1
|
Blackmore DG, Waters MJ. The multiple roles of GH in neural ageing and injury. Front Neurosci 2023; 17:1082449. [PMID: 36960169 PMCID: PMC10027725 DOI: 10.3389/fnins.2023.1082449] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/17/2023] [Indexed: 03/09/2023] Open
Abstract
Advanced age is typically associated with a decrease in cognitive function including impairment in the formation and retention of new memories. The hippocampus is critical for learning and memory, especially spatial learning, and is particularly affected by ageing. With advanced age, multiple neural components can be detrimentally affected including a reduction in the number of neural stem and precursor cells, a decrease in the formation of adult born neurons (neurogenesis), and deficits in neural circuitry, all of which ultimately contribute to impaired cognitive function. Importantly, physical exercise has been shown to ameliorate many of these impairments and is able to improve learning and memory. Relevantly, growth hormone (GH) is an important protein hormone that decreases with ageing and increases following physical exercise. Originally described due to its role in longitudinal growth, GH has now been identified to play several additional key roles, especially in relation to the brain. Indeed, the regular decrease in GH levels following puberty is one of the most well documented components of neuroendocrine ageing. Growth hormone deficiency (GHD) has been described to have adverse effects on brain function, which can be ameliorated via GH replacement therapy. Physical exercise has been shown to increase circulating GH levels. Furthermore, we recently demonstrated the increase in exercise-mediated GH is critical for improved cognitive function in the aged mouse. Here we examine the multiple roles that GH plays, particularly in the aged brain and following trauma, irradiation and stroke, and how increasing GH levels can ameliorate deficits in cognition.
Collapse
Affiliation(s)
- Daniel G. Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Michael J. Waters
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- *Correspondence: Michael J. Waters,
| |
Collapse
|
2
|
Juárez-Aguilar E, Olivares-Hernández JD, Regalado-Santiago C, García-García F. The role of growth hormone in hippocampal function. VITAMINS AND HORMONES 2021; 118:289-313. [PMID: 35180930 DOI: 10.1016/bs.vh.2021.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Growth hormone is a multifunctional molecule with broad cellular targets. This pituitary hormone is currently used as a therapeutic agent against several brain injuries due to its neurotrophic activity. The hippocampus is one of the brain regions where the growth hormone plays a role in normal and pathologic conditions. This brain structure is associated with several cognitive functions such as learning, memory, and mood, which are frequently affected by brain traumatism. The present chapter describes the experimental and clinical evidence that supports a central role of growth hormone in the hippocampus functionality.
Collapse
Affiliation(s)
- Enrique Juárez-Aguilar
- Departmento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Veracruz, Mexico.
| | - Juan David Olivares-Hernández
- Laboratorio D-01, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | | | - Fabio García-García
- Departmento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Veracruz, Mexico
| |
Collapse
|
3
|
Gasco V, Cambria V, Bioletto F, Ghigo E, Grottoli S. Traumatic Brain Injury as Frequent Cause of Hypopituitarism and Growth Hormone Deficiency: Epidemiology, Diagnosis, and Treatment. Front Endocrinol (Lausanne) 2021; 12:634415. [PMID: 33790864 PMCID: PMC8005917 DOI: 10.3389/fendo.2021.634415] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI)-related hypopituitarism has been recognized as a clinical entity for more than a century, with the first case being reported in 1918. However, during the 20th century hypopituitarism was considered only a rare sequela of TBI. Since 2000 several studies strongly suggest that TBI-mediated pituitary hormones deficiency may be more frequent than previously thought. Growth hormone deficiency (GHD) is the most common abnormality, followed by hypogonadism, hypothyroidism, hypocortisolism, and diabetes insipidus. The pathophysiological mechanisms underlying pituitary damage in TBI patients include a primary injury that may lead to the direct trauma of the hypothalamus or pituitary gland; on the other hand, secondary injuries are mainly related to an interplay of a complex and ongoing cascade of specific molecular/biochemical events. The available data describe the importance of GHD after TBI and its influence in promoting neurocognitive and behavioral deficits. The poor outcomes that are seen with long standing GHD in post TBI patients could be improved by GH treatment, but to date literature data on the possible beneficial effects of GH replacement therapy in post-TBI GHD patients are currently scarce and fragmented. More studies are needed to further characterize this clinical syndrome with the purpose of establishing appropriate standards of care. The purpose of this review is to summarize the current state of knowledge about post-traumatic GH deficiency.
Collapse
|
4
|
Baltazar-Lara R, Ávila-Mendoza J, Martínez-Moreno CG, Carranza M, Pech-Pool S, Vázquez-Martínez O, Díaz-Muñoz M, Luna M, Arámburo C. Neuroprotective Effects of Growth Hormone (GH) and Insulin-Like Growth Factor Type 1 (IGF-1) after Hypoxic-Ischemic Injury in Chicken Cerebellar Cell Cultures. Int J Mol Sci 2020; 22:ijms22010256. [PMID: 33383827 PMCID: PMC7795313 DOI: 10.3390/ijms22010256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
It has been reported that growth hormone (GH) and insulin-like growth factor 1 (IGF-1) exert protective and regenerative actions in response to neural damage. It is also known that these peptides are expressed locally in nervous tissues. When the central nervous system (CNS) is exposed to hypoxia-ischemia (HI), both GH and IGF-1 are upregulated in several brain areas. In this study, we explored the neuroprotective effects of GH and IGF-1 administration as well as the involvement of these endogenously expressed hormones in embryonic chicken cerebellar cell cultures exposed to an acute HI injury. To induce neural damage, primary cultures were first incubated under hypoxic-ischemic (<5% O2, 1g/L glucose) conditions for 12 h (HI), and then incubated under normal oxygenation and glucose conditions (HI + Ox) for another 24 h. GH and IGF-1 were added either during or after HI, and their effect upon cell viability, apoptosis, or necrosis was evaluated. In comparison with normal controls (Nx, 100%), a significant decrease of cell viability (54.1 ± 2.1%) and substantial increases in caspase-3 activity (178.6 ± 8.7%) and LDH release (538.7 ± 87.8%) were observed in the HI + Ox group. On the other hand, both GH and IGF-1 treatments after injury (HI + Ox) significantly increased cell viability (77.2 ± 4.3% and 72.3 ± 3.9%, respectively) and decreased both caspase-3 activity (118.2 ± 3.8% and 127.5 ± 6.6%, respectively) and LDH release (180.3 ± 21.8% and 261.6 ± 33.9%, respectively). Incubation under HI + Ox conditions provoked an important increase in the local expression of GH (3.2-fold) and IGF-1 (2.5-fold) mRNAs. However, GH gene silencing with a specific small-interfering RNAs (siRNAs) decreased both GH and IGF-1 mRNA expression (1.7-fold and 0.9-fold, respectively) in the HI + Ox group, indicating that GH regulates IGF-1 expression under these incubation conditions. In addition, GH knockdown significantly reduced cell viability (35.9 ± 2.1%) and substantially increased necrosis, as determined by LDH release (1011 ± 276.6%). In contrast, treatments with GH and IGF-1 stimulated a partial recovery of cell viability (45.2 ± 3.7% and 53.7 ± 3.2%) and significantly diminished the release of LDH (320.1 ± 25.4% and 421.7 ± 62.2%), respectively. Our results show that GH, either exogenously administered and/or locally expressed, can act as a neuroprotective factor in response to hypoxic-ischemic injury, and that this effect may be mediated, at least partially, through IGF-1 expression.
Collapse
Affiliation(s)
- Rosario Baltazar-Lara
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (R.B.-L.); (J.Á.-M.); (C.G.M.-M.); (M.C.); (S.P.-P.); (O.V.-M.); (M.D.-M.)
| | - José Ávila-Mendoza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (R.B.-L.); (J.Á.-M.); (C.G.M.-M.); (M.C.); (S.P.-P.); (O.V.-M.); (M.D.-M.)
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Carlos G. Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (R.B.-L.); (J.Á.-M.); (C.G.M.-M.); (M.C.); (S.P.-P.); (O.V.-M.); (M.D.-M.)
| | - Martha Carranza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (R.B.-L.); (J.Á.-M.); (C.G.M.-M.); (M.C.); (S.P.-P.); (O.V.-M.); (M.D.-M.)
| | - Santiago Pech-Pool
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (R.B.-L.); (J.Á.-M.); (C.G.M.-M.); (M.C.); (S.P.-P.); (O.V.-M.); (M.D.-M.)
| | - Olivia Vázquez-Martínez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (R.B.-L.); (J.Á.-M.); (C.G.M.-M.); (M.C.); (S.P.-P.); (O.V.-M.); (M.D.-M.)
| | - Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (R.B.-L.); (J.Á.-M.); (C.G.M.-M.); (M.C.); (S.P.-P.); (O.V.-M.); (M.D.-M.)
| | - Maricela Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (R.B.-L.); (J.Á.-M.); (C.G.M.-M.); (M.C.); (S.P.-P.); (O.V.-M.); (M.D.-M.)
