1
|
Epardo D, Balderas-Márquez JE, Rodríguez-Arzate CA, Thébault SC, Carranza M, Luna M, Ávila-Mendoza J, Calderón-Vallejo D, Quintanar JL, Arámburo C, Martínez-Moreno CG. Growth Hormone Neuroprotective Effects After an Optic Nerve Crush in the Male Rat. Invest Ophthalmol Vis Sci 2024; 65:17. [PMID: 39504048 PMCID: PMC11549927 DOI: 10.1167/iovs.65.13.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/12/2024] [Indexed: 11/11/2024] Open
Abstract
Purpose Growth hormone (GH) has neuroprotective effects that have not been evaluated in the mammalian visual system. This study tested the hypothesis that GH administration can promote retinal neuroprotection in an optic nerve crush (ONC) model in male rats. Methods The ON was compressed for 10 seconds, and bovine GH was injected concomitantly to injury for 14 days (0.5 µg/g every 12 hours). At 24 hours and 14 days after ONC, we evaluated the effects of GH upon several markers by quantitative PCR (qPCR), Western blot, and immunohistochemistry; the ON integrity was assessed using CTB Alexa 488 anterograde tracer, and retinal function was tested by full-field electroretinogram. Results GH partially prevented the ONC-induced death of retinal ganglion cells (RGCs), as well as the increase in gliosis marker GFAP at 14 days. Most of the ONC-induced changes in mRNA retinal levels of several neurotrophic, survival, synaptogenic, gliosis, and excitotoxicity markers were prevented by GH, both at 24 hours and 14 days, and treatment also stimulated the expression of antiapoptotic proteins Bcl-2 and Bcl-xL at 24 hours. Additionally, GH partially maintained the ON integrity and active anterograde transport, as well as retinal function by avoiding the reduced amplitude and slowing of the A- and B-waves and oscillatory potentials associated with the ONC at 14 days. Conclusions GH has neuroprotective effects in the ONC model in male rats, it promoted RGC survival, gliosis reduction, and axonal transport increase, likely through the regulation of genes involved in neuroprotection, survival, and synaptogenesis. Furthermore, GH prevented functional impairment, indicating its potential as a therapeutic option for retinal neurodegenerative diseases.
Collapse
Affiliation(s)
- David Epardo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., México
| | - Jerusa E Balderas-Márquez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., México
| | - Cynthia A Rodríguez-Arzate
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., México
| | - Stéphanie C Thébault
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., México
| | - Martha Carranza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., México
| | - Maricela Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., México
| | - José Ávila-Mendoza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., México
| | - Denisse Calderón-Vallejo
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ags., México
| | - J Luis Quintanar
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ags., México
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., México
| | - Carlos G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., México
| |
Collapse
|
2
|
Martínez-Moreno CG, Calderón-Vallejo D, Díaz-Galindo C, Hernández-Jasso I, Olivares-Hernández JD, Ávila-Mendoza J, Epardo D, Balderas-Márquez JE, Urban-Sosa VA, Baltazar-Lara R, Carranza M, Luna M, Arámburo C, Quintanar JL. Neurotrophic and synaptic effects of GnRH and/or GH upon motor function after spinal cord injury in rats. Sci Rep 2024; 14:26420. [PMID: 39488642 PMCID: PMC11531546 DOI: 10.1038/s41598-024-78073-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024] Open
Abstract
Thoracic spinal cord injury (SCI) profoundly impairs motor and sensory functions, significantly reducing life quality without currently available effective treatments for neuroprotection or full functional regeneration. This study investigated the neurotrophic and synaptic recovery potential of gonadotropin-releasing hormone (GnRH) and growth hormone (GH) treatments in ovariectomized rats subjected to thoracic SCI. Employing a multidisciplinary approach, we evaluated the effects of these hormones upon gene expression of classical neurotrophins (NGF, BDNF, and NT3) as well as indicative markers of synaptic function (Nlgn1, Nxn1, SNAP25, SYP, and syntaxin-1), together with morphological assessments of myelin sheath integrity (Klüver-Barrera staining and MBP immunoreactivity) and synaptogenic proteins (PSD95, SYP) by immunohystochemistry (IHC) , and also on the neuromotor functional recovery of hindlimbs in the lesioned animals. Results demonstrated that chronic administration of GnRH and GH induced notable upregulation in the expression of several neurotrophic and synaptogenic activity genes. Additionally, the treatment showed a significant impact on the restoration of functional synaptic markers and myelin integrity. Intriguingly, while individual GnRH application induced certain recovery benefits, the combined treatment with GH appeared to inhibit neuromotor recovery, suggesting a complex interplay in hormonal regulation post-SCI. GnRH and GH are bioactive and participate in modulating neurotrophic responses and synaptic restoration under neural damage conditions, offering insights into novel therapeutic approaches for SCI. However, the intricate effects of combined hormonal treatment accentuate the necessity for further investigation that conduce to optimal and novel therapeutic strategies for patients with spinal cord lesions.
