the clinical and genetic characteristics of the largest PILBOS cohort to date, showing that both sexes can exhibit PILBOS features, although males are overrepresented. A mouse model carrying a human-derived Chd1 missense variant (Chd1 R616Q/+) displays female-restricted phenotypes, including growth deficiency, anxiety and hypotonia. Orchiectomy unmasks a growth deficiency phenotype in male Chd1 R616Q/+ mice, while testosterone rescues the phenotype in females, implicating androgens in phenotype modulation. In the gnomAD and UK Biobank databases, rare missense variants in CHD1 are overrepresented in males, supporting a male protective effect. We identify 33 additional highly constrained autosomal genes with missense variant overrepresentation in males. Our results support androgen-regulated sexual dimorphism in PILBOS and open novel avenues to understand the mechanistic basis of sexual dimorphism in other autosomal Mendelian disorders.
Collapse
Affiliation(s)
- Kimberley Jade Anderson
- Department of Genetics and Molecular Medicine, Landspitali University Hospital, Reykjavik, Iceland
| | | | - Ilana M. Nodelman
- T.C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, MD, USA
| | - Sara Tholl Halldorsdottir
- The Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Stefania Benonisdottir
- Institute of Physical Sciences, University of Iceland, Reykjavik, Iceland
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
| | - Malak Alghamdi
- Medical Genetics Division, Pediatric Department, College of Medicine, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Naif Almontashiri
- College of Applied Medical Sciences and Center for Genetics and Inherited Diseases, Taibah University, Madinah, Kingdom of Saudi Arabia
- Research Department, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - Brenda J. Barry
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Matthias Begemann
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jacquelyn F. Britton
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Burke
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Benjamin Cogne
- Nantes Université, CHU de Nantes, CNRS, INSERM, l’institut du thorax, F-44000 Nantes, France
- Nantes Université, CHU de Nantes, Service de Génétique médicale, F-44000 Nantes, France
| | - Ana S.A. Cohen
- Department of Pathology and Laboratory Medicine, Genomic Medicine Center, Children’s Mercy-Kansas City, Kansas City, MO, USA
- The University of Missouri-Kansas City, School of Medicine, Kansas City, MO, USA
| | | | - Evan E. Eichler
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Elizabeth C. Engle
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Ophthalmology, Boston Children’s Hospital, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jill A. Fahrner
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laurence Faivre
- INSERM UMR1231, GAD team, Univeristé de Bourgogne Europe, Dijon, France
- Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, de l’Interrégion Est, Centre Hospitalier Universitaire Dijon, 21079 Dijon, France
| | - Mélanie Fradin
- Service de Genetique Medicale, Centre Labellisé Anomalies du Développement de l’Ouest, CHU Rennes, Rennes, France
| | - Nico Fuhrmann
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christine W. Gao
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gunjan Garg
- Department of Clinical Genetics, Liverpool Hospital, Sydney, New South Wales, Australia
- Hunter Genetics, Waratah, New South Wales, Australia
- School of Women’s and Children’s Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Dagmar Grečmalová
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Czech Republic
| | - Mina Grippa
- SSD Medical Genetics, Maternal and Child Department, AOU Policlinico Modena, Modena, Italy
| | - Jacqueline R. Harris
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kennedy Krieger Institute, Department of Neurology, Baltimore, Maryland, USA
| | - Kendra Hoekzema
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Sydney Hubbard
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Katrien Janssens
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Julie A. Jurgens
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cordula Knopp
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Meral Aktas Koptagel
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Farah A. Ladha
- Baylor College of Medicine, Department of Molecular and Human Genetics, Houston, TX, USA
| | - Pablo Lapunzina
- INGEMM-Institute of Medical and Molecular Genetics, IdiPAZ- CIBERER- Hospital Universitario La Paz, Madrid, Spain and ERNITHACA, Madrid, Spain
| | - Tobias Lindau
- Department of Pediatrics, Gemeinschaftsklinikum Mittelrhein Kemperhof, Koblenzer Straße 115-155, 56073 Koblenz, Germany
| | - Marije Meuwissen
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Andreina Minicucci
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, U.O. Genetica Medica, 40138 Bologna, Italy
| | - Emily Neuhaus
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Mathilde Nizon
- Service de Génétique Médicale, Unité de Génétique Clinique, Nantes, France
| | - Lenka Nosková
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kristen Park
- Departments of Pediatrics and Neurology, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
| | - Chirag Patel
- Genetic Health Queensland, Royal Brisbane & Women’s Hospital Campus, Herston, Brisbane, Australia
| | - Rolph Pfundt
- Department of Human Genetics, Donders Institute, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pankaj Prasun
- Division of Genetics, Department of Pediatrics West Virginia School of Medicine, Morgantown, USA
| | - Nils Rahner
- MVZ Institute for Clinical Genetics and Tumor Genetics, Bonn, Germany
| | - Nathaniel H. Robin
- Department of Genetics, UAB Heersink School of Medicine, Birmingham AL, USA
| | - Carey Ronspies
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jasmin Roohi
- Department of Genetics, Mid-Atlantic Permanente Medical Group, Washington, DC, USA
| | - Jill Rosenfeld
- Baylor College of Medicine, Department of Molecular and Human Genetics, Houston, TX, USA
- Baylor Genetics, Houston, TX, USA
| | - Margarita Saenz
- Departments of Pediatrics and Neurology, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
| | - Carol Saunders
- Department of Pathology and Laboratory Medicine, Genomic Medicine Center, Children’s Mercy-Kansas City, Kansas City, MO, USA
- The University of Missouri-Kansas City, School of Medicine, Kansas City, MO, USA