- Correspondence: (M.L.); (C.A.); Tel.: +52-55-5623-4066 (M.L.); +52-55-5623-4065 (C.A.); Fax: +52-55-5623-4005 (M.L. & C.A.)
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (R.B.-L.); (J.Á.-M.); (C.G.M.-M.); (M.C.); (S.P.-P.); (O.V.-M.); (M.D.-M.)
- Correspondence: (M.L.); (C.A.); Tel.: +52-55-5623-4066 (M.L.); +52-55-5623-4065 (C.A.); Fax: +52-55-5623-4005 (M.L. & C.A.)
| |
Collapse
|
5
|
Martínez-Moreno CG, Arámburo C. Growth hormone (GH) and synaptogenesis. VITAMINS AND HORMONES 2020; 114:91-123. [PMID: 32723552 DOI: 10.1016/bs.vh.2020.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Growth hormone (GH) is known to exert several roles during development and function of the nervous system. Initially, GH was exclusively considered a pituitary hormone that regulates body growth and metabolism, but now its alternative extrapituitary production and pleiotropic functions are widely accepted. Through excess and deficit models, the critical role of GH in nervous system development and adult brain function has been extensively demonstrated. Moreover, neurotrophic actions of GH in neural tissues include pro-survival effects, neuroprotection, axonal growth, synaptogenesis, neurogenesis and neuroregeneration. The positive effects of GH upon memory, behavior, mood, sensorimotor function and quality of life, clearly implicate a beneficial action in synaptic physiology. Experimental and clinical evidence about GH actions in synaptic function modulation, protection and restoration are revised in this chapter.
Collapse
Affiliation(s)
- Carlos G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México.
| |
Collapse
|
6
|
Fleming T, Balderas-Márquez JE, Epardo D, Ávila-Mendoza J, Carranza M, Luna M, Harvey S, Arámburo C, Martínez-Moreno CG. Growth Hormone Neuroprotection Against Kainate Excitotoxicity in the Retina is Mediated by Notch/PTEN/Akt Signaling. Invest Ophthalmol Vis Sci 2020; 60:4532-4547. [PMID: 31675424 DOI: 10.1167/iovs.19-27473] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose In the retina, growth hormone (GH) promotes axonal growth, synaptic restoration, and protective actions against excitotoxicity. Notch signaling pathway is critical for neural development and participates in the retinal neuroregenerative process. We investigated the interaction of GH with Notch signaling pathway during its neuroprotective effect against excitotoxic damage in the chicken retina. Methods Kainate (KA) was used as excitotoxic agent and changes in the mRNA expression of several signaling markers were determined by qPCR. Also, changes in phosphorylation and immunoreactivity were determined by Western blotting. Histology and immunohistochemistry were performed for morphometric analysis. Overexpression of GH was performed in the quail neuroretinal-derived immortalized cell line (QNR/D) cell line. Exogenous GH was administered to retinal primary cell cultures to study the activation of signaling pathways. Results KA disrupted the retinal cytoarchitecture and induced significant cell loss in several retinal layers, but the coaddition of GH effectively prevented these adverse effects. We showed that GH upregulates the Notch signaling pathway during neuroprotection leading to phosphorylation of the PI3K/Akt signaling pathways through downregulation of PTEN. In contrast, cotreatment of GH with the Notch signaling inhibitor, DAPT, prevented its neuroprotective effect against KA. We identified binding sites in Notch1 and Notch2 genes for STAT5. Also, GH prevented Müller cell transdifferentiation and downregulated Sox2, FGF2, and PCNA after cotreatment with KA. Additionally, GH modified TNF receptors immunoreactivity suggesting anti-inflammatory actions. Conclusions Our data indicate that the neuroprotective effects of GH against KA injury in the retina are mediated through the regulation of Notch signaling. Additionally, anti-inflammatory and antiproliferative effects were observed.
Collapse
Affiliation(s)
- Thomas Fleming
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México.,Department of Physiology, University of Alberta, Edmonton, Canada
| | - Jerusa E Balderas-Márquez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - David Epardo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - José Ávila-Mendoza
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
| | - Martha Carranza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Maricela Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Steve Harvey
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Carlos G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| |
Collapse
|
7
|
Posavi M, Diaz-Ortiz M, Liu B, Swanson CR, Skrinak RT, Hernandez-Con P, Amado DA, Fullard M, Rick J, Siderowf A, Weintraub D, McCluskey L, Trojanowski JQ, Dewey RB, Huang X, Chen-Plotkin AS. Characterization of Parkinson's disease using blood-based biomarkers: A multicohort proteomic analysis. PLoS Med 2019; 16:e1002931. [PMID: 31603904 PMCID: PMC6788685 DOI: 10.1371/journal.pmed.1002931] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative disease affecting about 5 million people worldwide with no disease-modifying therapies. We sought blood-based biomarkers in order to provide molecular characterization of individuals with PD for diagnostic confirmation and prediction of progression. METHODS AND FINDINGS In 141 plasma samples (96 PD, 45 neurologically normal control [NC] individuals; 45.4% female, mean age 70.0 years) from a longitudinally followed Discovery Cohort based at the University of Pennsylvania (UPenn), we measured levels of 1,129 proteins using an aptamer-based platform. We modeled protein plasma concentration (log10 of relative fluorescence units [RFUs]) as the effect of treatment group (PD versus NC), age at plasma collection, sex, and the levodopa equivalent daily dose (LEDD), deriving first-pass candidate protein biomarkers based on p-value for PD versus NC. These candidate proteins were then ranked by Stability Selection. We confirmed findings from our Discovery Cohort in a Replication Cohort of 317 individuals (215 PD, 102 NC; 47.9% female, mean age 66.7 years) from the multisite, longitudinally followed National Institute of Neurological Disorders and Stroke Parkinson's Disease Biomarker Program (PDBP) Cohort. Analytical approach in the Replication Cohort mirrored the approach in the Discovery Cohort: each protein plasma concentration (log10 of RFU) was modeled as the effect of group (PD versus NC), age at plasma collection, sex, clinical site, and batch. Of the top 10 proteins from the Discovery Cohort ranked by Stability Selection, four associations were replicated in the Replication Cohort. These blood-based biomarkers were bone sialoprotein (BSP, Discovery false discovery rate [FDR]-corrected p = 2.82 × 10-2, Replication FDR-corrected p = 1.03 × 10-4), osteomodulin (OMD, Discovery FDR-corrected p = 2.14 × 10-2, Replication FDR-corrected p = 9.14 × 10-5), aminoacylase-1 (ACY1, Discovery FDR-corrected p = 1.86 × 10-3, Replication FDR-corrected p = 2.18 × 10-2), and growth hormone receptor (GHR, Discovery FDR-corrected p = 3.49 × 10-4, Replication FDR-corrected p = 2.97 × 10-3). Measures of these proteins were not significantly affected by differences in sample handling, and they did not change comparing plasma samples from 10 PD participants sampled both on versus off dopaminergic medication. Plasma measures of OMD, ACY1, and GHR differed in PD versus NC but did not differ between individuals with amyotrophic lateral sclerosis (ALS, n = 59) versus NC. In the Discovery Cohort, individuals with baseline levels of GHR and ACY1 in the lowest tertile were more likely to progress to mild cognitive impairment (MCI) or dementia in Cox proportional hazards analyses adjusting for age, sex, and disease duration (hazard ratio [HR] 2.27 [95% CI 1.04-5.0, p = 0.04] for GHR, and HR 3.0 [95% CI 1.24-7.0, p = 0.014] for ACY1). GHR's association with cognitive decline was confirmed in the Replication Cohort (HR 3.6 [95% CI 1.20-11.1, p = 0.02]). The main limitations of this study were its reliance on the aptamer-based platform for protein measurement and limited follow-up time available for some cohorts. CONCLUSIONS In this study, we found that the blood-based biomarkers BSP, OMD, ACY1, and GHR robustly associated with PD across multiple clinical sites. Our findings suggest that biomarkers based on a peripheral blood sample may be developed for both disease characterization and prediction of future disease progression in PD.