Collapse
Affiliation(s)
- C G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - D Calderón-Vallejo
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| | - C Díaz-Galindo
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| | - I Hernández-Jasso
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| | - J D Olivares-Hernández
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - J Ávila-Mendoza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - D Epardo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - J E Balderas-Márquez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - V A Urban-Sosa
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - R Baltazar-Lara
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - M Carranza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - M Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - C Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México.
| | - J L Quintanar
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México.
| |
Collapse
|
3
|
Ávila-Mendoza J, Delgado-Rueda K, Urban-Sosa VA, Carranza M, Luna M, Martínez-Moreno CG, Arámburo C. KLF13 Regulates the Activity of the GH-Induced JAK/STAT Signaling by Targeting Genes Involved in the Pathway. Int J Mol Sci 2023; 24:11187. [PMID: 37446365 DOI: 10.3390/ijms241311187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
The Krüppel-like factor 13 (KLF13) has emerged as an important transcription factor involved in essential processes of the central nervous system (CNS). It predominantly functions as a transcriptional repressor, impacting the activity of several signaling pathways with essential roles in the CNS, including the JAK/STAT pathway, which is the canonical mediator of growth hormone (GH) signaling. It is now recognized that GH has important actions as a neurotrophic factor. Therefore, we analyzed the effects of KLF13 on the activity of the JAK/STAT signaling pathway in the hippocampus-derived cell line HT22. Results showed that KLF13 directly regulates the expression of several genes involved in the JAK-STAT pathway, including Jak1, Jak2, Jak3, and Socs1, by associating with their proximal gene promoters. In addition, it was found that in KLF13-deficient HT22 neurons, the expression of Jak1, Stat3, Socs1, Socs3, and Igf1 was dysregulated, exhibiting mRNA levels that went up to 7-fold higher than the control cell line. KLF13 displayed a differential effect on the GH-induced JAK/STAT pathway activity, decreasing the STAT3 branch while enhancing the STAT5 branch. In KLF13-deficient HT22 cells, the activity of the STAT3 branch was enhanced, mediating the GH-dependent augmented expression of the JAK/STAT output genes Socs1, Socs3, Igf1, and Bdnf. Furthermore, GH treatment increased both the nuclear content of KLF13 and Klf13 mRNA levels, suggesting that KLF13 could be part of the mechanisms that maintain the homeostatic state of this pathway. These findings support the notion that KLF13 is a regulator of JAK/STAT activity.