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Parkville, Victoria, Australia
| | - Isabelle Thiffault
- Department of Pathology and Laboratory Medicine, Genomic Medicine Center, Children’s Mercy-Kansas City, Kansas City, MO, USA
- The University of Missouri-Kansas City, School of Medicine, Kansas City, MO, USA
| | - Sarah Thull
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Danita Velasco
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Clara Velmans
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jolijn Verseput
- Human Genetics Department, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Antonio Vitobello
- INSERM UMR1231, GAD team, Univeristé de Bourgogne Europe, Dijon, France
| | - Tianyun Wang
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- Neuroscience Research Institute, Peking University; Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing 100191, China
- Autism Research Center, Peking University Health Science Center, Beijing 100191, China
| | - Karin Weiss
- The Genetics Institute Rambam Health Care Campus Haifa Israel
| | | | | | - Thor Eysteinsson
- Department of Physiology, Faculty of Medicine, University of Iceland
- Department of Ophthalmology, Landspitali University Hospital, Reykjavik, Iceland
| | | | - Kári Stefánsson
- deCODE Genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Agnar Helgason
- deCODE Genetics/Amgen Inc., Reykjavik, Iceland
- Department of Anthropology, University of Iceland, Reykjavik, Iceland
| | - Gregory D. Bowman
- T.C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, MD, USA
| | - Hans Tomas Bjornsson
- Department of Genetics and Molecular Medicine, Landspitali University Hospital, Reykjavik, Iceland
- The Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Clina JG, Danon JC, Helsel BC, Lepping RJ, Martin LE, Sherman JR, Brucks MG, Donnelly JE, Ptomey LT. The association between cardiorespiratory fitness and resting-state functional connectivity in adults with Down syndrome. Alzheimers Dement 2025; 21:e70297. [PMID: 40394893 DOI: 10.1002/alz.70297] [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: 12/20/2024] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION Resting-state functional connectivity (FC) of the default mode network (DMN) is linked to Alzheimer's disease in people with Down syndrome (DS). In adults without DS, cardiorespiratory fitness is associated with DMN FC; however, this has not been unexplored in DS. METHODS This analysis used baseline data from an intervention in adults with DS. Resting-state functional magnetic resonance imaging measured connectivity from the posterior cingulate cortex seed to DMN nodes. Fitness was measured by the maximal treadmill test. Pearson correlations and linear regressions were used to examine the associations between fitness and FC. RESULTS Data from 40 adults with DS (26.0 years, 58% female) showed fitness was associated with overall DMN connectivity (r = 0.472, p = 0.004) and medial prefrontal cortex connectivity (r = 0.431, p = 0.010). The association between fitness and DMN FC remained significant after adjustment for age and sex (β = 0.0072, p = 0.04). DISCUSSION Fitness may be associated with DMN FC in DS. HIGHLIGHTS In adults with DS, cardiorespiratory fitness was associated with overall DMN connectivity, which remained significant after adjusting for age and sex. No associations were found between moderate to vigorous physical activity and DMN connectivity. Increasing fitness may be a therapeutic strategy for AD prevention or delay in DS.
Collapse
Affiliation(s)
- Julianne G Clina
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jessica C Danon
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Brian C Helsel
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Rebecca J Lepping
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Laura E Martin
- Department of Population Health, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Joseph R Sherman
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Morgan G Brucks
- Department of Population Health, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Joseph E Donnelly
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Lauren T Ptomey
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
3
|
Guo Y, Piasecki J, Swiecicka A, Ireland A, Phillips BE, Atherton PJ, Stashuk D, Rutter MK, McPhee JS, Piasecki M. Circulating testosterone and dehydroepiandrosterone are associated with individual motor unit features in untrained and highly active older men. GeroScience 2022; 44:1215-1228. [PMID: 34862585 PMCID: PMC9213614 DOI: 10.1007/s11357-021-00482-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/28/2021] [Indexed: 10/31/2022] Open
Abstract
Long-term exercise training has been considered as an effective strategy to counteract age-related hormonal declines and minimise muscle atrophy. However, human data relating circulating hormone levels with motor nerve function are scant. The aims of the study were to explore associations between circulating sex hormone levels and motor unit (MU) characteristics in older men, including masters athletes competing in endurance and power events. Forty-three older men (mean ± SD age: 69.9 ± 4.6 years) were studied based on competitive status. The serum concentrations of dehydroepiandrosterone (DHEA), total testosterone (T) and estradiol were quantified using liquid chromatography mass spectrometry. Intramuscular electromyographic signals were recorded from vastus lateralis (VL) during 25% of maximum voluntary isometric contractions and processed to extract MU firing rate (FR), and motor unit potential (MUP) features. After adjusting for athletic status, MU FR was positively associated with DHEA levels (p = 0.019). Higher testosterone and estradiol were associated with lower MUP complexity; these relationships remained significant after adjusting for athletic status (p = 0.006 and p = 0.019, respectively). Circulating DHEA was positively associated with MU firing rate in these older men. Higher testosterone levels were associated with reduced MUP complexity, indicating reduced electrophysiological temporal dispersion, which is related to decreased differences in conduction times along axonal branches and/or MU fibres. Although evident in males only, this work highlights the potential of hormone administration as a therapeutic interventional strategy specifically targeting human motor units in older age.