Collapse
Affiliation(s)
- Marijan Posavi
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Maria Diaz-Ortiz
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Benjamine Liu
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christine R Swanson
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- National Institute of Neurological Disease and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - R Tyler Skrinak
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Pilar Hernandez-Con
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Defne A Amado
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michelle Fullard
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jacqueline Rick
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Andrew Siderowf
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel Weintraub
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Leo McCluskey
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Richard B Dewey
- Department of Neurology and Neurotherapeutics, Clinical Center for Movement Disorders at the University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Xuemei Huang
- Department of Neurology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
8
|
Martinez-Moreno CG, Epardo D, Balderas-Márquez JE, Fleming T, Carranza M, Luna M, Harvey S, Arámburo C. Regenerative Effect of Growth Hormone (GH) in the Retina after Kainic Acid Excitotoxic Damage. Int J Mol Sci 2019; 20:E4433. [PMID: 31509934 PMCID: PMC6770150 DOI: 10.3390/ijms20184433] [Citation(s) in RCA: 12] [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: 08/12/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
In addition to its role as an endocrine messenger, growth hormone (GH) also acts as a neurotrophic factor in the central nervous system (CNS), whose effects are involved in neuroprotection, axonal growth, and synaptogenic modulation. An increasing amount of clinical evidence shows a beneficial effect of GH treatment in patients with brain trauma, stroke, spinal cord injury, impaired cognitive function, and neurodegenerative processes. In response to injury, Müller cells transdifferentiate into neural progenitors and proliferate, which constitutes an early regenerative process in the chicken retina. In this work, we studied the long-term protective effect of GH after causing severe excitotoxic damage in the retina. Thus, an acute neural injury was induced via the intravitreal injection of kainic acid (KA, 20 µg), which was followed by chronic administration of GH (10 injections [300 ng] over 21 days). Damage provoked a severe disruption of several retinal layers. However, in KA-damaged retinas treated with GH, we observed a significant restoration of the inner plexiform layer (IPL, 2.4-fold) and inner nuclear layer (INL, 1.5-fold) thickness and a general improvement of the retinal structure. In addition, we also observed an increase in the expression of several genes involved in important regenerative pathways, including: synaptogenic markers (DLG1, NRXN1, GAP43); glutamate receptor subunits (NR1 and GRIK4); pro-survival factors (BDNF, Bcl-2 and TNF-R2); and Notch signaling proteins (Notch1 and Hes5). Interestingly, Müller cell transdifferentiation markers (Sox2 and FGF2) were upregulated by this long-term chronic GH treatment. These results are consistent with a significant increase in the number of BrdU-positive cells observed in the KA-damaged retina, which was induced by GH administration. Our data suggest that GH is able to facilitate the early proliferative response of the injured retina and enhance the regeneration of neurite interconnections.
Collapse
Affiliation(s)
- Carlos G Martinez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico.
| | - David Epardo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| | - Jerusa E Balderas-Márquez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico.
| | - Thomas Fleming
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico.
| | - Martha Carranza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico.
| | - Maricela Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico.
| | - Steve Harvey
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico.
| |
Collapse
|
9
|
Chen Y, Tsai YH, Tseng BJ, Tseng SH. Influence of Growth Hormone and Glutamine on Intestinal Stem Cells: A Narrative Review. Nutrients 2019; 11:E1941. [PMID: 31426533 PMCID: PMC6724402 DOI: 10.3390/nu11081941] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/21/2022] Open
Abstract
Growth hormone (GH) and glutamine (Gln) stimulate the growth of the intestinal mucosa. GH activates the proliferation of intestinal stem cells (ISCs), enhances the formation of crypt organoids, increases ISC stemness markers in the intestinal organoids, and drives the differentiation of ISCs into Paneth cells and enterocytes. Gln enhances the proliferation of ISCs and increases crypt organoid formation; however, it mainly acts on the post-proliferation activity of ISCs to maintain the stability of crypt organoids and the intestinal mucosa, as well as to stimulate the differentiation of ISCs into goblet cells and possibly Paneth cells and enteroendocrine cells. Since GH and Gln have differential effects on ISCs. Their use in combination may have synergistic effects on ISCs. In this review, we summarize the evidence of the actions of GH and/or Gln on crypt cells and ISCs in the literature. Overall, most studies demonstrated that GH and Gln in combination exerted synergistic effects to activate the proliferation of crypt cells and ISCs and enhance crypt organoid formation and mucosal growth. This treatment influenced the proliferation of ISCs to a similar degree as GH treatment alone and the differentiation of ISCs to a similar degree as Gln treatment alone.
Collapse
Affiliation(s)
- Yun Chen
- Department of Surgery, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei 220, Taiwan
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan 320, Taiwan
| | - Ya-Hui Tsai
- Department of Surgery, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei 220, Taiwan
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan 320, Taiwan
| | - Bor-Jiun Tseng
- Department of Surgery, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei 220, Taiwan
| | - Sheng-Hong Tseng
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan.
| |
Collapse
|
10
|
Cognitive Evolution of a Patient Who Suffered a Subarachnoid Haemorrhage Eight Years Ago, after Being Treated with Growth Hormone, Melatonin and Neurorehabilitation. REPORTS 2018. [DOI: 10.3390/reports1010002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
11
|
Bianchi VE, Locatelli V, Rizzi L. Neurotrophic and Neuroregenerative Effects of GH/IGF1. Int J Mol Sci 2017; 18:ijms18112441. [PMID: 29149058 PMCID: PMC5713408 DOI: 10.3390/ijms18112441] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/12/2022] Open
Abstract
Introduction. Human neurodegenerative diseases increase progressively with age and present a high social and economic burden. Growth hormone (GH) and insulin-like growth factor-1 (IGF-1) are both growth factors exerting trophic effects on neuronal regeneration in the central nervous system (CNS) and peripheral nervous system (PNS). GH and IGF-1 stimulate protein synthesis in neurons, glia, oligodendrocytes, and Schwann cells, and favor neuronal survival, inhibiting apoptosis. This study aims to evaluate the effect of GH and IGF-1 on neurons, and their possible therapeutic clinical applications on neuron regeneration in human subjects. Methods. In the literature, we searched the clinical trials and followed up studies in humans, which have evaluated the effect of GH/IGF-1 on CNS and PNS. The following keywords have been used: “GH/IGF-1” associated with “neuroregeneration”, “amyotrophic lateral sclerosis”, “Alzheimer disease”, “Parkinson’s disease”, “brain”, and “neuron”. Results. Of the retrieved articles, we found nine articles about the effect of GH in healthy patients who suffered from traumatic brain injury (TBI), and six studies (four using IGF-1 and two GH therapy) in patients with amyotrophic lateral sclerosis (ALS). The administration of GH in patients after TBI showed a significantly positive recovery of brain and mental function. Treatment with GH and IGF-1 therapy in ALS produced contradictory results. Conclusions. Although strong findings have shown the positive effects of GH/IGF-1 administration on neuroregeneration in animal models, a very limited number of clinical studies have been conducted in humans. GH/IGF-1 therapy had different effects in patients with TBI, evidencing a high recovery of neurons and clinical outcome, while in ALS patients, the results are contradictory. More complex clinical protocols are necessary to evaluate the effect of GH/IGF-1 efficacy in neurodegenerative diseases. It seems evident that GH and IGF-1 therapy favors the optimal recovery of neurons when a consistent residual activity is still present. Furthermore, the effect of GH/IGF-1 could be mediated by, or be overlapped with that of other hormones, such as estradiol and testosterone.
Collapse
Affiliation(s)
- Vittorio Emanuele Bianchi
- Endocrinology and Metabolism, Clinical Center Stella Maris, Strada Rovereta, 42-47891 Falciano, San Marino.
| | - Vittorio Locatelli
- School of Medicine and Surgery, University of Milano-Bicocca via Cadore, 48-20900 Monza Brianza, Italy.
| | - Laura Rizzi
- Molecular Biology, School of Medicine and Surgery, University of Milano-Bicocca, via Cadore, 48-20900 Monza Brianza, Italy.
| |
Collapse
|
12
|
Kawa MP, Stecewicz I, Piecyk K, Paczkowska E, Rogińska D, Sobuś A, Łuczkowska K, Pius-Sadowska E, Gawrych E, Petriczko E, Walczak M, Machaliński B. The Impact of Growth Hormone Therapy on the Apoptosis Assessment in CD34+ Hematopoietic Cells from Children with Growth Hormone Deficiency. Int J Mol Sci 2017; 18:ijms18010111. [PMID: 28067847 PMCID: PMC5297745 DOI: 10.3390/ijms18010111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 12/10/2016] [Accepted: 12/21/2016] [Indexed: 01/07/2023] Open
Abstract
Growth hormone (GH) modulates hematopoietic cell homeostasis and is associated with apoptosis control, but with limited mechanistic insights. Aim of the study was to determine whether GH therapeutic supplementation (GH-TS) could affect apoptosis of CD34+ cells enriched in hematopoietic progenitor cells of GH deficient (GHD) children. CD34+ cells from peripheral blood of 40 GHD children were collected before and in 3rd and 6th month of GH-TS and compared to 60 controls adjusted for bone age, sex, and pubertal development. Next, apoptosis assessment via different molecular techniques was performed. Finally, to comprehensively characterize apoptosis process, global gene expression profile was determined using genome-wide RNA microarray technology. Results showed that GH-TS significantly reduced spontaneous apoptosis in CD34+ cells (p < 0.01) and results obtained using different methods to detect early and late apoptosis in analyzed cells population were consistent. GH-TS was also associated with significant downregulation of several members of TNF-alpha superfamily and other genes associated with apoptosis and stress response. Moreover, the significant overexpression of cyto-protective and cell cycle-associated genes was detected. These findings suggest that recombinant human GH has a direct anti-apoptotic activity in hematopoietic CD34+ cells derived from GHD subjects in course of GH-TS.