Collapse
Affiliation(s)
- José Ávila-Mendoza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Karen Delgado-Rueda
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Valeria A Urban-Sosa
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Martha Carranza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Maricela Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Carlos G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| |
Collapse
|
4
|
Jayakumar P, Martínez-Moreno CG, Lorenson MY, Walker AM, Morales T. Prolactin Attenuates Neuroinflammation in LPS-Activated SIM-A9 Microglial Cells by Inhibiting NF-κB Pathways Via ERK1/2. Cell Mol Neurobiol 2022; 42:2171-2186. [PMID: 33821330 PMCID: PMC11421592 DOI: 10.1007/s10571-021-01087-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 03/27/2021] [Indexed: 10/21/2022]
Abstract
Prolactin (PRL) is a pleiotropic hormone with multiple functions in several tissues and organs, including the brain. PRL decreases lesion-induced microgliosis and modifies gene expression related to microglial functions in the hippocampus, thereby providing a possible mechanism through which it might participate in neuroimmune modulatory responses and prevent neuronal cell damage. However, the direct contribution of microglial cells to PRL-mediated neuroprotection is still unclear and no studies have yet documented whether PRL can directly activate cellular pathways in microglial cells. The aim of this study is to elucidate in vitro actions of PRL on the immortalized SIM-A9 microglia cell line in basal and LPS-stimulated conditions. PRL alone induced a time-dependent extracellular signal-regulated kinase 1/2 (ERK1/2) activation. Pretreatment with PRL attenuated LPS (200 ng/ml) stimulated pro-inflammatory markers: nitric oxide (NO) levels, inducible nitric oxide synthase (iNOS), interleukins (IL)-6, -1β and tumor necrosis factor (TNF-α) expression at 20 nM dosage. PRL suppressed LPS-induced nuclear factor (NF)-κappaB (NF-κB) p65 subunit phosphorylation and its upstream p-ERK1/2 activity. In conclusion, PRL exhibits anti-inflammatory effects in LPS-stimulated SIM-A9 microglia by downregulating pro-inflammatory mediators corresponding to suppression of LPS-activated ERK1/2 and NF-κB phosphorylation.
Collapse
Affiliation(s)
- Preethi Jayakumar
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Carlos G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Mary Y Lorenson
- Department of Biomedical Sciences, University of California, Riverside, CA, USA
| | - Ameae M Walker
- Department of Biomedical Sciences, University of California, Riverside, CA, USA
| | - Teresa Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico.
| |
Collapse
|
5
|
Klepper S, Jung S, Dittmann L, Geppert CI, Hartmann A, Beier N, Trollmann R. Further Evidence of Neuroprotective Effects of Recombinant Human Erythropoietin and Growth Hormone in Hypoxic Brain Injury in Neonatal Mice. Int J Mol Sci 2022; 23:ijms23158693. [PMID: 35955834 PMCID: PMC9368903 DOI: 10.3390/ijms23158693] [Citation(s) in RCA: 1] [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: 07/07/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023] Open
Abstract
Experimental in vivo data have recently shown complementary neuroprotective actions of rhEPO and growth hormone (rhGH) in a neonatal murine model of hypoxic brain injury. Here, we hypothesized that rhGH and rhEPO mediate stabilization of the blood−brain barrier (BBB) and regenerative vascular effects in hypoxic injury to the developing brain. Using an established model of neonatal hypoxia, neonatal mice (P7) were treated i.p. with rhGH (4000 µg/kg) or rhEPO (5000 IU/kg) 0/12/24 h after hypoxic exposure. After a regeneration period of 48 h or 7 d, cerebral mRNA expression of Vegf-A, its receptors and co-receptors, and selected tight junction proteins were determined using qRT-PCR and ELISA. Vessel structures were assessed by Pecam-1 and occludin (Ocln) IHC. While Vegf-A expression increased significantly with rhGH treatment (p < 0.01), expression of the Vegfr and TEK receptor tyrosine kinase (Tie-2) system remained unchanged. RhEPO increased Vegf-A (p < 0.05) and Angpt-2 (p < 0.05) expression. While hypoxia reduced the mean vessel area in the parietal cortex compared to controls (p < 0.05), rhGH and rhEPO prevented this reduction after 48 h of regeneration. Hypoxia significantly reduced the Ocln+ fraction of cortical vascular endothelial cells. Ocln signal intensity increased in the cortex in response to rhGH (p < 0.05) and in the cortex and hippocampus in response to rhEPO (p < 0.05). Our data indicate that rhGH and rhEPO have protective effects on hypoxia-induced BBB disruption and regenerative vascular effects during the post-hypoxic period in the developing brain.