Collapse
Affiliation(s)
- Yuxiao Guo
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham BRC, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jessica Piasecki
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, Nottingham Trent University, Nottingham, UK
| | - Agnieszka Swiecicka
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| | - Alex Ireland
- Department of Sport and Exercise Sciences, Musculoskeletal Science and Sports Medicine Research Centre, Manchester Metropolitan University, Manchester, UK
| | - Bethan E. Phillips
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham BRC, School of Medicine, University of Nottingham, Nottingham, UK
| | - Philip J. Atherton
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham BRC, School of Medicine, University of Nottingham, Nottingham, UK
| | - Daniel Stashuk
- Department of Systems Design Engineering, University of Waterloo, Waterloo, ON Canada
| | - Martin K. Rutter
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Diabetes, Endocrinology and Metabolism Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Jamie S. McPhee
- Department of Sport and Exercise Sciences, Musculoskeletal Science and Sports Medicine Research Centre, Manchester Metropolitan University, Manchester, UK
| | - Mathew Piasecki
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham BRC, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
4
|
McLeod VM, Chiam MDF, Perera ND, Lau CL, Boon WC, Turner BJ. Mapping Motor Neuron Vulnerability in the Neuraxis of Male SOD1 G93A Mice Reveals Widespread Loss of Androgen Receptor Occurring Early in Spinal Motor Neurons. Front Endocrinol (Lausanne) 2022; 13:808479. [PMID: 35273564 PMCID: PMC8902593 DOI: 10.3389/fendo.2022.808479] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/19/2022] [Indexed: 12/11/2022] Open
Abstract
Sex steroid hormones have been implicated as disease modifiers in the neurodegenerative disorder amyotrophic lateral sclerosis (ALS). Androgens, signalling via the androgen receptor (AR), predominate in males, and have widespread actions in the periphery and the central nervous system (CNS). AR translocates to the cell nucleus when activated upon binding androgens, whereby it regulates transcription of target genes via the classical genomic signalling pathway. We previously reported that AR protein is decreased in the lumbar spinal cord tissue of symptomatic male SOD1G93A mice. Here, we further explored the changes in AR within motor neurons (MN) of the CNS, assessing their nuclear AR content and propensity to degenerate by endstage disease in male SOD1G93A mice. We observed that almost all motor neuron populations had undergone significant loss in nuclear AR in SOD1G93A mice. Interestingly, loss of nuclear AR was evident in lumbar spinal MNs as early as the pre-symptomatic age of 60 days. Several MN populations with high AR content were identified which did not degenerate in SOD1G93A mice. These included the brainstem ambiguus and vagus nuclei, and the sexually dimorphic spinal MNs: cremaster, dorsolateral nucleus (DLN) and spinal nucleus of bulbocavernosus (SNB). In conclusion, we demonstrate that AR loss directly associates with MN vulnerability and disease progression in the SOD1G93A mouse model of ALS.
Collapse
Affiliation(s)
- Victoria M. McLeod
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Mathew D. F. Chiam
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Nirma D. Perera
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Chew L. Lau
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Wah Chin Boon
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Bradley J. Turner
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Perron Institute for Neurological and Translational Science, Queen Elizabeth Medical Centre, Nedlands, WA, Australia
- *Correspondence: Bradley J. Turner,
| |
Collapse
|
5
|
Namekawa T, Kitayama S, Ikeda K, Horie-Inoue K, Suzuki T, Okamoto K, Ichikawa T, Yano A, Kawakami S, Inoue S. HIF1α inhibitor 2-methoxyestradiol decreases NRN1 expression and represses in vivo and in vitro growth of patient-derived testicular germ cell tumor spheroids. Cancer Lett 2020; 489:79-86. [PMID: 32544513 DOI: 10.1016/j.canlet.2020.05.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/25/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022]
Abstract
Testicular germ cell tumor (GCT) is the most common type of malignancy in young males. Patients with nonseminomatous GCT still have poor prognosis. To identify new therapeutic targets, we generated patient-derived cells (PDCs) and their xenograft (PDCX) models from 3 distinct GCT patients' specimens. The pathological features of GCT PDCs and PDCX tumors recapitulated those of nonseminomatous components exhibiting in the corresponding patients' specimens. Notably, stemness-related markers and hypoxia-related genes, including hypoxia inducible factor 1α (HIF1A) and neuritin 1 (NRN1), were abundantly expressed in three-dimensional spheroid cultures of GCT PDCs. We identified functional HIF1α response elements in the NRN1 promoter and defined that their transcriptional activities were substantially activated by hypoxia. HIF1α inhibition by siRNAs or an inhibitor, 2-methoxyestradiol, significantly suppressed NRN1 expression and decreased the in vitro and in vivo growth of PDC spheroids. Moreover, NRN1 knockdown efficiently suppressed PDC proliferation. These results suggest that HIF1α and NRN1 are potential diagnostic and therapeutic targets, and that 2-methoxyestradiol could be applied to clinical management of GCT. Overall, our GCT PDC and PDCX models would be useful as preclinical models for precision medicine targeting each patient.
Collapse
Affiliation(s)
- Takeshi Namekawa
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan; Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Chiba, 260-8677, Japan
| | - Sachi Kitayama
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Koji Okamoto
- Division of Cancer Differentiation, National Cancer Center Hospital, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tomohiko Ichikawa
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Chiba, 260-8677, Japan
| | - Akihiro Yano
- Department of Urology, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, 350-8550, Japan
| | - Satoru Kawakami
- Department of Urology, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, 350-8550, Japan
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan; Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, 173-0015, Japan.