Collapse
Affiliation(s)
- Miłosz Piotr Kawa
- Department of General Pathology, Pomeranian Medical University in Szczecin, 72 Powstancow Wlkp. Street, 70-111 Szczecin, Poland.
| | - Iwona Stecewicz
- Department of Pediatrics, Endocrinology, Diabetology, Metabolic Diseases and Cardiology of the Developmental Age, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland.
| | - Katarzyna Piecyk
- Department of General Pathology, Pomeranian Medical University in Szczecin, 72 Powstancow Wlkp. Street, 70-111 Szczecin, Poland.
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, 72 Powstancow Wlkp. Street, 70-111 Szczecin, Poland.
| | - Dorota Rogińska
- Department of General Pathology, Pomeranian Medical University in Szczecin, 72 Powstancow Wlkp. Street, 70-111 Szczecin, Poland.
| | - Anna Sobuś
- Department of General Pathology, Pomeranian Medical University in Szczecin, 72 Powstancow Wlkp. Street, 70-111 Szczecin, Poland.
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, 72 Powstancow Wlkp. Street, 70-111 Szczecin, Poland.
| | - Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, 72 Powstancow Wlkp. Street, 70-111 Szczecin, Poland.
| | - Elżbieta Gawrych
- Department of Pediatric and Oncological Surgery, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland.
| | - Elżbieta Petriczko
- Department of Pediatrics, Endocrinology, Diabetology, Metabolic Diseases and Cardiology of the Developmental Age, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland.
| | - Mieczysław Walczak
- Department of Pediatrics, Endocrinology, Diabetology, Metabolic Diseases and Cardiology of the Developmental Age, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland.
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University in Szczecin, 72 Powstancow Wlkp. Street, 70-111 Szczecin, Poland.
| |
Collapse
|
13
|
Devesa J, Lema H, Zas E, Munín B, Taboada P, Devesa P. Learning and Memory Recoveries in a Young Girl Treated with Growth Hormone and Neurorehabilitation. J Clin Med 2016; 5:jcm5020014. [PMID: 26821051 PMCID: PMC4773770 DOI: 10.3390/jcm5020014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/16/2016] [Accepted: 01/19/2016] [Indexed: 11/16/2022] Open
Abstract
Background-To describe the results obtained after treating a non growth hormone-deficient 10-year-old girl who suffered asphyxia during delivery, resulting in important cognitive deficits, with growth hormone (GH) and neurorehabilitation. Methods-GH was administered (mg/day) at doses of 0.5 over three months followed by 0.9, every two weeks over three months, and then alternating 1.2 three days/week and 0.3 two days/week. Neurorehabilitation consisted of daily sessions of neurostimulation, speech therapy, occupational therapy and auditive stimulation. Treatment lasted nine months. Results-Scores obtained in all the areas treated showed that, at discharge, the patient clearly increased her cognitive abilities, memory and language competence index; her intelligence quotient score increased from 51 to 80, and the index of functional independence measure reached a value of 120 over 126 (maximal value). Conclusions-This case suggests that GH administration may play a role in improving cognitive deficits during neurorehabilitation in children with brain damage suffered during delivery. This agrees with the known effects of GH on cognition.
Collapse
Affiliation(s)
- Jesús Devesa
- Medical Centre Foltra, Travesía de Montouto 24, Teo 15886, Spain.
- Department of Physiology, School of Medicine, University of Santiago de Compostela, Santiago de Compostela 15710, Spain.
| | - Hortensia Lema
- Medical Centre Foltra, Travesía de Montouto 24, Teo 15886, Spain.
| | - Eva Zas
- Medical Centre Foltra, Travesía de Montouto 24, Teo 15886, Spain.
| | - Borja Munín
- Medical Centre Foltra, Travesía de Montouto 24, Teo 15886, Spain.
| | - Pilar Taboada
- Medical Centre Foltra, Travesía de Montouto 24, Teo 15886, Spain.
| | - Pablo Devesa
- Medical Centre Foltra, Travesía de Montouto 24, Teo 15886, Spain.
| |
Collapse
|
14
|
Regalado-Santiago C, Juárez-Aguilar E, Olivares-Hernández JD, Tamariz E. Mimicking Neural Stem Cell Niche by Biocompatible Substrates. Stem Cells Int 2016; 2016:1513285. [PMID: 26880934 PMCID: PMC4736764 DOI: 10.1155/2016/1513285] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/19/2015] [Accepted: 11/23/2015] [Indexed: 01/17/2023] Open
Abstract
Neural stem cells (NSCs) participate in the maintenance, repair, and regeneration of the central nervous system. During development, the primary NSCs are distributed along the ventricular zone of the neural tube, while, in adults, NSCs are mainly restricted to the subependymal layer of the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus in the hippocampus. The circumscribed areas where the NSCs are located contain the secreted proteins and extracellular matrix components that conform their niche. The interplay among the niche elements and NSCs determines the balance between stemness and differentiation, quiescence, and proliferation. The understanding of niche characteristics and how they regulate NSCs activity is critical to building in vitro models that include the relevant components of the in vivo niche and to developing neuroregenerative approaches that consider the extracellular environment of NSCs. This review aims to examine both the current knowledge on neurogenic niche and how it is being used to develop biocompatible substrates for the in vitro and in vivo mimicking of extracellular NSCs conditions.
Collapse
Affiliation(s)
- Citlalli Regalado-Santiago
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Enrique Juárez-Aguilar
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Juan David Olivares-Hernández
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Elisa Tamariz
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| |
Collapse
|
15
|
Kawa MP, Stecewicz I, Piecyk K, Pius-Sadowska E, Paczkowska E, Rogińska D, Sobuś A, Łuczkowska K, Gawrych E, Petriczko E, Walczak M, Machaliński B. Effects of growth hormone therapeutic supplementation on hematopoietic stem/progenitor cells in children with growth hormone deficiency: focus on proliferation and differentiation capabilities. Endocrine 2015; 50:162-75. [PMID: 25920498 PMCID: PMC4546702 DOI: 10.1007/s12020-015-0591-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/30/2015] [Indexed: 11/04/2022]
Abstract
We investigated the direct effects of growth hormone (GH) replacement therapy (GH-RT) on hematopoiesis in children with GH deficiency (GHD) with the special emphasis on proliferation and cell cycle regulation. Peripheral blood (PB) was collected from sixty control individuals and forty GHD children before GH-RT and in 3rd and 6th month of GH-RT to measure hematological parameters and isolate CD34(+)-enriched hematopoietic progenitor cells (HPCs). Selected parameters of PB were analyzed by hematological analyzer. Moreover, collected HPCs were used to analyze GH receptor (GHR) and IGF1 expression, clonogenicity, and cell cycle activity. Finally, global gene expression profile of collected HPCs was analyzed using genome-wide RNA microarrays. GHD resulted in a decrease in several hematological parameters related to RBCs and significantly diminished clonogenicity of erythroid progenies. In contrast, GH-RT stimulated increases in clonogenic growth of erythroid lineage and RBC counts as well as significant up-regulation of cell cycle-propagating genes, including MAP2K1, cyclins D1/E1, PCNA, and IGF1. Likewise, GH-RT significantly modified GHR expression in isolated HPCs and augmented systemic IGF1 levels. Global gene expression analysis revealed significantly higher expression of genes associated with cell cycle, proliferation, and differentiation in HPCs from GH-treated subjects. (i) GH-RT significantly augments cell cycle progression in HPCs and increases clonogenicity of erythroid progenitors; (ii) GHR expression in HPCs is modulated by GH status; (iii) molecular mechanisms by which GH influences hematopoiesis might provide a basis for designing therapeutic interventions for hematological complications related to GHD.