Collapse
Affiliation(s)
- Simon Klepper
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
| | - Susan Jung
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
| | - Lara Dittmann
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
| | - Carol I. Geppert
- Institute of Pathology, Friedrich-Alexander Universität Erlangen-Nürnberg, Krankenhausstr. 8, 91054 Erlangen, Germany
| | - Arnd Hartmann
- Institute of Pathology, Friedrich-Alexander Universität Erlangen-Nürnberg, Krankenhausstr. 8, 91054 Erlangen, Germany
| | - Nicole Beier
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
| | - Regina Trollmann
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-8533753; Fax: +49-9131-8533389
| |
Collapse
|
6
|
Wei W, Hu P, Qin M, Chen G, Wang F, Yao S, Jin M, Xie Z, Zhang X. SIRT4 Is Highly Expressed in Retinal Müller Glial Cells. Front Neurosci 2022; 16:840443. [PMID: 35185463 PMCID: PMC8854368 DOI: 10.3389/fnins.2022.840443] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/13/2022] [Indexed: 11/18/2022] Open
Abstract
Sirtuin 4 (SIRT4) is one of seven mammalian sirtuins that possesses ADP-ribosyltransferase, lipoamidase and deacylase activities and plays indispensable role in metabolic regulation. However, the role of SIRT4 in the retina is not clearly understood. The purpose of this study was to explore the location and function of SIRT4 in the retina. Therefore, immunofluorescence was used to analyze the localization of SIRT4 in rat, mouse and human retinas. Western blotting was used to assess SIRT4 and glutamine synthetase (GS) protein expression at different developmental stages in C57BL/6 mice retinas. We further analyzed the retinal structure, electrophysiological function and the expression of GS protein in SIRT4-deficient mice. Excitotoxicity was caused by intravitreal injection of glutamate (50 nmol) in mice with long-term intraperitoneal injection of resveratrol (20 mg/Kg), and then retinas were subjected to Western blotting and paraffin section staining to analyze the effect of SIRT4 on excitotoxicity. We show that SIRT4 co-locates with Müller glial cell markers (GS and vimentin). The protein expression pattern of SIRT4 was similar to that of GS, and both increased with development. There were no significant retinal structure or electrophysiological function changes in 2-month SIRT4-deficient mice, while the expression of GS protein was decreased. Moreover, long-term administration of resveratrol can upregulate the expression of SIRT4 and GS while reducing the retinal injury caused by excessive glutamate. These results suggest that SIRT4 is highly expressed in retinal Müller glial cells and is relevant to the expression of GS. SIRT4 does not appear to be essential in retinal development, but resveratrol, as an activator of SIRT4, can upregulate GS protein expression and protect the retina from excitotoxicity.
Collapse
|
7
|
Cheng F, Cheng Y, Zhao X, An L, Yang L, Li Z, Zhang L, He R. NEDD4 E3 ubiquitin protein ligase serves an important role in cutaneous melanoma occurrence and development. Exp Ther Med 2021; 22:1382. [PMID: 34650630 PMCID: PMC8506948 DOI: 10.3892/etm.2021.10818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/25/2021] [Indexed: 11/18/2022] Open
Abstract
The present study aimed to discuss the effects and relative mechanisms of NEDD4 E3 ubiquitin protein ligase (NEDD4) in cutaneous melanoma (CMM) occurrence and development. Clinical cancer and adjacent normal tissues samples were collected to analyze pathological changes and protein expression of NEDD4. Moreover, small interfering (si)RNA was used to knockdown NEDD4 expression in SK-MEL-2 and Malme-3M cells. Cellular proliferation, apoptosis, invasiveness and migration were examined using colony formation, flow cytometric, Transwell and wound-healing assays, respectively. In addition, the relative mRNA and protein expression levels of NEDD4, notch receptor 1 (Notch1) and PTEN were evaluated via reverse transcription-quantitative (RT-q) PCR and western blotting. It was found that NEDD4 mRNA and protein expression were significantly upregulated (both P<0.01). Following NEDD4-knockdown, colony number was significantly decreased, while the apoptotic rate was significantly increased, the invasive cell number was significantly inhibited and the wound-healing capacity was significantly decreased. Following si-NEDD4 transfection, RT-qPCR and western blotting revealed that NEDD4 and Notch1 mRNA and protein expression levels were significantly downregulated, while those of PTEN were significantly upregulated in the SK-MEL-2 and Malme-3M cell lines. Collectively, the current results suggest that NEDD4-knockdown effectively suppressed CMM biological activity by regulating the Notch1/PTEN pathway in vitro.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Dermatology, Affiliated Xingtai People's Hospital of Hebei Medical University, Xingtai, Hebei 054001, P.R. China