| |
Collapse
|
6
|
Rahmawati E, Yang WCV, Lei YP, Maurya PK, Chen HW, Tzeng CR. Decreased Level of Neurotrophic Factor Neuritin 1 in Women with Ovarian Endometriosis after Receiving Gonadotropin-Releasing Hormone Agonist Treatment. Int J Mol Sci 2019; 20:E4352. [PMID: 31491902 PMCID: PMC6770869 DOI: 10.3390/ijms20184352] [Citation(s) in RCA: 4] [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: 08/23/2019] [Revised: 08/31/2019] [Accepted: 09/03/2019] [Indexed: 12/19/2022] Open
Abstract
This study aimed to investigate the effect of gonadotropin-releasing hormone agonist (GnRHa) treatment on the expression of neuritin 1 (NRN1) in women with ovarian endometriosis. We collected tissues and serum from women with endometriosis treated with (n = 45) or without (n = 37) GnRHa. NRN1 mRNA and protein levels were measured using qPCR and Western blot. Immunolocalization of NRN1 in endometriotic tissues was examined using immunohistochemistry. In addition, a follow-up study was carried out to monitor the serum level of NRN1 in patients before and after GnRHa treatment. Both mRNA (p = 0.046) and protein (p = 0.0155) levels of NRN1 were significantly lower in endometriotic tissues from patients receiving GnRHa treatment compared to the untreated group. Both epithelial and stromal cells of endometriotic tissues from untreated women with endometriosis exhibited stronger staining of NRN1 but not in those who were treated with GnRHa. The follow-up study showed that the serum level of the NRN1 concentration decreased significantly from 1149 ± 192.3 to 379.2 ± 80.16 pg/mL after GnRHa treatment (p = 0.0098). The expression of NRN1 was significantly lower in women with ovarian endometriosis treated with GnRHa. These results suggest that NRN1 may be a biomarker response to the effect of GnRHa treatment for patients with ovarian endometriosis.
Collapse
Affiliation(s)
- Endah Rahmawati
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Obstetrics and Gynecology, Faculty of Medicine Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia.
| | - Wei-Chung Vivian Yang
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yen-Ping Lei
- Department of Obstetrics and Gynecology, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India.
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| | - Chii-Ruey Tzeng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Obstetrics and Gynecology, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| |
Collapse
|
7
|
Chen C, Tian Y, Wang J, Zhang X, Nan L, Dai P, Gao Y, Zheng S, Liu W, Zhang Y. Testosterone propionate can promote effects of acellular nerve allograft-seeded bone marrow mesenchymal stem cells on repairing canine sciatic nerve. J Tissue Eng Regen Med 2019; 13:1685-1701. [PMID: 31267700 DOI: 10.1002/term.2922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/27/2019] [Accepted: 06/30/2019] [Indexed: 12/22/2022]
Abstract
Peripheral human nerves fail to regenerate across long tube implants (>2 cm), and tissue-engineered nerve grafts represent a promising treatment alternative. The present study aims to investigate the testosterone propionate (TP) repair effect of acellular nerve allograft (ANA) seeded with allogeneic bone marrow mesenchymal stem cells (BMSCs) on 3-cm canine sciatic nerve defect. ANA cellularized with allogeneic BMSCs was implanted to the defect, and TP was injected into the lateral crus of the defected leg. The normal group, the autograft group, the ANA + BMSCs group, the ANA group, and the nongrafted group were used as control. Five months postoperatively, dogs in the TP + ANA + BMSCs group were capable of load bearing, normal walking, and skipping, the autograft group and the ANA + BMSCs group demonstrated nearly the same despite a slight limp. The compound muscle action potentials (CMAPs) on the injured side to the uninjured site in the TP + ANA + BMSCs group were significantly higher than that in the ANA + BMSCs group [CMAPs ratio at A: F(3, 20) = 191.40; 0.02, CMAPs ratio at B: F(3, 20) = 43.27; 0.01]. Masson trichrome staining revealed that in the TP + ANA + BMSCs group, both the diameter ratio of the myelinated nerve and the thickness ratio of regenerated myelin sheath were significantly larger than that in the other groups [the diameter of myelinated nerve fibers: F(3, 56) = 13.45; P < .01, the thickness ratio of regenerated myelin sheath: F(3, 56) = 51.25; P < .01]. In conclusion, TP could significantly increase the repairing effects of the ANA + BMSCs group, and their combination was able to repair 3-cm canine sciatic nerve defect. It therefore represents a promising therapeutic approach.
Collapse
Affiliation(s)
- Chen Chen
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China.,Department of General-, Visceral-, Transplantation and Vascular Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Yuanyuan Tian
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China
| | - Jinglu Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China
| | - Xinke Zhang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China
| | - Liangliang Nan
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China.,Institute for Infectious Diseases and Zoonoses, LMU Munich, Munich, Germany
| | - Pengxiu Dai
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China
| | - Yongping Gao
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China
| | - Shuxin Zheng
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China
| | - Wengang Liu
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China
| | - Yihua Zhang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China
| |
Collapse
|
8
|
Mohajeri M, Martín-Jiménez C, Barreto GE, Sahebkar A. Effects of estrogens and androgens on mitochondria under normal and pathological conditions. Prog Neurobiol 2019; 176:54-72. [DOI: 10.1016/j.pneurobio.2019.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 02/23/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023]
|
9
|
Yuan W, Cui L, Li G, Wang N, Zhang P, Zhang Y, Yu N, Wang H, Zhu J, Yang L, Huang J. Recombinant neuritin affects the senescence, apoptosis, proliferation, and migration of rat bone marrow-derived mesenchymal stem cells. Biotechnol Lett 2017; 39:1649-1655. [DOI: 10.1007/s10529-017-2411-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 08/18/2017] [Indexed: 12/01/2022]
|
10
|
Charous SJ, Hotaling JM, Burgess BD, Sappington JM, Park J, Turek G, Foecking EM. Muscle-Nerve-Muscle Grafting for Facial Reanimation in Rats. Ann Otol Rhinol Laryngol 2017; 126:261-267. [DOI: 10.1177/0003489416686587] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: Facial paralysis is a devastating condition leaving patients with a myriad of aesthetic and functional consequences. Muscle-nerve-muscle (MNM) neurotization is a reinnervation technique that involves implanting an autogenous nerve graft as a conduit between an innervated “donor” muscle and a denervated “recipient” muscle. We investigated the use of MNM reinnervation, alone or in combination with electrical stimulation (ES) and testosterone propionate (TP) in comparison to nerve reanastomosis (RE), on functional recovery following rat facial nerve injury. Methods: Thirty-one male, Sprague-Dawley rats were assigned to groups: no graft (control), MNM grafting alone (MNM), MNM grafting with ES and TP (MNM+ES+TP), or RE. Harvested right facial nerve branches were used as the MNM graft. Functional recovery was assessed by behavioral observations and electromyographic recordings. Results: The MNM grafting improved muscle tone and vibrissae movement. The ES+TP treatment further enhanced muscle tone as well as reduced recovery time for coordinated movement in a manner that is comparable to those of RE. Electromyographic recordings demonstrated electrical conductance across all MNM grafts. Conclusion: These data have important implications for patients with unilateral paralysis from facial or laryngeal nerve injury, particularly those who are not candidates for nerve reanastomosis.
