1
|
Lopes de Lima I, Ap. Rosini Silva A, Brites C, Angelo da Silva Miyaguti N, Raposo Passos Mansoldo F, Vaz Nunes S, Henrique Godoy Sanches P, Regiani Cataldi T, Pais de Carvalho C, Reis da Silva A, Ribeiro da Rosa J, Magalhães Borges M, Vilarindo Oliveira W, Canevari TC, Beatriz Vermelho A, Nogueira Eberlin M, M. Porcari A. Mass Spectrometry-Based Metabolomics Reveals a Salivary Signature for Low-Severity COVID-19. Int J Mol Sci 2024; 25:11899. [PMID: 39595969 PMCID: PMC11593410 DOI: 10.3390/ijms252211899] [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: 07/16/2024] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 11/28/2024] Open
Abstract
Omics approaches were extensively applied during the coronavirus disease 2019 (COVID-19) pandemic to understand the disease, identify biomarkers with diagnostic and prognostic value, and discover new molecular targets for medications. COVID-19 continues to challenge the healthcare system as the virus mutates, becoming more transmissible or adept at evading the immune system, causing resurgent epidemic waves over the last few years. In this study, we used saliva from volunteers who were negative and positive for COVID-19 when Omicron and its variants became dominant. We applied a direct solid-phase extraction approach followed by non-target metabolomics analysis to identify potential salivary signatures of hospital-recruited volunteers to establish a model for COVID-19 screening. Our model, which aimed to differentiate COVID-19-positive individuals from controls in a hospital setting, was based on 39 compounds and achieved high sensitivity (85%/100%), specificity (82%/84%), and accuracy (84%/92%) in training and validation sets, respectively. The salivary diagnostic signatures were mainly composed of amino acids and lipids and were related to a heightened innate immune antiviral response and an attenuated inflammatory profile. The higher abundance of thyrotropin-releasing hormone in the COVID-19 positive group highlighted the endocrine imbalance in low-severity disease, as first reported here, underscoring the need for further studies in this area.
Collapse
Affiliation(s)
- Iasmim Lopes de Lima
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Alex Ap. Rosini Silva
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| | - Carlos Brites
- LAPI-Laboratory of Research in Infectology, University Hospital Professor Edgard Santos (HUPES), Federal University of Bahia (UFBA), Salvador 40110-060, BA, Brazil; (C.B.); (S.V.N.)
| | - Natália Angelo da Silva Miyaguti
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| | - Felipe Raposo Passos Mansoldo
- BIOINOVAR-Biotechnology Laboratories, Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil; (F.R.P.M.); (A.B.V.)
| | - Sara Vaz Nunes
- LAPI-Laboratory of Research in Infectology, University Hospital Professor Edgard Santos (HUPES), Federal University of Bahia (UFBA), Salvador 40110-060, BA, Brazil; (C.B.); (S.V.N.)
| | - Pedro Henrique Godoy Sanches
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| | - Thais Regiani Cataldi
- Department of Genetics, Luiz de Queiroz College of Agriculture, University of São Paulo (USP/ESALQ), Piracicaba 13418-900, SP, Brazil;
| | - Caroline Pais de Carvalho
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Adriano Reis da Silva
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Jonas Ribeiro da Rosa
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| | - Mariana Magalhães Borges
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Wellisson Vilarindo Oliveira
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Thiago Cruz Canevari
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
| | - Alane Beatriz Vermelho
- BIOINOVAR-Biotechnology Laboratories, Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil; (F.R.P.M.); (A.B.V.)