| | - Yi Cheng
- Department of Dermatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiaoling Zhao
- Department of Dermatology, Affiliated Xingtai People's Hospital of Hebei Medical University, Xingtai, Hebei 054001, P.R. China
| | - Lihui An
- Department of Dermatology, Affiliated Xingtai People's Hospital of Hebei Medical University, Xingtai, Hebei 054001, P.R. China
| | - Linfang Yang
- Department of Dermatology, Affiliated Xingtai People's Hospital of Hebei Medical University, Xingtai, Hebei 054001, P.R. China
| | - Zihan Li
- Department of Dermatology, Affiliated Xingtai People's Hospital of Hebei Medical University, Xingtai, Hebei 054001, P.R. China
| | - Lei Zhang
- Department of Dermatology, The Second People's Hospital of Guiyang, Guiyang, Guizhou 550023, P.R. China
| | - Runzhi He
- Department of Neurosurgery, Affiliated Xingtai People's Hospital of Hebei Medical University, Xingtai, Hebei 054001, P.R. China
| |
Collapse
|
8
|
Prospects for the application of Müller glia and their derivatives in retinal regenerative therapies. Prog Retin Eye Res 2021; 85:100970. [PMID: 33930561 DOI: 10.1016/j.preteyeres.2021.100970] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/28/2021] [Accepted: 03/31/2021] [Indexed: 02/07/2023]
Abstract
Neural cell death is the main feature of all retinal degenerative disorders that lead to blindness. Despite therapeutic advances, progression of retinal disease cannot always be prevented, and once neuronal cell damage occurs, visual loss cannot be reversed. Recent research in the stem cell field, and the identification of Müller glia with stem cell characteristics in the human eye, have provided hope for the use of these cells in retinal therapies to restore vision. Müller glial cells, which are the major structural cells of the retina, play a very important role in retinal homeostasis during health and disease. They are responsible for the spontaneous retinal regeneration observed in zebrafish and lower vertebrates during early postnatal life, and despite the presence of Müller glia with stem cell characteristics in the adult mammalian retina, there is no evidence that they promote regeneration in humans. Like many other stem cells and neurons derived from pluripotent stem cells, Müller glia with stem cell potential do not differentiate into retinal neurons or integrate into the retina when transplanted into the vitreous of experimental animals with retinal degeneration. However, despite their lack of integration, grafted Müller glia have been shown to induce partial restoration of visual function in spontaneous or induced experimental models of photoreceptor or retinal ganglion cell damage. This improvement in visual function observed after Müller cell transplantation has been ascribed to the release of neuroprotective factors that promote the repair and survival of damaged neurons. Due to the development and availability of pluripotent stem cell lines for therapeutic uses, derivation of Müller cells from retinal organoids formed by iPSC and ESC has provided more realistic prospects for the application of these cells to retinal therapies. Several opportunities for research in the regenerative field have also been unlocked in recent years due to a better understanding of the genomic and proteomic profiles of the developing and regenerating retina in zebrafish, providing the basis for further studies of the human retina. In addition, the increased interest on the nature and function of cellular organelle release and the characterization of molecular components of exosomes released by Müller glia, may help us to design new approaches that could be applied to the development of more effective treatments for retinal degenerative diseases.
Collapse
|
9
|
Growth Hormone and the Auditory Pathway: Neuromodulation and Neuroregeneration. Int J Mol Sci 2021; 22:ijms22062829. [PMID: 33799503 PMCID: PMC7998811 DOI: 10.3390/ijms22062829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/26/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Growth hormone (GH) plays an important role in auditory development during the embryonic stage. Exogenous agents such as sound, noise, drugs or trauma, can induce the release of this hormone to perform a protective function and stimulate other mediators that protect the auditory pathway. In addition, GH deficiency conditions hearing loss or central auditory processing disorders. There are promising animal studies that reflect a possible regenerative role when exogenous GH is used in hearing impairments, demonstrated in in vivo and in vitro studies, and also, even a few studies show beneficial effects in humans presented and substantiated in the main text, although they should not exaggerate the main conclusions.