Collapse
Affiliation(s)
- Steven J. Charous
- Loyola University of Chicago, Department of Otolaryngology-Head and Neck Surgery, Maywood, Illinois, USA
| | - Jeffery M. Hotaling
- Loyola University of Chicago, Department of Otolaryngology-Head and Neck Surgery, Maywood, Illinois, USA
| | - Bridget D. Burgess
- Loyola University of Chicago, Department of Otolaryngology-Head and Neck Surgery, Maywood, Illinois, USA
| | - Joshua M. Sappington
- Loyola University of Chicago, Department of Otolaryngology-Head and Neck Surgery, Maywood, Illinois, USA
| | - James Park
- Loyola University of Chicago, Stritch School of Medicine, Maywood, Illinois, USA
| | - Grant Turek
- Loyola University of Chicago, Stritch School of Medicine, Maywood, Illinois, USA
| | - Eileen M. Foecking
- Loyola University of Chicago, Department of Otolaryngology-Head and Neck Surgery, Maywood, Illinois, USA
- Edward Hines Jr. VA Hospital, Hines, Illinois, USA
| |
Collapse
|
11
|
Song J, Jung C, Kim OY. The Novel Implication of Androgen in Diabetes-induced Alzheimer's Disease. J Lipid Atheroscler 2017. [DOI: 10.12997/jla.2017.6.2.66] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Gwangju, Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju, Korea
| | - Oh Yoen Kim
- Department of Food Science and Nutrition, Dong-A University, Busan, Korea
| |
Collapse
|
12
|
Karegar M, Mohammadi R. Assessment of neuroregenerative effect of dihydrotestosterone, on peripheral nerve regeneration using allografts: a rat sciatic nerve model. Neurol Res 2015; 37:908-15. [PMID: 26187472 DOI: 10.1179/1743132815y.0000000076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Effects of dihydrotestosterone on nerve allograft were studied in a rat sciatic nerve model. METHODS 30 healthy male white Wistar rats were castrated and randomised into three experimental groups (n = 10): Normal control group (NC), autograft group (AUTO), allograft group (ALLO) and dihydrotestosterone-treated group (ALLO/DHT). In NC group, left sciatic nerve was exposed and left intact. In autograft group, a segment of sciatic nerve was transected and reimplanted reversely. In the ALLO group, the left sciatic nerve was exposed and transected where a 10-mm segment was excised. The same procedure was performed in the ALLO/DHT group. The harvested nerves of the rats of ALLO group were served as allograft for ALLO/DHT group and vice versa. The NC, AUTO and ALLO groups received 300 μl phosphate buffered saline (PBS) intraperitoneally once a day for 1 week and the ALLO/DHT group received 300 μl DHT (1 mg/kg/day) interaperitoneally once a day for 1 week. RESULTS The results showed earlier regeneration of axons in ALLO/DHT than in ALLO group (P < 0.05). Histomorphometic and immunohistochemical studies also showed earlier regeneration of axons in ALLO/DHT than in ALLO group (P < 0.05). DISCUSSIONS Administration of DHT could accelerate functional recovery after nerve allografting in sciatic nerve and may have implications in clinical practice.
Collapse
|
13
|
Wang X, Liu C, Xu F, Cui L, Tan S, Chen R, Yang L, Huang J. Effects of neuritin on the migration, senescence and proliferation of human bone marrow mesenchymal stem cells. ACTA ACUST UNITED AC 2015. [DOI: 10.1515/cmble-2015-0026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractNeuritin is a neurotrophic factor associated with neuroplasticity. Most studies on neuritin focus on the nervous system; however, there has been no comprehensive evaluation of neuritin in non-neuronal cells. In this study, we screened 11 cell lines and found that neuritin was not expressed in bone marrowderived mesenchymal stem cells (BMSCs). Neuritin-expressing BMSCs were obtained by transfection. In the neuritin-expressing BMSC model, we observed significantly greater cell migration and improved anti-senescence protection, in addition to reduced proliferation and viability. In conclusion, neuritin not only plays an important role in the nervous system but also has an effect on the migration, senescence, proliferation, and viability of stem cells. This study provides a theoretical basis for understanding the function of neuritin.