| | - Marcos Nogueira Eberlin
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Andreia M. Porcari
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| |
Collapse
|
2
|
Mao R, Zhou F, Hong Y, Li Y, Zhu C, Jin L, Li S. CRH upregulates supervillin through ERK and AKT pathways to promote bladder cancer cell migration. Cell Biol Int 2024; 48:1743-1754. [PMID: 39090812 DOI: 10.1002/cbin.12227] [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: 01/11/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Corticotropin-releasing hormone (CRH) has been well documented playing a role in the regulation of cellular processes, immune responses, and inflammatory processes that can influence the occurrence and development of tumors. Supervillin (SVIL) is a membrane-associated and actin-binding protein, which is actively involved in the proliferation, spread, and migration of cancer cells. This work investigated CRH's influence on bladder cancer cells' migration and relevant mechanisms. By using human bladder cancer cells T24 and RT4 in wound healing experiments and transwell assay, we found that the migration ability of the T24 cells was significantly increased after CRH treatment. In vivo experiments showed that CRH significantly promoted the metastases of T24 cells in cell line-derived xenograft (CDX) mouse model. Interestingly, downregulation of SVIL by SVIL-specifc small hairpin RNAs significantly reduced the promoting effect of CRH on bladder cancer cell migration. Furthermore, CRH significantly increased SVIL messenger RNA and protein expression in T24 cells, accompanied with AKT and ERK phosphorylation in T24 cells. Pretreatment with AKT inhibitor (MK2206) blocked the CRH-induced SVIL expression and ERK phosphorylation. Also, inhibition of ERK signaling pathway by U0126 significantly reduced the CRH-induced SVIL expression and AKT phosphorylation. It suggested that cross-talking between AKT and ERK pathways was involved in the effect of CRH on SVIL. Taken together, we demonstrated that CRH induced migration of bladder cancer cells, in which AKT and ERK pathways -SVIL played a key role.
Collapse
Affiliation(s)
- Rongchen Mao
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Feier Zhou
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yali Hong
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yongqi Li
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Chao Zhu
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lai Jin
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Shengnan Li
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Vassallo A, Di Filippo L, Frara S, Bertoli M, Pagani M, Presciuttini B. New onset of Graves' disease after controlled ovarian stimulation: A case report and brief literature review. Int J Gynaecol Obstet 2024. [PMID: 39396110 DOI: 10.1002/ijgo.15951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/22/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
De novo onset of Graves' disease (GD) after controlled ovarian stimulation (OS) is exceptional. Only one case of progression to GD after OS in a patient with pre-existing subclinical hyperthyroidism has been reported. We describe the case of a patient with neither previous thyroid disorders nor autoimmunity who developed GD after OS for primary infertility. A 40-year-old woman with primary infertility underwent four cycles of OS. Her thyroid function performed before the last cycle was unremarkable (thyroid stimulating hormone [TSH] 1.9 mU/L, fT4 1.3 ng/dL, fT3 2.4 pg/mL), and thyroid autoimmunity was negative (anti-thyroperoxidase antibodies and anti-thyroglobuline antibodies). Six weeks after the last cycle she developed overt thyrotoxicosis (TSH < 0.005 mU/L, fT4 4.79 ng/dL, fT3 15.6 pg/mL) with anti-thyrotropin receptor antibodies (TRAb) positivity (9.2 IU/L). She was diagnosed with GD and anti-thyroid therapy was instituted. After 1 year of treatment, thyroid function was still suboptimal (TSH 0.2 mU/L, fT4 1.04 ng/dL, fT3 2.2 pg/mL), and TRAb titer still elevated (8.75 IU/L). Despite her desire to achieve pregnancy, a further cycle of OS was postponed until complete remission of thyroid dysfunction and withdrawal of anti-thyroid therapy. Although TSH assay after OS is not recommended in euthyroid women without autoimmunity, in the presence of hyperthyroid symptoms throughout OS it is advisable to evaluate thyroid function and TRAb. It is advisable to carefully evaluate the course of GD before proceeding with further courses of OS that could lead to its exacerbation or recurrence. In cases where a strong desire for pregnancy persists, thyroidectomy may be proposed.