Collapse
|
10
|
Jung S, Terörde K, Dörr HG, Trollmann R. Recombinant Human Growth Hormone Activates Neuroprotective Growth Factors in Hypoxic Brain Injury in Neonatal Mice. Endocrinology 2021; 162:6129199. [PMID: 33545716 DOI: 10.1210/endocr/bqab008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 01/10/2023]
Abstract
Perinatal hypoxia severely disrupts cerebral metabolic and maturational programs beyond apoptotic cell death. Antiapoptotic treatments such as erythropoietin are suggested to improve outcomes in hypoxic brain injury; however, the results are controversial. We analyzed the neuroprotective effects of recombinant human growth hormone (rhGH) on regenerative mechanisms in the hypoxic developing mouse brain in comparison to controls. Using an established model of neonatal acute hypoxia (8% O2, 6 hours), P7 mice were treated intraperitoneally with rhGH (4000 µg/kg) 0, 12, and 24 hours after hypoxic exposure. After a regeneration period of 48 hours, expression of hypoxia-inducible neurotrophic factors (erythropoietin [EPO], vascular endothelial growth factor A [VEGF-A], insulin-like growth factors 1 and 2 [IGF-1/-2], IGF binding proteins) and proinflammatory markers was analyzed. In vitro experiments were performed using primary mouse cortical neurons (E14, DIV6). rhGH increased neuronal gene expression of EPO, IGF-1, and VEGF (P < .05) in vitro and diminished apoptosis of hypoxic neurons in a dose-dependent manner. In the developing brain, rhGH treatment led to a notable reduction of apoptosis in the subventricular zone and hippocampus (P < .05), abolished hypoxia-induced downregulation of IGF-1/IGF-2 expression (P < .05), and led to a significant accumulation of endogenous EPO protein and anti-inflammatory effects through modulation of interleukin-1β and tumor necrosis factor α signaling as well as upregulation of cerebral phosphorylated extracellularly regulated kinase 1/2 levels (ERK1/2). Indicating stabilizing effects on the blood-brain barrier (BBB), rhGH significantly modified cerebrovascular occludin expression. Thus, we conclude that rhGH mediates neuroprotective effects by the activation of endogenous neurotrophic growth factors and BBB stabilization. In addition, the modification of ERK1/2 pathways is involved in neuroprotective actions of rhGH. The present study adds further evidence that pharmacologic activation of neurotrophic growth factors may be a promising target for neonatal neuroprotection.
Collapse
Affiliation(s)
- Susan Jung
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Klara Terörde
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Helmuth-Günther Dörr
- Department of Pediatrics, Pediatric Endocrinology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Regina Trollmann
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
11
|
Delétage N, Le Douce J, Callizot N, Godfrin Y, Lemarchant S. SCO-spondin-derived Peptide Protects Neurons from Glutamate-induced Excitotoxicity. Neuroscience 2021; 463:317-336. [PMID: 33577953 DOI: 10.1016/j.neuroscience.2021.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/22/2022]
Abstract
Subcommissural organ (SCO)-spondin is a brain-specific glycoprotein produced during embryogenesis, that strongly contributes to neuronal development. The SCO becomes atrophic in adults, halting SCO-spondin production and its neuroprotective functions. Using rat and human neuronal cultures, we evaluated the neuroprotective effect of an innovative peptide derived from SCO-spondin against glutamate excitotoxicity. Primary neurons were exposed to glutamate and treated with the linear (NX210) and cyclic (NX210c) forms of the peptide. Neuronal survival and neurite networks were assessed using immunohistochemistry or biochemistry. The mechanism of action of both peptide forms was investigated by exposing neurons to inhibitors targeting receptors and intracellular mediators that trigger apoptosis, neuronal survival, or neurite growth. NX210c promoted neuronal survival and prevented neurite network retraction in rat cortical and hippocampal neurons, whereas NX210 was efficient only in neuronal survival (cortical neurons) or neurite networks (hippocampal neurons). They triggered neuroprotection via integrin receptors and γ-secretase substrate(s), activation of the PI3K/mTOR pathway and disruption of the apoptotic cascade. The neuroprotective effect of NX210c was confirmed in human cortical neurons via the reduction of lactate dehydrogenase release and recovery of normal basal levels of apoptotic cells. Together, these results show that NX210 and NX210c protect against glutamate neurotoxicity through common and distinct mechanisms of action and that, most often, NX210c is more efficient than NX210. Proof of concept in central nervous system animal models are under investigation to evaluate the neuroprotective action of SCO-spondin-derived peptide.