Collapse
Affiliation(s)
- M. P. Kawa
- />Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - I. Stecewicz
- />Department of Pediatrics, Endocrinology, Diabetology, Metabolic Diseases and Cardiology of the Developmental Age, Pomeranian Medical University, Szczecin, Poland
| | - K. Piecyk
- />Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - E. Pius-Sadowska
- />Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - E. Paczkowska
- />Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - D. Rogińska
- />Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - A. Sobuś
- />Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - K. Łuczkowska
- />Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - E. Gawrych
- />Department of Pediatric and Oncological Surgery, Pomeranian Medical University, Szczecin, Poland
| | - E. Petriczko
- />Department of Pediatrics, Endocrinology, Diabetology, Metabolic Diseases and Cardiology of the Developmental Age, Pomeranian Medical University, Szczecin, Poland
| | - M. Walczak
- />Department of Pediatrics, Endocrinology, Diabetology, Metabolic Diseases and Cardiology of the Developmental Age, Pomeranian Medical University, Szczecin, Poland
| | - B. Machaliński
- />Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
16
|
Kong L, Hu Y, Yao Y, Jiao Y, Li S, Yang J. The Coumarin Derivative Osthole Stimulates Adult Neural Stem Cells, Promotes Neurogenesis in the Hippocampus, and Ameliorates Cognitive Impairment in APP/PS1 Transgenic Mice. Biol Pharm Bull 2015; 38:1290-301. [DOI: 10.1248/bpb.b15-00142] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Yu Hu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Yingjia Yao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Yanan Jiao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Shaoheng Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Jingxian Yang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| |
Collapse
|
17
|
Desai M, Li T, Han G, Ross MG. Programmed hyperphagia secondary to increased hypothalamic SIRT1. Brain Res 2014; 1589:26-36. [PMID: 25245521 DOI: 10.1016/j.brainres.2014.09.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 09/09/2014] [Accepted: 09/11/2014] [Indexed: 01/10/2023]
Abstract
Small for gestational age (SGA) offspring exhibit reduced hypothalamic neural satiety pathways leading to programmed hyperphagia and adult obesity. Appetite regulatory site, the hypothalamic arcuate nucleus (ARC) contains appetite (NPY/AgRP) and satiety (POMC) neurons. Using in vitro culture of hypothalamic neuroprogenitor cells (NPC) which form the ARC, we demonstrated that SGA offspring exhibit reduced NPC proliferation and neuronal differentiation. bHLH protein Hes1 promotes NPC self-renewal and inhibits differentiation by repressing neuronal differentiation genes (Mash1, neurogenin3). We hypothesized that Hes1/Mash1 and ultimately ARC neuronal differentiation and expression of NPY/POMC neurons are influenced by SIRT1 which is a nutrient sensor and a histone deacetylase. Control dams received ad libitum food, whereas study dams were 50% food-restricted from pregnancy day 10 to 21 (SGA). In vivo studies showed that SGA newborns and adult offspring had increased protein expression of hypothalamic/ARC SIRT1 and AgRP with decreased POMC. Additionally, SGA newborns had decreased expression of hypothalamic neurogenic factors with reduced in vivo NPC proliferation. In vitro culture of hypothalamic NPCs showed similar changes with elevated SIRT1 binding to Hes1 in SGA newborn. Silencing SIRT1 increased NPC proliferation and Hes1 and Tuj1expression in both Control and SGA NPCs. Although SGA NPC proliferation remained below that of Controls, it was higher than Control NPCs in the absence of SIRT1 siRNA. The direct impact of SIRT1 on NPC proliferation and differentiation were further confirmed with pharmacologic SIRT1 inhibitor and activator. Thus, in SGA newborns elevated SIRT1 induces premature differentiation of NPCs, reducing the NPC pool and cell proliferation.
Collapse
Affiliation(s)
- Mina Desai
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California 90502.
| | - Tie Li
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California 90502
| | - Guang Han
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California 90502
| | - Michael G Ross
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California 90502
| |
Collapse
|
18
|
Devesa P, Agasse F, Xapelli S, Almengló C, Devesa J, Malva JO, Arce VM. Growth hormone pathways signaling for cell proliferation and survival in hippocampal neural precursors from postnatal mice. BMC Neurosci 2014; 15:100. [PMID: 25156632 PMCID: PMC4155078 DOI: 10.1186/1471-2202-15-100] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 08/15/2014] [Indexed: 02/08/2023] Open
Abstract
Background Accumulating evidence suggests that growth hormone (GH) may play a major role in the regulation of postnatal neurogenesis, thus supporting the possibility that it may be also involved in promoting brain repair after brain injury. In order to gain further insight on this possibility, in this study we have investigated the pathways signaling the effect of GH treatment on the proliferation and survival of hippocampal subgranular zone (SGZ)-derived neurospheres. Results Our results demonstrate that GH treatment promotes both proliferation and survival of SGZ neurospheres. By using specific chemical inhibitors we have been also able to demonstrate that GH treatment promotes the activation of both Akt-mTOR and JNK signaling pathways, while blockade of these pathways either reduces or abolishes the GH effects. In contrast, no effect of GH on the activation of the Ras-ERK pathway was observed after GH treatment, despite blockade of this signaling path also resulted in a significant reduction of GH effects. Interestingly, SGZ cells were also capable of producing GH, and blockade of endogenous GH also resulted in a decrease in the proliferation and survival of SGZ neurospheres. Conclusions Altogether, our findings suggest that GH treatment may promote the proliferation and survival of neural progenitors. This effect may be elicited by cooperating with locally-produced GH in order to increase the response of neural progenitors to adequate stimuli. On this view, the possibility of using GH treatment to promote neurogenesis and cell survival in some acquired neural injuries may be envisaged.
Collapse
Affiliation(s)
| | | | | | | | - Jesús Devesa
- Department of Physiology, School of Medicine, University of Santiago de Compostela, 15710 Santiago de Compostela, Spain.
| | | | | |
Collapse
|
19
|
Johansson J, Grönbladh A, Hallberg M. Gamma-hydroxybutyrate (GHB) induces cognitive deficits and affects GABAB receptors and IGF-1 receptors in male rats. Behav Brain Res 2014; 269:164-74. [DOI: 10.1016/j.bbr.2014.04.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 04/16/2014] [Accepted: 04/19/2014] [Indexed: 12/30/2022]
|
20
|
McLenachan S, Zhang D, Palomo ABA, Edel MJ, Chen FK. mRNA transfection of mouse and human neural stem cell cultures. PLoS One 2013; 8:e83596. [PMID: 24386231 PMCID: PMC3873397 DOI: 10.1371/journal.pone.0083596] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 11/13/2013] [Indexed: 11/18/2022] Open
Abstract
The use of synthetic mRNA as an alternative gene delivery vector to traditional DNA-based constructs provides an effective method for inducing transient gene expression in cell cultures without genetic modification. Delivery of mRNA has been proposed as a safer alternative to viral vectors in the induction of pluripotent cells for regenerative therapies. Although mRNA transfection of fibroblasts, dendritic and embryonic stem cells has been described, mRNA delivery to neurosphere cultures has not been previously reported. Here we sought to establish an efficient method for delivering mRNA to primary neurosphere cultures. Neurospheres derived from the subventricular zone of adult mice or from human embryonic stem cells were transfected with EGFP mRNA by lipofection and electroporation. Transfection efficiency and expression levels were monitored by flow cytometry. Cell survival following transfection was examined using live cell counting and the MTT assay. Both lipofection and electroporation provided high efficiency transfection of neurospheres. In comparison with lipofection, electroporation resulted in increased transfection efficiencies, but lower expression per cell and shorter durations of expression. Additional rounds of lipofection renewed EGFP expression in neurospheres, suggesting this method may be suitable for reprogramming applications. In summary, we have developed a protocol for achieving high efficiency transfection rates in mouse and human neurosphere cell culture that can be applied for future studies of gene function studies in neural stem cells, such as defining efficient differentiation protocols for glial and neuronal linages.
Collapse
Affiliation(s)
- Samuel McLenachan
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Western Australia, Australia ; Ocular Tissue Engineering Laboratory, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Dan Zhang
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Western Australia, Australia ; Ocular Tissue Engineering Laboratory, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Ana Belén Alvarez Palomo
- Control of Pluripotency Laboratory, Molecular Genetics Research Group, Department of Physiological Sciences I, University of Barcelona, Barcelona, Spain
| | - Michael J Edel
- Control of Pluripotency Laboratory, Molecular Genetics Research Group, Department of Physiological Sciences I, University of Barcelona, Barcelona, Spain ; University of Sydney Medical School, Faculty of Medicine, Westmead Childrens Hospital, Division of Pediatrics and Child Health, Sydney, Australia
| | - Fred K Chen
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Western Australia, Australia ; Ocular Tissue Engineering Laboratory, Lions Eye Institute, Nedlands, Western Australia, Australia ; Ophthalmology Department, Royal Perth Hospital, Wellington Street, Perth, Western Australia, Australia
| |
Collapse
|
21
|
Regalado-Santiago C, López-Meraz ML, Santiago-García J, Fernández-Pomares C, Juárez-Aguilar E. Growth hormone (GH) is a survival rather than a proliferative factor for embryonic striatal neural precursor cells. Growth Horm IGF Res 2013; 23:179-186. [PMID: 23891194 DOI: 10.1016/j.ghir.2013.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/30/2013] [Accepted: 07/04/2013] [Indexed: 01/15/2023]
Abstract
OBJECTIVE A possible role of GH during central nervous system (CNS) development has been suggested by the presence of this hormone and its receptor in brain areas before its production by the pituitary gland. Although several effects have been reported for GH, the specific role of this hormone during CNS development remains unclear. Here, we examined the effect of GH on proliferation, survival and neurosphere formation in primary cultures of striatal tissue from 14-day-old (E14) mouse embryos. DESIGN GH receptor gene expression was confirmed by RT-PCR. Primary cultures of embryonic striatal cells were treated with different doses of GH in serum free media, then the number of neurospheres was determined. To examine the GH effect on proliferation and survival of the striatal primary cultures, bromodeoxyuridine (BrdU) and TUNEL immunoreactivity was conducted. RESULTS In the presence of the epidermal growth factor (EGF), GH increased the formation of neurospheres, with a maximal response at 10 ng/ml, higher doses were inhibitory. In absence of EGF, GH failed to stimulate neurosphere formation. Proliferation rate in the primary striatal cultures was inhibited by 24 or 48 h incubation with GH. However, in the absence of EGF, GH increased BrdU incorporation. GH treatment decreases the rate of apoptosis of nestin and GFAP positive cells in the primary striatal cultures, enhancing neurosphere formation. CONCLUSIONS Our in vitro data demonstrate that GH plays a survival role on the original population of embryonic striatal cells, improving Neural Precursor Cells (NPCs) expansion. We suggest that this GH action could be predominant during striatal neurodevelopment.