Collapse
Affiliation(s)
- Alberto Vassallo
- Institute of Endocrine and Metabolic Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - Luigi Di Filippo
- Institute of Endocrine and Metabolic Sciences, Vita-Salute San Raffaele University, Milan, Italy
- Endocrinology Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Stefano Frara
- Institute of Endocrine and Metabolic Sciences, Vita-Salute San Raffaele University, Milan, Italy
- Endocrinology Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimo Bertoli
- Infertility Unit, Department Obstetrics and Gynecology, ASST Mantova, Mantua, Italy
| | | | - Barbara Presciuttini
- Medical Department, ASST Mantova, Mantua, Italy
- Endocrinology Unit, Medical Department, ASST Mantova, Mantua, Italy
| |
Collapse
|
4
|
Wójcik M, Bordoni B, Siatkowski I, Żekanowska E. The Effect of Craniosacral Therapy on Blood Levels of Stress Hormones in Male Firefighter Cadets: A Randomized Clinical Trial. Behav Sci (Basel) 2023; 13:914. [PMID: 37998661 PMCID: PMC10669461 DOI: 10.3390/bs13110914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
(1) Background: Fire department cadets preparing to become firefighters and paramedics experience high levels of stress when participating in incidents like traffic accidents and fires. Stress adversely affects health, and coping with it proves difficult. Unfortunately, there is no single method that reduces stress completely in humans. One non-invasive method for lowering stress hormone levels is craniosacral therapy. (2) Methods: Fifty-seven firefighting cadets aged 18-24 years (21.63 ± 1.41) participated in the study. They were randomly assigned to either a test group or a control group. Participants' blood levels of cortisol and CRH (corticotropin-releasing hormone) were assessed before and after the study. The study group underwent 5-week craniosacral therapy (1× per week). (3) Results: The Kruskal-Wallis test showed that the therapy group's results were statistically significant for CRH values (p-value = 0.00067) and for cortisol values (p-value ≤ 0.0001). Wilxocon and Dunn tests showed statistical significance for cortisol after CS therapy between the control and study groups (p = 0.0377), and for CRH between the control and study groups before (p = 0.00634) and after the study (p = 0.000887), and in the study group before and after the study (p = 0.0101). (4) Conclusions: The application of craniosacral therapy reduced stress hormone levels in male firefighter cadets. The results indicate that craniosacral therapy (five sessions, one per week) has an effect on the reduction of stress hormones.
Collapse
Affiliation(s)
- Małgorzata Wójcik
- Department of Physiotherapy, Faculty of Sport Sciences in Gorzow Wielkopolski, Poznan University of Physical Education, 61-871 Poznan, Poland
| | - Bruno Bordoni
- Department of Cardiology, Foundation Don Carlo Gnocchi IRCCS, Institute of Hospitalization and Care, 20100 Milan, Italy;
| | - Idzi Siatkowski
- Department of Mathematical and Statistical Methods, Poznan University of Life Science, 60-637 Poznan, Poland;
| | - Ewa Żekanowska
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland;
| |
Collapse
|
5
|
Mumme H, Thomas BE, Bhasin SS, Krishnan U, Dwivedi B, Perumalla P, Sarkar D, Ulukaya GB, Sabnis HS, Park SI, DeRyckere D, Raikar SS, Pauly M, Summers RJ, Castellino SM, Wechsler DS, Porter CC, Graham DK, Bhasin M. Single-cell analysis reveals altered tumor microenvironments of relapse- and remission-associated pediatric acute myeloid leukemia. Nat Commun 2023; 14:6209. [PMID: 37798266 PMCID: PMC10556066 DOI: 10.1038/s41467-023-41994-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/25/2023] [Indexed: 10/07/2023] Open
Abstract
Acute myeloid leukemia (AML) microenvironment exhibits cellular and molecular differences among various subtypes. Here, we utilize single-cell RNA sequencing (scRNA-seq) to analyze pediatric AML bone marrow (BM) samples from diagnosis (Dx), end of induction (EOI), and relapse timepoints. Analysis of Dx, EOI scRNA-seq, and TARGET AML RNA-seq datasets reveals an AML blasts-associated 7-gene signature (CLEC11A, PRAME, AZU1, NREP, ARMH1, C1QBP, TRH), which we validate on independent datasets. The analysis reveals distinct clusters of Dx relapse- and continuous complete remission (CCR)-associated AML-blasts with differential expression of genes associated with survival. At Dx, relapse-associated samples have more exhausted T cells while CCR-associated samples have more inflammatory M1 macrophages. Post-therapy EOI residual blasts overexpress fatty acid oxidation, tumor growth, and stemness genes. Also, a post-therapy T-cell cluster associated with relapse samples exhibits downregulation of MHC Class I and T-cell regulatory genes. Altogether, this study deeply characterizes pediatric AML relapse- and CCR-associated samples to provide insights into the BM microenvironment landscape.
Collapse
Affiliation(s)
- Hope Mumme
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Beena E Thomas
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Swati S Bhasin
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Upaasana Krishnan
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Bhakti Dwivedi
- Department of Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Pruthvi Perumalla
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Debasree Sarkar
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Gulay B Ulukaya
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Himalee S Sabnis
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sunita I Park
- Department of Pathology, Children's Healthcare of Atlanta, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sunil S Raikar
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Melinda Pauly
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan J Summers
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sharon M Castellino
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel S Wechsler
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher C Porter
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Manoj Bhasin
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA.
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA.