Collapse
|
14
|
Cardona-Rossinyol A, Mir M, Caraballo-Miralles V, Lladó J, Olmos G. Neuroprotective effects of estradiol on motoneurons in a model of rat spinal cord embryonic explants. Cell Mol Neurobiol 2013; 33:421-32. [PMID: 23322321 PMCID: PMC11498032 DOI: 10.1007/s10571-013-9908-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 01/05/2013] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset degenerative disorder characterized by motoneuron death. Clinical and experimental studies in animal models of ALS have found gender differences in the incidence and onset of disease, suggesting that female hormones may play a beneficial role. Cumulative evidence indicates that 17β-estradiol (17βE2) has a neuroprotective role in the central nervous system. We have previously developed a new culture system by using rat spinal cord embryonic explants in which motoneurons have the singularity of migrating outside the spinal cord, growing as a monolayer in the presence of glial cells. In this study, we have validated this new culture system as a useful model for studying neuroprotection by estrogens on spinal cord motoneurons. We show for the first time that spinal cord motoneurons express classical estrogen receptors and that 17βE2 activates, specifically in these cells, the Akt anti-apoptotic signaling pathway and two of their downstream effectors: GSK-3β and Bcl-2. To further validate our system, we demonstrated neuroprotective effects of 17βE2 on spinal cord motoneurons when exposed to the proinflammatory cytokines TNF-α and IFN-γ. These effects of 17βE2 were fully reverted in the presence of the estrogen receptor antagonist ICI 182,780. Our new culture model and the results presented here may provide the basis for further studies on the effects of estrogens, and selective estrogen receptor modulators, on spinal cord motoneurons in the context of ALS or other motoneuron diseases.
Collapse
Affiliation(s)
- Andrea Cardona-Rossinyol
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| | - Margalida Mir
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| | - Víctor Caraballo-Miralles
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| | - Jerònia Lladó
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| | - Gabriel Olmos
- Grup de Neurobiologia Cel·lular, Institut Universitari d’Investigacions en Ciències de la Salut, IUNICS and Departament de Biologia, Universitat de les Illes Balears, Ctra. de Valldemossa km 7.5, Palma de Mallorca, 07122 Spain
| |
Collapse
|
15
|
Xin J, Fargo KN, Tanzer L, Sanders VM, Jones KJ. Immune cell-mediated neuroprotection is independent of estrogen action through estrogen receptor-alpha. Metab Brain Dis 2012; 27:23-8. [PMID: 21975535 DOI: 10.1007/s11011-011-9265-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 09/22/2011] [Indexed: 10/17/2022]
Abstract
It has been well documented that both estrogen and immune cells (CD4+ T cells) mediate neuroprotection in the mouse facial nerve axotomy model. Estrogen has been shown to play an important role in regulating the immune response. However, it is unclear whether immune cell-mediated neuroprotection is dependent on estrogen signaling. In this study, using FACS staining, we confirmed that the majority of CD4+ T cells express high levels of estrogen receptor-alpha (ERα), suggesting that CD4+ T cell-mediated neuroprotection may be modulated by estrogen signaling. We previously found that immunodeficient Rag-2KO mice showed a significant increase in axotomy-induced facial motoneuron death compared to immunocompetent wild-type mice. Therefore, we investigated axotomy-induced facial motoneuron loss in immunodeficient Rag-2KO mice that received 17β-estradiol treatment or adoptive transfer of immune cells from mice lacking functional ERα. Our results indicate that while estradiol treatment failed to rescue facial motoneurons from axotomy-induced cell death in Rag-2KO mice, immune cells lacking ERα successfully restored facial motoneuron survival in Rag-2 KO mice to a wild-type level. Collectively, we concluded that CD4+ T cell-mediated neuroprotection is independent of estrogen action through ERα.
Collapse
Affiliation(s)
- Junping Xin
- Neuroscience Institute, Loyola University Medical Center, 2160 South First Avenue, Maywood, IL 60153, USA.
| | | | | | | | | |
Collapse
|
16
|
Electrical stimulation and testosterone differentially enhance expression of regeneration-associated genes. Exp Neurol 2010; 223:183-91. [DOI: 10.1016/j.expneurol.2009.04.031] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 04/26/2009] [Accepted: 04/28/2009] [Indexed: 02/04/2023]
|
17
|
Fargo KN, Foster AM, Sengelaub DR. Neuroprotective effect of testosterone treatment on motoneuron recruitment following the death of nearby motoneurons. Dev Neurobiol 2009; 69:825-35. [PMID: 19658088 DOI: 10.1002/dneu.20743] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Motoneuron loss is a significant medical problem, capable of causing severe movement disorders or even death. We have previously shown that motoneuron death induces marked dendritic atrophy in surviving nearby motoneurons. Additionally, in quadriceps motoneurons, this atrophy is accompanied by decreases in motor nerve activity. However, treatment with testosterone partially attenuates changes in both the morphology and activation of quadriceps motoneurons. Testosterone has an even larger neuroprotective effect on the morphology of motoneurons of the spinal nucleus of the bulbocavernosus (SNB), in which testosterone treatment can completely prevent dendritic atrophy. The present experiment was performed to determine whether the greater neuroprotective effect of testosterone on SNB motoneuron morphology was accompanied by a greater neuroprotective effect on motor activation. Right side SNB motoneurons were killed by intramuscular injection of cholera toxin-conjugated saporin in adult male Sprague-Dawley rats. Animals were either given Silastic testosterone implants or left untreated. Four weeks later, left side SNB motor activation was assessed with peripheral nerve recording. The death of right side SNB motoneurons resulted in several changes in the electrophysiological response properties of surviving left side SNB motoneurons, including decreased background activity, increased response latency, increased activity duration, and decreased motoneuron recruitment. Treatment with exogenous testosterone attenuated the increase in activity duration and completely prevented the decrease in motoneuron recruitment. These data provide a functional correlate to the known protective effects of testosterone treatment on the morphology of these motoneurons, and further support a role for testosterone as a therapeutic agent in the injured nervous system.