Collapse
Affiliation(s)
| | | | - Noëlle Callizot
- Neuro-Sys, 410 Chemin Départemental 60, 13120 Gardanne, France.
| | - Yann Godfrin
- Axoltis Pharma, 60 Avenue Rockefeller, 69008 Lyon, France; Godfrin Life Sciences, 8 impasse de la source, 69300 Caluire-et-Cuire, France.
| | | |
Collapse
|
12
|
Corona R, Jayakumar P, Carbajo Mata MA, Del Valle-Díaz MF, Luna-García LA, Morales T. Sexually dimorphic effects of prolactin treatment on the onset of puberty and olfactory function in mice. Gen Comp Endocrinol 2021; 301:113652. [PMID: 33122037 DOI: 10.1016/j.ygcen.2020.113652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 11/28/2022]
Abstract
The onset of puberty is associated with the psychophysiological maturation of the adolescent to an adult capable of reproduction when olfactory signals play an important role. This period begins with the secretion of the gonadotropin-releasing hormone (GnRH) from GnRH neurons within the hypothalamus. This is regulated by kisspeptin neurons that express high levels of transmembrane prolactin receptors (PRLR) that bind to and are activated by prolactin (PRL). The elevated levels of serum PRL found during lactation, or caused by chronic PRL infusion, decreases the secretion of gonadotropins and kisspeptin and compromised the estrous cyclicity and the ovulation. In the present work, we aimed to evaluate the effects of either increased or decreased PRL circulating levels within the peripubertal murine brain by administration of PRL or treatment with cabergoline (Cab) respectively. We showed that either treatment delayed the onset of puberty in females, but not in males. This was associated with the augmentation of the PRL receptor (Prlr) mRNA expression in the arcuate nucleus and decreased Kiss1 expression in the anteroventral periventricular zone. Then, during adulthood, we assessed the activation of the mitral and granular cells of the main (MOB) and accessory olfactory bulb (AOB) by cFos immunoreactivity (ir) after the exposure to soiled bedding of the opposite sex. In the MOB, the PRL treatment promoted an increased cFos-ir of the mitral cells of males and females. In the granular cells of male of either treatment an augmented activation was observed. In the AOB, an impaired cFos-ir was observed in PRL and Cab treated females after exposure to male soiled bedding. However, in males, only Cab impaired its activation. No effects were observed in the AOB-mitral cells. In conclusion, our results demonstrate that PRL contributes to pubertal development and maturation of the MOB-AOB during the murine juvenile period in a sex-dependent way.
Collapse
Affiliation(s)
- Rebeca Corona
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Mexico.
| | - Preethi Jayakumar
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Mexico
| | | | | | | | - Teresa Morales
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Mexico
| |
Collapse
|
13
|
Lin C, Qian P, Zhang Y, Liu Z, Dai K, Sun D. Plac1 promotes nasopharyngeal carcinoma cells proliferation, migration and invasion via Furin/NICD/PTEN pathway. Tissue Cell 2021; 69:101480. [PMID: 33418237 DOI: 10.1016/j.tice.2020.101480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 01/17/2023]
Abstract
Placenta-specific protein 1 (Plac1) has critical functions in multiple human malignancies, but its role in nasopharyngeal carcinoma (NPC) was unclear. Clinical samples of NPC and adjacent normal tissue were collected. Plac1 expressions in both tissues and cells were measured. After cell transfection, NPC cell viability, proliferation, migration and invasion were detected using cell counting kit-8 (CCK-8) assay, colony formation assay, scratch assay and Transwell assay. Relative expressions of Plac1 and proteins related to migration and invasion (E-Cadherin, N-cadherin, Matrix metalloproteinase2 (MMP2), and MMP9), Furin/Notch1 intracellular domain (NICD)/phosphate and tension homology (PTEN) pathway (NICD, PTEN, phosphorylated-Akt (p-Akt), Akt) were quantified by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot as needed. The interaction between Plac1 and Furin, a member of Furin/NICD/PTEN Pathway, was analyzed using co-Immunoprecipitation (co-IP) assay. Plac1 expression was upregulated in both NPC tissue and cells. Overexpressed Plac1 promoted Plac1 and Furin expressions and increased cell viability, proliferation, migration and invasion of NPC cells, while silencing Plac1 showed the opposite effects. Plac1 interacted with Furin, overexpression of Furin reversed the inhibitory effects of silencing Plac1 on NPC cell proliferation, migration, and invasion, and also reversed the effects of silencing Plac1 on Furin/NICD/PTEN pathway-, cell migration-, and invasion-related protein expressions. Plac1 promoted NPC cell proliferation, migration and invasion via Furin/NICD/PTEN Pathway. The findings of this study provide a possible therapeutic method for NPC treatment.