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
6
|
Pedroza-García KA, Calderón-Vallejo D, Cervantes-García D, Quintanar-Stephano A, Salinas E, Quintanar JL. Effect of Leuprolide Acetate, a GnRH Agonist, on Neuroinflammation and Anxiety-Like Behavior after Mild Hypoxic-Ischemic Encephalopathy in Rat Model. Neuroimmunomodulation 2023; 30:206-212. [PMID: 37607495 PMCID: PMC10614564 DOI: 10.1159/000533388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Mild hypoxic-ischemic encephalopathy (HIE) is a condition that predisposes to negative outcomes such as neuroanatomical injury, mood disorders, and motor or cognitive disabilities. The neuroinflammation plays an important role in the neurological damage; therefore, reducing it could provide neuroprotection. The leuprolide acetate (LA) has shown to have neuroregenerative and immunomodulator properties in other nervous system injuries. OBJECTIVE The aim of this study was to evaluate the immunomodulatory effect of LA in the acute phase of mild HIE and its effects in motor activity and behavior in a subacute phase. METHOD Forty-five Wistar rats on postnatal day 7 were divided into Sham, HIE treated with saline solution (HIE-SS), and HIE-LA. The HIE was performed cutting of the right carotid artery followed by 60 min of hypoxia. The expression of the inflammatory cytokines interleukin (IL)-1β, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and the chemokine CXCL-1 were evaluated 72 h after HIE by RT-qPCR and the motor activity and behavior were evaluated by open field test at postnatal day 33. RESULTS HIE-SS animals showed increased expression of IL-1β, TNF-α, IFN-γ, and CXCL-1 genes in injured tissue. However, the HIE-LA group exhibited similar expression levels of IL-1β and TNF-α to the Sham group, while IFN-γ and CXCL-1 mRNA expression were attenuated with LA treatment. LA treatment also prevented anxiety-like behavior in the open field test. CONCLUSION Treatment with LA partially reverses HIE-induced neuroinflammation and prevents anxiety-like behavior in neonatal rats.
Collapse
Affiliation(s)
- Karina Alejandra Pedroza-García
- Laboratorio de Neurofisiología, Departamento de Fisiología y Farmacología, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Denisse Calderón-Vallejo
- Laboratorio de Neurofisiología, Departamento de Fisiología y Farmacología, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
- Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Daniel Cervantes-García
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
- Consejo Nacional de Ciencia y Tecnología, Mexico City, Mexico
| | - Andrés Quintanar-Stephano
- Laboratorio de Neuroendocrinología, Departamento de Fisiología y Farmacología, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Eva Salinas
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - J. Luis Quintanar
- Laboratorio de Neurofisiología, Departamento de Fisiología y Farmacología, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
7
|
Valli E, Dalotto-Moreno T, Sterle HA, Méndez-Huergo SP, Paulazo MA, García SI, Pirola CJ, Klecha AJ, Rabinovich GA, Cremaschi GA. Hypothyroidism-associated immunosuppression involves induction of galectin-1-producing regulatory T cells. FASEB J 2023; 37:e22865. [PMID: 36934391 DOI: 10.1096/fj.202200884r] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 01/31/2023] [Accepted: 02/27/2023] [Indexed: 03/20/2023]
Abstract
Hypothyroidism exerts deleterious effects on immunity, but the precise role of the hypothalamic-pituitary-thyroid (HPT) axis in immunoregulatory and tolerogenic programs is barely understood. Here, we investigated the mechanisms underlying hypothyroid-related immunosuppression by examining the regulatory role of components of the HPT axis. We first analyzed lymphocyte activity in mice overexpressing the TRH gene (Tg-Trh). T cells from Tg-Trh showed increased proliferation than wild-type (WT) euthyroid mice in response to polyclonal activation. The release of Th1 pro-inflammatory cytokines was also increased in Tg-Trh and TSH levels correlated with T-cell proliferation. To gain further mechanistic insights into hypothyroidism-related immunosuppression, we evaluated T-cell subpopulations in lymphoid tissues of hypothyroid and control mice. No differences were observed in CD3/CD19 or CD4/CD8 ratios between these strains. However, the frequency of regulatory T cells (Tregs) was significantly increased in hypothyroid mice, and not in Tg-Trh mice. Accordingly, in vitro Tregs differentiation was more pronounced in naïve T cells isolated from hypothyroid mice. Since Tregs overexpress galectin-1 (Gal-1) and mice lacking this lectin (Lgals1-/- ) show reduced Treg function, we investigated the involvement of this immunoregulatory lectin in the control of Tregs in settings of hypothyroidism. Increased T lymphocyte reactivity and reduced frequency of Tregs were found in hypothyroid Lgals1-/- mice when compared to hypothyroid WT animals. This effect was rescued by the addition of recombinant Gal-1. Finally, increased expression of Gal-1 was found in Tregs purified from hypothyroid WT mice compared with their euthyroid counterpart. Thus, a substantial increase in the frequency and activity of Gal-1-expressing Tregs underlies immunosuppression associated with hypothyroid conditions, with critical implications in immunopathology, metabolic disorders, and cancer.