Collapse
|
22
|
Baquedano E, Chowen JA, Argente J, Frago LM. Differential effects of GH and GH-releasing peptide-6 on astrocytes. J Endocrinol 2013; 218:263-74. [PMID: 23792323 DOI: 10.1530/joe-13-0053] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
GH and GH secretagogues (GHSs) are involved in many cellular activities such as stimulation of mitosis, proliferation and differentiation. As astrocytes are involved in developmental and protective functions, our aim was to analyse the effects of GH and GH-releasing hexapeptide on astrocyte proliferation and differentiation in the hypothalamus and hippocampus. Treatment of adult male Wistar rats with GH (i.v., 100 μg/day) for 1 week increased the levels of glial fibrillary acidic protein (GFAP) and decreased the levels of vimentin in the hypothalamus and hippocampus. These changes were not accompanied by increased proliferation. By contrast, GH-releasing hexapeptide (i.v., 150 μg/day) did not affect GFAP levels but increased proliferation in the areas studied. To further study the intracellular mechanisms involved in these effects, we treated C6 astrocytoma cells with GH or GH-releasing hexapeptide and the phosphatidylinositol 3'-kinase (PI3K) inhibitor, LY294002, and observed that the presence of this inhibitor reverted the increase in GFAP levels induced by GH and the proliferation induced by GH-releasing hexapeptide. We conclude that although GH-releasing hexapeptide is a GHS, it may exert GH-independent effects centrally on astrocytes when administered i.v., although the effects of both substances appear to be mediated by the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Eva Baquedano
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | |
Collapse
|
23
|
Arce VM, Devesa P, Devesa J. Role of growth hormone (GH) in the treatment on neural diseases: from neuroprotection to neural repair. Neurosci Res 2013; 76:179-86. [PMID: 23602740 DOI: 10.1016/j.neures.2013.03.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 02/26/2013] [Accepted: 03/26/2013] [Indexed: 12/13/2022]
Abstract
Growth hormone (GH) is a pleiotropic hormone that exerts important functions in the control of brain development as well as in the regulation neuronal differentiation and function, together with several behavioral and psychological effects that have been linked to its modulatory actions on brain neurotransmitters. In addition, the possibility that GH may play a role on brain repair after injury has been also envisaged, and a number of reports have shown that GH administration following injury confers neuroprotection and accelerates the recovery of some neural functions. In this review we have analyzed the state of the art of GH administration in several neural diseases. Though more studies are still necessary in order to completely understand the importance of GH in these processes, the promising results obtained so far, together with the absence of untoward effects during GH therapy, encourages the development of clinical assays in order to further support the use GH treatment in neural diseases in which neuroprotection and/or neuroregeneration are involved.
Collapse
Affiliation(s)
- Víctor M Arce
- Departamento de Fisioloxía, Facultade de Medicina, Universidade de Santiago de Compostela, Spain.
| | | | | |
Collapse
|
24
|
Devesa J, Reimunde P, Devesa P, Barberá M, Arce V. Growth hormone (GH) and brain trauma. Horm Behav 2013; 63:331-44. [PMID: 22405763 DOI: 10.1016/j.yhbeh.2012.02.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 02/21/2012] [Accepted: 02/22/2012] [Indexed: 12/27/2022]
Abstract
Growth hormone (GH) is a pleiotropic hormone with known neurotrophic effects. We aimed to study whether GH administration might be useful together with rehabilitation in the recovery of TBI patients. 13 TBI patients (8 M, 5 F; age: 6-53 years old) were studied. Time after TBI: 2.5 months to 11 years; 5 patients showed acquired GH-deficiency (GHD). Disabilities observed: cognitive disorders; motor plegias; neurogenic dysphagia (n=5), vegetative coma (n=2) and amaurosis (n=1). All but one TBI patient followed intense rehabilitation for years. Treatment consisted of GH administration (maximal dose 1 mg/day, 5 days/week, resting 15-days every 2-months, until a maximum of 8 months) and clinical rehabilitation according to the individual needs (3-4 h/day, 5 days/week, during 6-12 months). Informed consent was obtained before commencing GH administration. GH significantly increased plasma IGF-1 values (ng.mL(-1)) in both GHD and no GHD patients, being then similar between both groups (GHD: 275.6±35.6 [p<0.01 vs. baseline], no GHD: 270.2±64 [p<0.05 vs. baseline]). In all the cases clear significant improvements were observed during and at the end of the combined treatment. Cognitive improvements appeared earlier and were more important than motor improvements. Swallowing improved significantly in all TBI patients with neurogenic dysphagia (2 of them in a vegetative state). Visual performance was ameliorated in the patient with amaurosis. No undesirable side-effects were observed. Our data indicate that GH can be combined with rehabilitation for improving disabilities in TBI patients, regardless of whether or not they are GHD.
Collapse
Affiliation(s)
- Jesús Devesa
- Department of Physiology, School of Medicine, University of Santiago de Compostela, Spain.
| | | | | | | | | |
Collapse
|
25
|
Christie KJ, Turnley AM. Regulation of endogenous neural stem/progenitor cells for neural repair-factors that promote neurogenesis and gliogenesis in the normal and damaged brain. Front Cell Neurosci 2013; 6:70. [PMID: 23346046 PMCID: PMC3548228 DOI: 10.3389/fncel.2012.00070] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 12/30/2012] [Indexed: 01/17/2023] Open
Abstract
Neural stem/precursor cells in the adult brain reside in the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus in the hippocampus. These cells primarily generate neuroblasts that normally migrate to the olfactory bulb (OB) and the dentate granule cell layer respectively. Following brain damage, such as traumatic brain injury, ischemic stroke or in degenerative disease models, neural precursor cells from the SVZ in particular, can migrate from their normal route along the rostral migratory stream (RMS) to the site of neural damage. This neural precursor cell response to neural damage is mediated by release of endogenous factors, including cytokines and chemokines produced by the inflammatory response at the injury site, and by the production of growth and neurotrophic factors. Endogenous hippocampal neurogenesis is frequently also directly or indirectly affected by neural damage. Administration of a variety of factors that regulate different aspects of neural stem/precursor biology often leads to improved functional motor and/or behavioral outcomes. Such factors can target neural stem/precursor proliferation, survival, migration and differentiation into appropriate neuronal or glial lineages. Newborn cells also need to subsequently survive and functionally integrate into extant neural circuitry, which may be the major bottleneck to the current therapeutic potential of neural stem/precursor cells. This review will cover the effects of a range of intrinsic and extrinsic factors that regulate neural stem/precursor cell functions. In particular it focuses on factors that may be harnessed to enhance the endogenous neural stem/precursor cell response to neural damage, highlighting those that have already shown evidence of preclinical effectiveness and discussing others that warrant further preclinical investigation.
Collapse
Affiliation(s)
- Kimberly J Christie
- Neural Regeneration Laboratory, Department of Anatomy and Neuroscience, Centre for Neuroscience Research, The University of Melbourne Parkville, VIC, Australia
| | | |
Collapse
|
26
|
Dibajnia P, Morshead CM. Role of neural precursor cells in promoting repair following stroke. Acta Pharmacol Sin 2013; 34:78-90. [PMID: 23064725 PMCID: PMC4086492 DOI: 10.1038/aps.2012.107] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 07/02/2012] [Indexed: 01/01/2023]
Abstract
Stem cell-based therapies for the treatment of stroke have received considerable attention. Two broad approaches to stem cell-based therapies have been taken: the transplantation of exogenous stem cells, and the activation of endogenous neural stem and progenitor cells (together termed neural precursors). Studies examining the transplantation of exogenous cells have demonstrated that neural stem and progenitor cells lead to the most clinically promising results. Endogenous activation of neural precursors has also been explored based on the fact that resident precursor cells have the inherent capacity to proliferate, migrate and differentiate into mature neurons in the uninjured adult brain. Studies have revealed that these neural precursor cell behaviours can be activated following stroke, whereby neural precursors will expand in number, migrate to the infarct site and differentiate into neurons. However, this innate response is insufficient to lead to functional recovery, making it necessary to enhance the activation of endogenous precursors to promote tissue repair and functional recovery. Herein we will discuss the current state of the stem cell-based approaches with a focus on endogenous repair to treat the stroke injured brain.