Collapse
Affiliation(s)
- Keith N Fargo
- Research and Development Service, Edward Hines, Jr. VA Hospital, Hines, Illinois 60141, USA.
| | | | | |
Collapse
|
18
|
Loebrich S, Nedivi E. The function of activity-regulated genes in the nervous system. Physiol Rev 2009; 89:1079-103. [PMID: 19789377 DOI: 10.1152/physrev.00013.2009] [Citation(s) in RCA: 166] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The mammalian brain is plastic in the sense that it shows a remarkable capacity for change throughout life. The contribution of neuronal activity to brain plasticity was first recognized in relation to critical periods of development, when manipulating the sensory environment was found to profoundly affect neuronal morphology and receptive field properties. Since then, a growing body of evidence has established that brain plasticity extends beyond development and is an inherent feature of adult brain function, spanning multiple domains, from learning and memory to adaptability of primary sensory maps. Here we discuss evolution of the current view that plasticity of the adult brain derives from dynamic tuning of transcriptional control mechanisms at the neuronal level, in response to external and internal stimuli. We then review the identification of "plasticity genes" regulated by changes in the levels of electrical activity, and how elucidating their cellular functions has revealed the intimate role transcriptional regulation plays in fundamental aspects of synaptic transmission and circuit plasticity that occur in the brain on an every day basis.
Collapse
Affiliation(s)
- Sven Loebrich
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
19
|
Neuroprotective actions of androgens on motoneurons. Front Neuroendocrinol 2009; 30:130-41. [PMID: 19393684 PMCID: PMC2726741 DOI: 10.1016/j.yfrne.2009.04.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Revised: 04/03/2009] [Accepted: 04/14/2009] [Indexed: 01/30/2023]
Abstract
Androgens have a variety of protective and therapeutic effects in both the central and peripheral nervous systems. Here we review these effects as they related specifically to spinal and cranial motoneurons. Early in development, androgens are critical for the formation of important neuromuscular sex differences, decreasing the magnitude of normally occurring cell death in select motoneuron populations. Throughout the lifespan, androgens also protect against motoneuron death caused by axonal injury. Surviving motoneurons also display regressive changes to their neurites as a result of both direct axonal injury and loss of neighboring motoneurons. Androgen treatment enhances the ability of motoneurons to recover from these regressive changes and regenerate both axons and dendrites, restoring normal neuromuscular function. Androgens exert these protective effects by acting through a variety of molecular pathways. Recent work has begun to examine how androgen treatment can interact with other treatment strategies in promoting recovery from motoneuron injury.
Collapse
|
20
|
Pike CJ, Nguyen TVV, Ramsden M, Yao M, Murphy MP, Rosario ER. Androgen cell signaling pathways involved in neuroprotective actions. Horm Behav 2008; 53:693-705. [PMID: 18222446 PMCID: PMC2424283 DOI: 10.1016/j.yhbeh.2007.11.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 10/31/2007] [Accepted: 11/05/2007] [Indexed: 11/15/2022]
Abstract
As a normal consequence of aging in men, testosterone levels significantly decline in both serum and brain. Age-related testosterone depletion results in increased risk of dysfunction and disease in androgen-responsive tissues, including brain. Recent evidence indicates that one deleterious effect of age-related testosterone loss in men is increased risk for Alzheimer's disease (AD). We discuss recent findings from our laboratory and others that identify androgen actions implicated in protecting the brain against neurodegenerative diseases and begin to define androgen cell signaling pathways that underlie these protective effects. Specifically, we focus on the roles of androgens as (1) endogenous negative regulators of beta-amyloid accumulation, a key event in AD pathogenesis, and (2) neuroprotective factors that utilize rapid non-genomic signaling to inhibit neuronal apoptosis. Continued elucidation of cell signaling pathways that contribute to protective actions of androgens should facilitate the development of targeted therapeutic strategies to combat AD and other age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Fargo KN, Galbiati M, Foecking EM, Poletti A, Jones KJ. Androgen regulation of axon growth and neurite extension in motoneurons. Horm Behav 2008; 53:716-28. [PMID: 18387610 PMCID: PMC2408920 DOI: 10.1016/j.yhbeh.2008.01.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 01/11/2008] [Accepted: 01/18/2008] [Indexed: 01/10/2023]
Abstract
Androgens act on the CNS to affect motor function through interaction with a widespread distribution of intracellular androgen receptors (AR). This review highlights our work on androgens and process outgrowth in motoneurons, both in vitro and in vivo. The actions of androgens on motoneurons involve the generation of novel neuronal interactions that are mediated by the induction of androgen-dependent neurite or axonal outgrowth. Here, we summarize the experimental evidence for the androgenic regulation of the extension and regeneration of motoneuron neurites in vitro using cultured immortalized motoneurons, and axons in vivo using the hamster facial nerve crush paradigm. We place particular emphasis on the relevance of these effects to SBMA and peripheral nerve injuries.