Collapse
Affiliation(s)
- Eduardo Valli
- Laboratorio de Neuroinmunomodulación y Oncología Molecular, Instituto de Investigaciones Biomédicas (BIOMED), Pontificia Universidad Católica Argentina (UCA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Tomás Dalotto-Moreno
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Helena A Sterle
- Laboratorio de Neuroinmunomodulación y Oncología Molecular, Instituto de Investigaciones Biomédicas (BIOMED), Pontificia Universidad Católica Argentina (UCA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Santiago P Méndez-Huergo
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - María A Paulazo
- Laboratorio de Neuroinmunomodulación y Oncología Molecular, Instituto de Investigaciones Biomédicas (BIOMED), Pontificia Universidad Católica Argentina (UCA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Silvia I García
- Facultad de Medicina, Instituto de investigaciones Médicas Alfredo Lanari, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Departamento de Cardiología Molecular., Instituto de Investigaciones Médicas (IDIM), Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Carlos J Pirola
- Biología de Sistemas de Enfermedades Complejas, Centro de Altos Estudios en Ciencias Humanas y de la Salud (CAECIHS), Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Alicia J Klecha
- Laboratorio de Neuroinmunomodulación y Oncología Molecular, Instituto de Investigaciones Biomédicas (BIOMED), Pontificia Universidad Católica Argentina (UCA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Graciela A Cremaschi
- Laboratorio de Neuroinmunomodulación y Oncología Molecular, Instituto de Investigaciones Biomédicas (BIOMED), Pontificia Universidad Católica Argentina (UCA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
8
|
Chuphal B, Sathoria P, Rai U, Roy B. Crosstalk between reproductive and immune systems: the teleostean perspective. JOURNAL OF FISH BIOLOGY 2023; 102:302-316. [PMID: 36477945 DOI: 10.1111/jfb.15284] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/30/2022] [Indexed: 06/17/2023]
Abstract
The bidirectional interaction between the hypothalamic-pituitary-gonadal (HPG) axis and the immune system plays a crucial role in the adaptation of an organism to its environment, its survival and the continuance of a species. Nonetheless, very little is known about this interaction among teleost, the largest group of extant vertebrates. Fishes being seasonal breeders, their immune system is exposed to seasonally changing levels of HPG hormones. On the contrary, the presence and infiltration of leukocytes, the expression of pattern recognition receptors as well as cytokines in gonads suggest their key role in teleostean gametogenesis as in the case of mammals. Moreover, the modulation of gametogenesis and steroidogenesis by lipopolysaccharide implicates the pathological significance of inflammation on reproduction. Thus, it is important to engage in the understanding of the interaction between these two important physiological systems, not only from a phylogenetic perspective but also due to the importance of fish as an important economic resource. In view of this, the authors have reviewed the crosstalk between the reproductive and immune systems in teleosts and tried to explore the importance of this interaction in their survival and reproductive fitness.