Collapse
Affiliation(s)
- Pooya Dibajnia
- Department of Surgery, Division of Anatomy, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Cindi M Morshead
- Department of Surgery, Division of Anatomy, University of Toronto, Toronto, ON M5S 3E1, Canada
- Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
27
|
Grönbladh A, Johansson J, Nyberg F, Hallberg M. Recombinant human growth hormone affects the density and functionality of GABAB receptors in the male rat brain. Neuroendocrinology 2013; 97:203-11. [PMID: 22710737 DOI: 10.1159/000339821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 05/27/2012] [Indexed: 11/19/2022]
Abstract
The beneficial effects of growth hormone (GH) on memory and learning have previously been confirmed in both humans and in animal models. An important role of GABAB receptors for multiple forms of learning and memory has also been reported. In this study, we examined the effect of GH on the density and functionality of the metabotropic GABAB receptors in the rat brain. Male Sprague-Dawley rats (n = 24) divided into 3 groups were injected twice daily with recombinant human GH (0.07 or 0.7 IU/kg) for 7 days. The effects of the hormone were determined by quantitative autoradiography and by GABAB stimulated [(35)S]-GTPγS binding using the selective GABAB receptor agonist baclofen. The results demonstrate moderate but significant alterations in both receptor density and functionality in a number of brain regions. For example, a dose-dependent upregulation of GABAB receptors was found in the cingulate cortex, primary motor cortex and caudate putamen, whereas attenuation in the receptor density was encountered in, for example, the medial geniculate nucleus. Although the GH-induced effects on the GABAB receptor in brain areas associated with cognition were fairly pronounced, they were significant and we propose that the physiological responses observed after GH administration at least partly can be mediated through a mechanism involving GABAB receptors.
Collapse
Affiliation(s)
- Alfhild Grönbladh
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | | |
Collapse
|
28
|
GH mediates exercise-dependent activation of SVZ neural precursor cells in aged mice. PLoS One 2012; 7:e49912. [PMID: 23209615 PMCID: PMC3507946 DOI: 10.1371/journal.pone.0049912] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 10/15/2012] [Indexed: 12/21/2022] Open
Abstract
Here we demonstrate, both in vivo and in vitro, that growth hormone (GH) mediates precursor cell activation in the subventricular zone (SVZ) of the aged (12-month-old) brain following exercise, and that GH signaling stimulates precursor activation to a similar extent to exercise. Our results reveal that both addition of GH in culture and direct intracerebroventricular infusion of GH stimulate neural precursor cells in the aged brain. In contrast, no increase in neurosphere numbers was observed in GH receptor null animals following exercise. Continuous infusion of a GH antagonist into the lateral ventricle of wild-type animals completely abolished the exercise-induced increase in neural precursor cell number. Given that the aged brain does not recover well after injury, we investigated the direct effect of exercise and GH on neural precursor cell activation following irradiation. This revealed that physical exercise as well as infusion of GH promoted repopulation of neural precursor cells in irradiated aged animals. Conversely, infusion of a GH antagonist during exercise prevented recovery of precursor cells in the SVZ following irradiation.
Collapse
|
29
|
Porlan E, Perez-Villalba A, Delgado AC, Ferrón SR. Paracrine regulation of neural stem cells in the subependymal zone. Arch Biochem Biophys 2012; 534:11-9. [PMID: 23073070 DOI: 10.1016/j.abb.2012.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 09/27/2012] [Accepted: 10/05/2012] [Indexed: 12/31/2022]
Abstract
Stem cells maintain their self-renewal and multipotency capacities through a self-organizing network of transcription factors and intracellular pathways activated by extracellular signaling from the microenvironment or "niche" in which they reside in vivo. In the adult mammalian brain new neurons continue to be generated throughout life of the organisms and this lifelong process of neurogenesis is supported by a reservoir of neural stem cells in the germinal regions. The discovery of adult neurogenesis in the mammalian brain has sparked great interest in defining the conditions that guide neural stem cell (NSC) maintenance and differentiation into the great variety of neuronal and glial subtypes. Here we review current knowledge regarding the paracrine regulation provided by the components of the niche and its function, focusing on the main germinal region of the adult central nervous system (CNS), the subependymal zone (SEZ).
Collapse
Affiliation(s)
- Eva Porlan
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Spain
| | | | | | | |
Collapse
|
30
|
Application of in vitro [³⁵S]GTPγ-S autoradiography in studies of growth hormone effects on opioid receptors in the male rat brain. Brain Res Bull 2012; 90:100-6. [PMID: 23063719 DOI: 10.1016/j.brainresbull.2012.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 09/07/2012] [Accepted: 09/12/2012] [Indexed: 11/21/2022]
Abstract
Chronic treatment with opiates may inhibit cell growth and trigger apoptosis. On the contrary, growth hormone (GH) has been demonstrated to stimulate neurogenesis and counteract apoptosis. We recently demonstrated that recombinant human GH (rhGH) may reverse opiate-induced apoptosis in cells derived from prenatal mouse hippocampus. Thus, GH might be able to prevent the impaired cognitive capabilities that may occur in both humans and other mammals in connection to chronic opiate treatment. In order to explore the mechanism by which GH exerts its beneficial effects we here examined the impact of GH treatment on the levels of delta and mu opioid peptide (DOP and MOP, respectively) receptors in the male rat brain. The rats were treated with rhGH (Genotropin®) at two different doses (0.07 and 0.7 IU/kg), twice daily, during 7 days. Following decapitation, the levels of DOP and MOP receptor functionality were determined using [³⁵S]GTPγS autoradiography. The results demonstrate that rhGH affects the levels of the MOP receptor functionality in certain areas of the brain. These alterations were seen in e.g. amygdala and thalamus, i.e. regions that recently have been implicated in learning and memory. The activity level of DOP receptors was not affected. Thus, the data support that the beneficial effect of GH on counteracting apoptosis might involve a direct or indirect effect on the MOP but not the DOP receptor.
Collapse
|
31
|
Blackmore DG, Reynolds BA, Golmohammadi MG, Large B, Aguilar RM, Haro L, Waters MJ, Rietze RL. Growth hormone responsive neural precursor cells reside within the adult mammalian brain. Sci Rep 2012; 2:250. [PMID: 22355762 PMCID: PMC3274722 DOI: 10.1038/srep00250] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 01/16/2012] [Indexed: 12/02/2022] Open
Abstract
The detection of growth hormone (GH) and its receptor in germinal regions of the mammalian brain prompted our investigation of GH and its role in the regulation of endogenous neural precursor cell activity. Here we report that the addition of exogenous GH significantly increased the expansion rate in long-term neurosphere cultures derived from wild-type mice, while neurospheres derived from GH null mice exhibited a reduced expansion rate. We also detected a doubling in the frequency of large (i.e. stem cell-derived) colonies for up to 120 days following a 7-day intracerebroventricular infusion of GH suggesting the activation of endogenous stem cells. Moreover, gamma irradiation induced the ablation of normally quiescent stem cells in GH-infused mice, resulting in a decline in olfactory bulb neurogenesis. These results suggest that GH activates populations of resident stem and progenitor cells, and therefore may represent a novel therapeutic target for age-related neurodegeneration and associated cognitive decline.
Collapse
Affiliation(s)
- Daniel G Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane Queensland, 4072, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Devesa P, Reimunde P, Gallego R, Devesa J, Arce VM. Growth hormone (GH) treatment may cooperate with locally-produced GH in increasing the proliferative response of hippocampal progenitors to kainate-induced injury. Brain Inj 2011; 25:503-10. [PMID: 21456999 DOI: 10.3109/02699052.2011.559611] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PRIMARY OBJECTIVE This study was designed to investigate the effect of growth hormone treatment on the proliferation of endogenous neural progenitor cells in the dentate gyrus (DG) of the brain stimulated by kainic acid (KA)-induced neurotoxicity. RESEARCH DESIGN Neurotoxicity was induced by intraperitoneal injection of KA. GH treatment lasted 4 days, starting either immediately or after 10 days of administration of the neurotoxic insult. METHODS AND PROCEDURE Proliferating cells were immunodetected after labelling by in vivo administration of 5-bromodeoxyuridine (BrdU). GH expression was detected by in situ hybridization and immunofluorescence. MAIN OUTCOMES AND RESULTS KA administration stimulated the proliferation of hippocampal precursors and this effect was significantly enhanced by GH treatment. Hippocampal GH expression was also up-regulated in response to KA administration. CONCLUSIONS The findings support the possibility that the proliferative response observed in the hippocampus of rats treated with KA and GH is a consequence of cooperation between the exogenous and the locally-produced hormone and their synergism with other mitogenic factors generated in response to the neurotoxic damage. Therefore, GH treatment could be used to cooperate with other physiological or pathological stimuli in order to promote cell proliferation.