Collapse
Affiliation(s)
- Chuanbao Lin
- Otorhinolaryngology Head and Neck Surgery, Weifang Ruiqing Hospital, Weicheng District, Weifang City, Shandong Province, 261021, China
| | - Pengfei Qian
- Department of Pediatrics, W.F. Maternal and Child Health Hospital, Weicheng District, Weifang City, Shandong Province, 261021, China
| | - Yan Zhang
- Department of Pathology, Z.C. Maternal and Child Health Hospital, Zhucheng City, Weifang City, Shandong Province, 262200, China
| | - Zhihui Liu
- Otorhinolaryngology Head and Neck Surgery, Weifang Ruiqing Hospital, Weicheng District, Weifang City, Shandong Province, 261021, China
| | - Kun Dai
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University, School of Medicine, Chongqing Road, Shanghai, 200025, China
| | - Dawei Sun
- Otorhinolaryngology Head and Neck Surgery, Weifang Ruiqing Hospital, Weicheng District, Weifang City, Shandong Province, 261021, China.
| |
Collapse
|
14
|
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
|
15
|
Chou Y, Zhang B, Gan L, Ma J, Zhong Y. Clinical efficacy of optical coherence tomography in sellar mass lesions: a meta-analysis. Pituitary 2020; 23:733-744. [PMID: 32767247 DOI: 10.1007/s11102-020-01072-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Although optical coherence tomography (OCT) of the eyes has been studied to detect and monitor sellar masses, there is no recommendation for selecting the most effective measurement of OCT in clinical practice. Thus, we conducted a meta-analysis to examine the efficacy of OCT in sellar mass lesions. METHODS We conducted a literature search in PubMed and EMBASE through April 26, 2020. The primary outcomes were the thickness of the peripapillary retinal nerve fiber layer (pRNFL) and the macular ganglion cell complex (mGCC). The secondary outcomes included the thickness of the macular ganglion cell and inner plexiform layer (mGCIPL) and macular thickness. Random-effects models were used in all meta-analyses. Additionally, we conducted meta-regressions and subgroup analyses. RESULTS We included 22 studies, involving 1347 eyes of patients and 1198 eyes of controls. When compared with the control group, the reductions in pRNFL, mGCC and macular thickness in the patient group were significantly different, whereas significant thinning of the mGCIPL was restricted to the nasal hemiretina. Furthermore, we found that before visual field (VF) defects occurred, significant thinning of the pRNFL and mGCC thickness could be detected by OCT. The change in OCT parameters also showed different patterns in different types of pituitary adenomas. CONCLUSIONS Sellar mass lesions were associated with the changes in OCT measurements. The characteristic patterns of the OCT parameters may refine the diagnostic accuracy. Moreover, the alterations of OCT metrics before VF defects indicate the efficacy of OCT in early detection. Different types of pituitary adenomas may vary in OCT measurements, and their specific features warrant further research efforts.
Collapse
Affiliation(s)
- Yuyu Chou
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Bilei Zhang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Linyang Gan
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jin Ma
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yong Zhong
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
16
|
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
|
17
|
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
|