Collapse
Affiliation(s)
- Bhawna Chuphal
- Department of Zoology, University of Delhi, Delhi, India
| | - Priyanka Sathoria
- Department of Zoology, Maitreyi College, University of Delhi, Delhi, India
| | - Umesh Rai
- University of Jammu, Jammu, Jammu and Kashmir, India
| | - Brototi Roy
- Department of Zoology, Maitreyi College, University of Delhi, Delhi, India
| |
Collapse
|
9
|
Marin L, Ambrosini G, Noventa M, Filippi F, Ragazzi E, Dessole F, Capobianco G, Andrisani A. Thyroid Dysfunction after Gonadotropin-Releasing Hormone Agonist Administration in Women with Thyroid Autoimmunity. Int J Endocrinol 2022; 2022:6331657. [PMID: 35465072 PMCID: PMC9019438 DOI: 10.1155/2022/6331657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/29/2022] [Indexed: 01/19/2023] Open
Abstract
GnRH agonists (GnRHa) are a useful tool for pretreatment before artificial endometrial preparation for frozen-thawed embryo-transfer (FET). Their prolonged administration has been associated with thyroid dysfunction, both hyper and hypothyroidism. The aim of this study is to investigate the impact of GnRHa administration on thyroid function in women undergoing artificial endometrial preparation. Seventy-eight euthyroid women undergoing endometrial preparation with hormone replacement for FET were retrospectively reviewed. They were divided into two groups according to pretreatment with GnRHa (group A, 42 women) or with an oral contraceptive (group B, 36 women). Group A was subsequently divided into two subgroups according to thyroid autoimmunity presence. Thyroid function has been evaluated and compared among groups and subgroups. Our results did not show any statistically significant differences in age, body mass index, and basal thyroid stimulating hormone (TSH). Total estradiol dosage, duration of treatment, and endometrial thickness were comparable among groups. When TSH was measured 14 days after embryo transfer, no significant differences between the two groups were reported. Among women of group A, TSH was significantly higher only in women with thyroid autoimmunity. GnRHa seems to be associated with thyroid dysfunction in women with thyroid autoimmunity undergoing hormone replacement therapy for FET.
Collapse
Affiliation(s)
- Loris Marin
- Department of Women's and Children's Health, University of Padua, Padua 35100, Italy
| | - Guido Ambrosini
- Department of Women's and Children's Health, University of Padua, Padua 35100, Italy
| | - Marco Noventa
- Department of Women's and Children's Health, University of Padua, Padua 35100, Italy
| | - Flavia Filippi
- Department of Women's and Children's Health, University of Padua, Padua 35100, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35100, Italy
| | - Francesco Dessole
- Department of Surgical, Microsurgical and Medical Sciences, Gynecologic and Obstetric Clinic, University of Sassari, Sassari 07100, Italy
| | - Giampiero Capobianco
- Department of Surgical, Microsurgical and Medical Sciences, Gynecologic and Obstetric Clinic, University of Sassari, Sassari 07100, Italy
| | - Alessandra Andrisani
- Department of Women's and Children's Health, University of Padua, Padua 35100, Italy
| |
Collapse
|
10
|
Wang Z, Cheng W, Zhao Z, Wang Z, Zhang C, Li G, Wu A, Jiang T. Comparative profiling of immune genes improves the prognoses of lower grade gliomas. Cancer Biol Med 2021; 19:j.issn.2095-3941.2021.0173. [PMID: 34623790 PMCID: PMC9088193 DOI: 10.20892/j.issn.2095-3941.2021.0173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/16/2021] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Lower grade gliomas (LGGs), classified as World Health Organization (WHO) grade II and grade III gliomas, comprise a heterogeneous group with a median survival time ranging from 4-13 years. Accurate prediction of the survival times of LGGs remains a major challenge in clinical practice. METHODS We reviewed the expression data of 865 LGG patients from 5 transcriptomics cohorts. The comparative profile of immune genes was analyzed for signature identification and validation. In-house RNAseq and microarray data from the Chinese Glioma Genome Atlas (CGGA) dataset were used as training and internal validation cohorts, respectively. The samples from The Cancer Genome Atlas (TCGA) and GSE16011 cohorts were used as external validation cohorts, and the real-time PCR of frozen LGG tissue samples (n = 36) were used for clinical validation. RESULTS A total of 2,214 immune genes were subjected to pairwise comparison to generate 2,449,791 immune-related gene pairs (IGPs). A total of 402 IGPs were identified with prognostic values for LGGs. The HOXA9-related and CRH-related scores facilitated identification of patients with different prognoses. An immune signature based on 10 IGPs was constructed to stratify patients into low and high risk groups, exhibiting different clinical outcomes. A nomogram, combining immune signature, 1p/19q status, and tumor grade, was able to predict the overall survival (OS) with c-indices of 0.85, 0.80, 0.80, 0.79, and 0.75 in the training, internal validation, external validation, and tissue sample cohorts, respectively. CONCLUSIONS This study was the first to report a comparative profiling of immune genes in large LGG cohorts. A promising individualized immune signature was developed to estimate the survival time for LGG patients.