Collapse
Affiliation(s)
- Keith N Fargo
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Chicago, Maywood, Illinois 60153, USA.
| | | | | | | | | |
Collapse
|
22
|
Richter HE, Diokno A, Kenton K, Norton P, Albo M, Kraus S, Moalli P, Chai TC, Zimmern P, Litman H, Tennstedt S. Predictors of treatment failure 24 months after surgery for stress urinary incontinence. J Urol 2008; 179:1024-30. [PMID: 18206917 DOI: 10.1016/j.juro.2007.10.074] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Indexed: 11/30/2022]
Abstract
PURPOSE We identified baseline demographic and clinical factors associated with treatment failure after surgical treatment of stress urinary incontinence. MATERIALS AND METHODS Data were obtained from 655 women randomized to Burch colposuspension or autologous rectus sling. Of those, 543 (83%) had stress failure status assessed at 24 months (269 Burch, 274 sling). Stress failure (261) was defined as self-report of stress urinary incontinence by the Medical, Epidemiological, and Social Aspects of Aging questionnaire, positive stress test or re-treatment for stress urinary incontinence. Nonstress failure (66) was defined as positive 24-hour pad test (more than 15 ml) or any incontinent episodes by 3-day voiding diary with none of the 3 criteria for stress failure. Subjects not meeting any failure criteria were considered a treatment success (185). Adjusting for surgical treatment group and clinical site, logistic regression models were developed to predict the probability of treatment failure. RESULTS Severity of urge incontinence symptoms (p = 0.041), prolapse stage (p = 0.013), and being postmenopausal without hormone therapy (p = 0.023) were significant predictors for stress failure. Odds of nonstress failure quadrupled for every 10-point increase in Medical, Epidemiological, and Social Aspects of Aging questionnaire urge score (OR 3.93 CI 1.45, 10.65) and decreased more than 2 times for every 10-point increase in stress score (OR 0.36, CI 0.16, 0.84). The associations of risk factors and failure remained similar regardless of surgical group. CONCLUSIONS Two years after surgery, risk factors for stress failure are similar after Burch and sling procedures and include greater baseline urge incontinence symptoms, more advanced prolapse, and menopausal not on hormone replacement therapy. Higher urge scores predicted failure by nonstress specific outcomes.
Collapse
Affiliation(s)
- Holly E Richter
- University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Cappelletti G, Galbiati M, Ronchi C, Maggioni MG, Onesto E, Poletti A. Neuritin (cpg15) enhances the differentiating effect of NGF on neuronal PC12 cells. J Neurosci Res 2008; 85:2702-13. [PMID: 17335086 DOI: 10.1002/jnr.21235] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Neuritin is a small, highly conserved GPI-anchored protein involved in neurite outgrowth. We have analyzed the involvement of neuritin in NGF-induced differentiation of PC12 cells by investigating the time-course of neuritin expression, the effects of its overexpression or silencing, and the possible mechanisms of its regulation and action. Real-time PCR analysis has shown that neuritin gene is upregulated by NGF in PC12 cells hours before neurite outgrowth becomes appreciable. PC12 cells transfected with a plasmid expressing neuritin display a significant increase in the response to NGF: 1) in the levels of SMI312 positive phosphorylated neurofilament proteins (markers for axonal processes) and tyrosine hydroxylase; 2) in the percentage of cells bearing neurites; as well as 3) in the average length of neurites when compared to control cells. On the contrary, neuritin silencing significantly reduces neurite outgrowth. These data suggest that neuritin is a modulator of NGF-induced neurite extension in PC12 cells. We also showed that neuritin potentiated the NGF-induced differentiation of PC12 cells without affecting TrkA or EGF receptor mRNAs expression. Moreover, the S-methylisothiourea (MIU), a potent inhibitor of inducible nitric oxide synthases, partially counteracts the NGF-mediated neuritin induction. These data suggest that NGF regulates neuritin expression in PC12 cells via the signaling pathway triggered by NO. This study reports the first evidence that neuritin plays a role in modulating neurite outgrowth during the progression of NGF-induced differentiation of PC12 cells. PC12 cells could be considered a valuable model to unravel the mechanism of action of neuritin on neurite outgrowth. (c) 2007 Wiley-Liss, Inc.
Collapse
|
24
|
Cantallops I, Cline HT. Rapid activity-dependent delivery of the neurotrophic protein CPG15 to the axon surface of neurons in intactXenopus tadpoles. Dev Neurobiol 2008; 68:744-59. [DOI: 10.1002/dneu.20529] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
25
|
Tetzlaff J, Tanzer L, Jones KJ. Cellular localization of androgen and estrogen receptors in mouse-derived motoneuron hybrid cells and mouse facial motoneurons. Dev Neurobiol 2007; 67:1362-70. [PMID: 17638386 DOI: 10.1002/dneu.20505] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The ability of gonadal steroid hormones to augment axonal regeneration after peripheral nerve injury has been well established in rat and hamster motoneuron systems, and provides a foundation for the use of these agents as neurotherapeutics. With the advent of mouse genetics and the availability of transgenic and knockout mice, the use of mice in studies of neuroprotection is growing. It has recently been demonstrated that both androgens and estrogens rescue motoneurons (MN) from injury in mouse-derived motoneuron hybrid cells in vitro and mouse facial motoneurons (FMN) in vivo (Tetzlaff et al. [2006] J Mol Neurosci 28:53-64). To elucidate the molecular mechanisms of these effects, the present study examined the cellular localization of androgen and estrogen receptors in mouse MN in vitro and in vivo. Immunoblotting and immunocytochemistry studies established the presence of androgen receptor (AR) and estrogen receptor alpha/beta in immortalized mouse motoneuron hybrid cells and AR and estrogen receptor alpha in mouse FMN.
Collapse
Affiliation(s)
- Julie Tetzlaff
- Neuroscience Graduate Program, Loyola University Chicago, Maywood, Illinois 60153, USA
| | | | | |
Collapse
|