Collapse
Affiliation(s)
- Pablo Devesa
- Department of Physiology, School of Medicine, University of Santiago de Compostela, Spain
| | | | | | | | | |
Collapse
|
33
|
Growth hormone and prolactin regulate human neural stem cell regenerative activity. Neuroscience 2011; 190:409-27. [PMID: 21664953 DOI: 10.1016/j.neuroscience.2011.05.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 05/10/2011] [Accepted: 05/12/2011] [Indexed: 12/18/2022]
Abstract
We have previously shown that the growth hormone (GH)/prolactin (PRL) axis has a significant role in regulating neuroprotective and/or neurorestorative mechanisms in the brain and that these effects are mediated, at least partly, via actions on neural stem cells (NSCs). Here, using NSCs with properties of neurogenic radial glia derived from fetal human forebrains, we show that exogenously applied GH and PRL promote the proliferation of NSCs in the absence of epidermal growth factor or basic fibroblast growth factor. When applied to differentiating NSCs, they both induce neuronal progenitor proliferation, but only PRL has proliferative effects on glial progenitors. Both GH and PRL also promote NSC migration, particularly at higher concentrations. Since human GH activates both GH and PRL receptors, we hypothesized that at least some of these effects may be mediated via the latter. Migration studies using receptor-specific antagonists confirmed that GH signals via the PRL receptor promote migration. Mechanisms of receptor signaling in NSC proliferation, however, remain to be elucidated. In summary, GH and PRL have complex stimulatory and modulatory effects on NSC activity and as such may have a role in injury-related recovery processes in the brain.
Collapse
|
34
|
Deleyrolle LP, Ericksson G, Morrison BJ, Lopez JA, Burrage K, Burrage P, Vescovi A, Rietze RL, Reynolds BA. Determination of somatic and cancer stem cell self-renewing symmetric division rate using sphere assays. PLoS One 2011; 6:e15844. [PMID: 21246056 PMCID: PMC3016423 DOI: 10.1371/journal.pone.0015844] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 11/25/2010] [Indexed: 12/21/2022] Open
Abstract
Representing a renewable source for cell replacement, neural stem cells have received substantial attention in recent years. The neurosphere assay represents a method to detect the presence of neural stem cells, however owing to a deficiency of specific and definitive markers to identify them, their quantification and the rate they expand is still indefinite. Here we propose a mathematical interpretation of the neurosphere assay allowing actual measurement of neural stem cell symmetric division frequency. The algorithm of the modeling demonstrates a direct correlation between the overall cell fold expansion over time measured in the sphere assay and the rate stem cells expand via symmetric division. The model offers a methodology to evaluate specifically the effect of diseases and treatments on neural stem cell activity and function. Not only providing new insights in the evaluation of the kinetic features of neural stem cells, our modeling further contemplates cancer biology as cancer stem-like cells have been suggested to maintain tumor growth as somatic stem cells maintain tissue homeostasis. Indeed, tumor stem cell's resistance to therapy makes these cells a necessary target for effective treatment. The neurosphere assay mathematical model presented here allows the assessment of the rate malignant stem-like cells expand via symmetric division and the evaluation of the effects of therapeutics on the self-renewal and proliferative activity of this clinically relevant population that drive tumor growth and recurrence.
Collapse
Affiliation(s)
- Loic P. Deleyrolle
- McKnight Brain Institute, Neurosurgery department, University of Florida, Gainesville, Florida, United States of America
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- * E-mail: (LPD); (BAR)
| | - Geoffery Ericksson
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Brian J. Morrison
- Griffith University, Brisbane, Queensland, Australia
- Queensland Institute of Medical Research, Royal Brisbane Hospital, Brisbane, Queensland, Australia
| | - J. Alejandro Lopez
- Griffith University, Brisbane, Queensland, Australia
- Queensland Institute of Medical Research, Royal Brisbane Hospital, Brisbane, Queensland, Australia
| | - Kevin Burrage
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Pamela Burrage
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | | | - Rodney L. Rietze
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Pfizer Regenerative Medicine, Cambridge, United Kingdom
| | - Brent A. Reynolds
- McKnight Brain Institute, Neurosurgery department, University of Florida, Gainesville, Florida, United States of America
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- * E-mail: (LPD); (BAR)
| |
Collapse
|
35
|
Chan L, Albertsson-Wikland K, Camacho-Hübner C, Hochberg Z. Signal transduction in child health: closing the gap between clinical and basic research. Sci Signal 2010; 3:mr3. [PMID: 20940424 DOI: 10.1126/scisignal.3143mr3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although some diseases are specific to children, many diseases of adults, including obesity and the metabolic syndrome, often originate in childhood. Thus, understanding the mechanisms of disease onset and progression in children is vital not only for child health, but for adult health as well. The NICHe (New Inroads to Child Health) Conference series focuses on future directions in child health, by bringing together clinical and basic scientists with the aim of sharing knowledge to facilitate the development of new therapeutic approaches. This year's conference, entitled "Child Health and Signal Transduction," focused on G protein-coupled receptor and receptor tyrosine kinase pathways involved in endocrine signaling, how disruption of these pathways can lead to disease, and how understanding these pathways may guide drug discovery.
Collapse
Affiliation(s)
- Li Chan
- Centre for Endocrinology, William Harvey Research Institute, St. Bartholomew's and The Royal London School of Medicine and Dentistry, London, UK.
| | | | | | | |
Collapse
|
36
|
Abstract
Growth hormone is widely used clinically to promote growth and anabolism and for other purposes. Its actions are mediated via the growth hormone receptor, both directly by tyrosine kinase activation and indirectly by induction of insulin-like growth factor 1 (IGF-1). Insensitivity to growth hormone (Laron syndrome) can result from mutations in the growth hormone receptor and can be treated with IGF-1. This treatment is, however, not fully effective owing to the loss of the direct actions of growth hormone and altered availability of exogenous IGF-1. Excessive activation of the growth hormone receptor by circulating growth hormone results in gigantism and acromegaly, whereas cell transformation and cancer can occur in response to autocrine activation of the receptor. Advances in understanding the mechanism of receptor activation have led to a model in which the growth hormone receptor exists as a constitutive dimer. Binding of the hormone realigns the subunits by rotation and closer apposition, resulting in juxtaposition of the catalytic domains of the associated tyrosine-protein kinase JAK2 below the cell membrane. This change results in activation of JAK2 by transphosphorylation, then phosphorylation of receptor tyrosines in the cytoplasmic domain, which enables binding of adaptor proteins, as well as direct phosphorylation of target proteins. This model is discussed in the light of salient information from closely related class 1 cytokine receptors, such as the erythropoietin, prolactin and thrombopoietin receptors.
Collapse
Affiliation(s)
- Andrew J Brooks
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Qld 4072, Australia.
| | | |
Collapse
|
37
|
Christophidis LJ, Gorba T, Gustavsson M, Williams CE, Werther GA, Russo VC, Scheepens A. Growth hormone receptor immunoreactivity is increased in the subventricular zone of juvenile rat brain after focal ischemia: a potential role for growth hormone in injury-induced neurogenesis. Growth Horm IGF Res 2009; 19:497-506. [PMID: 19524466 DOI: 10.1016/j.ghir.2009.05.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2009] [Revised: 04/30/2009] [Accepted: 05/14/2009] [Indexed: 02/05/2023]
Abstract
BACKGROUND During recovery from an ischemic brain injury, a cerebral growth hormone (GH) axis is activated. Whilst GH has been demonstrated to be neuroprotective both in vitro and in vivo, a role for GH in neuro-restorative processes after brain injury has yet to be studied. OBJECTIVE To explore a role for GH in injury-induced neurogenesis by examining GH receptor (GH-R) immunoreactivity within the subventricular zone (SVZ) of juvenile rats after brain injury and by testing the proliferative capacity of GH on embryonic mouse neural stem cells. DESIGN Twenty-one day old rats were subjected to unilateral hypoxic-ischemia of the brain and sacrificed 1-15days later. Coronal brain sections from these animals and age-matched naïve controls were immunostained for GH-R and cell markers of neurogenesis. The level of GH-R immunoreactivity in the ipsilateral and contralateral SVZ of each animal was semi-quantified both by independent blinded scoring by two examiners and blinded image analysis. To examine the effect of GH on proliferation of embryonic mouse neural stem cells, cells were treated with increasing concentrations of rat pituitary GH for 48h in the presence of 5'-bromo-2'-deoxyuridine. RESULTS The level of GH-R immunoreactivity in the ipsilateral SVZ was significantly increased 5days after injury vs. the contralateral SVZ, coinciding both spatially and temporally with injury-induced neurogenesis. The population of GH-R immunopositive cells in the ipsilateral SVZ at this time was found to include proliferating cells (Ki67 immunopositive), neural progenitor cells (nestin immunopositive) and post-proliferative migratory neuroblasts (doublecortin immunopositive). Stimulation of embryonic mouse NSCs with physiological concentrations of rat pituitary GH elicited a dose-dependent proliferative response. CONCLUSION These results indicate a novel role for GH and its receptor in injury-induced neurogenesis, and suggest that GH treatment may potentiate endogenous neuro-restorative processes after brain injury.
Collapse
|