Collapse
Affiliation(s)
- Zhiliang Wang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China
| | - Wen Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zheng Zhao
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China
| | - Zheng Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Chuanbao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Guanzhang Li
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China
| | - Anhua Wu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
11
|
Pereira G, Gillies H, Chanda S, Corbett M, Vernon SD, Milani T, Bateman L. Acute Corticotropin-Releasing Factor Receptor Type 2 Agonism Results in Sustained Symptom Improvement in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Front Syst Neurosci 2021; 15:698240. [PMID: 34539356 PMCID: PMC8441022 DOI: 10.3389/fnsys.2021.698240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex multi-symptom disease with widespread evidence of disrupted systems. The authors hypothesize that it is caused by the upregulation of the corticotropin-releasing factor receptor type 2 (CRFR2) in the raphé nuclei and limbic system, which impairs the ability to maintain homeostasis. The authors propose utilizing agonist-mediated receptor endocytosis to downregulate CRFR2. Materials and Methods This open-label trial tested the safety, tolerability and efficacy of an acute dose of CT38s (a short-lived, CRFR2-selective agonist, with no known off-target activity) in 14 ME/CFS patients. CT38s was subcutaneously-infused at one of four dose-levels (i.e., infusion rates of 0.01, 0.03, 0.06, and 0.20 μg/kg/h), for a maximum of 10.5 h. Effect was measured as the pre-/post-treatment change in the mean 28-day total daily symptom score (TDSS), which aggregated 13 individual patient-reported symptoms. Results ME/CFS patients were significantly more sensitive to the transient hemodynamic effects of CRFR2 stimulation than healthy subjects in a prior trial, supporting the hypothesized CRFR2 upregulation. Adverse events were generally mild, resolved without intervention, and difficult to distinguish from ME/CFS symptoms, supporting a CRFR2 role in the disease. The acute dose of CT38s was associated with an improvement in mean TDSS that was sustained (over at least 28 days post-treatment) and correlated with both total exposure and pre-treatment symptom severity. At an infusion rate of 0.03 μg/kg/h, mean TDSS improved by -7.5 ± 1.9 (or -25.7%, p = 0.009), with all monitored symptoms improving. Conclusion The trial supports the hypothesis that CRFR2 is upregulated in ME/CFS, and that acute CRFR2 agonism may be a viable treatment approach warranting further study. Clinical Trial Registration ClinicalTrials.gov, identifier NCT03613129.
Collapse
Affiliation(s)
| | | | | | | | | | - Tina Milani
- Bateman Horne Center, Salt Lake City, UT, United States
| | | |
Collapse
|
12
|
Hamed SA, Attiah FA, Abdulhamid SK, Fawzy M. Behavioral assessment of children and adolescents with Graves' disease: A prospective study. PLoS One 2021; 16:e0248937. [PMID: 33914772 PMCID: PMC8084231 DOI: 10.1371/journal.pone.0248937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/08/2021] [Indexed: 11/18/2022] Open
Abstract
Previous studies have identified frequent comorbid neuropsychiatric disorders and conditions in adults with thyrotoxicosis. These studies are scarce or even lacking in pediatric population. This work aimed to study the behavior of children and adolescents with Graves’ disease (GD). This study included 35 children with GD (boys = 15; girls = 25; mean age: 11.45±1.50yrs) and 40 healthy children (boys = 20; girls = 20; mean age: 12.54±1.62yrs). Behavior was assessed using Child Behavior Checklist (CBCL). Children with GD were assessed during periods of thyroid hormone elevation (active disease) and normalized thyroid hormones (with anti-thyroid drugs or ATDs). Compared to healthy children, patients during periods of thyroid hormone elevation (74.29%) and normalized thyroid hormones (31.43%) had higher frequencies of behavioral abnormalities and scorings of total CBCL scale (P = 0.01; P = 0.04, respectively) and its subscales’ [Anxious/Depressed (P = 0.02; P = 0.04), Withdrawn/Depressed (P = 0.03; P = 0.04) and Somatic Complaints (P = 0.03; P = 0.127) and Social (P = 0.01; P = 0.225), Thought (P = 0.01; P = 0.128) and Attention (P = 0.01; P = 0.01) problems], indicating internalizing and externalizing problems. The majority of patients had at least two different behavioral problems. Marked improvement was found during period of normalized thyroid hormones (P = 0.001). Correlation analyses showed significant associations between total CBCL scoring and age at onset (P = 0.01; P = 0.001) and lower concentrations of thyroid stimulating hormone (TSH) (P = 0.001; P = 0.04) and higher concentrations of free thyroxine (fT4) (P = 0.01; P = 0.02), triiodothyronine (fT3) (P = 0.01; P = 0.03) and thyrotropin receptor antibodies (TRAbs) (P = 0.001; P = 0.01) during periods of thyroid hormone elevation and normalized thyroid hormones, respectively. Multiple linear regression analysis showed that "at presentation" lower concentrations of TSH (P = 0.001; P = 0.03) and higher concentrations of fT4 (P = 0.001, P = 0.01), fT3 (P = 0.01; P = 0.06) and TRAbs (P = 0.001; P = 0.001) were predictors of behavioral problems during periods of active disease and normalized thyroid hormones. We conclude that GD is associated with higher frequencies and severities of anxiety, depression and inattention during periods of thyroid hormone elevation as well as normalized thyroid hormones with ATDs. Therefore, early diagnosis and optimizing management are required to improve children’s social life.
Collapse
Affiliation(s)
- Sherifa Ahmed Hamed
- Department of Neurology and Psychiatry, Assiut University Hospital, Assiut, Egypt
- * E-mail: ,
| | - Fadia Ahmed Attiah
- Department of Neurology and Psychiatry, Assiut University Hospital, Assiut, Egypt
| | | | - Mohamed Fawzy
- Department of Neurology and Psychiatry, Assiut University Hospital, Assiut, Egypt
| |
Collapse
|
13
|
Brauer VS, Zambuzi FA, Espíndola MS, Cavalcanti Neto MP, Prado MKB, Cardoso PM, Soares LS, Galvao-Lima LJ, Leopoldino AM, Cardoso CRDB, Frantz FG. The influence of dehydroepiandrosterone on effector functions of neutrophils. BRAZ J PHARM SCI 2021. [DOI: 10.1590/s2175-97902020000419139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
14
|
Zakharova L, Sharova V, Izvolskaia M. Mechanisms of Reciprocal Regulation of Gonadotropin-Releasing Hormone (GnRH)-Producing and Immune Systems: The Role of GnRH, Cytokines and Their Receptors in Early Ontogenesis in Normal and Pathological Conditions. Int J Mol Sci 2020; 22:ijms22010114. [PMID: 33374337 PMCID: PMC7795970 DOI: 10.3390/ijms22010114] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022] Open
Abstract
Different aspects of the reciprocal regulatory influence on the development of gonadotropin-releasing hormone (GnRH)-producing- and immune systems in the perinatal ontogenesis and their functioning in adults in normal and pathological conditions are discussed. The influence of GnRH on the development of the immune system, on the one hand, and the influence of proinflammatory cytokines on the development of the hypothalamic-pituitary-gonadal system, on the other hand, and their functioning in adult offspring are analyzed. We have focused on the effects of GnRH on the formation and functional activity of the thymus, as the central organ of the immune system, in the perinatal period. The main mechanisms of reciprocal regulation of these systems are discussed. The reproductive health of an individual is programmed by the establishment and development of physiological systems during critical periods. Regulatory epigenetic mechanisms of development are not strictly genetically controlled. These processes are characterized by a high sensitivity to various regulatory factors, which provides possible corrections for disorders.
Collapse
|
15
|
Li N, Xu M, Wu M, Zhao G. Cinnamtannin A2 protects the renal injury by attenuates the altered expression of kidney injury molecule 1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) expression in 5/6 nephrectomized rat model. AMB Express 2020; 10:87. [PMID: 32385622 PMCID: PMC7210374 DOI: 10.1186/s13568-020-01022-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/29/2020] [Indexed: 02/08/2023] Open
Abstract
Present investigation determines the protective effect of Cinnamtannin A2 against chronic renal failure (CRF). 5/6 nephrectomized rat model was used to induced CRF by removing the kidneys and rats were treated with Cinnamtannin A2 10 mg/kg, i.p. for the period 30 days. Nephroprotective effect Cinnamtannin A2 was assessed by estimating the biochemical parameters of renal function test and cytokines in the serum of nephractomized rats. Oxidative stress parameters were estimated in the kidney tissue and western blot assay and qRT-PCR assay was performed to determine the expression of protein in renal tissue of nephractomized rats. Moreover histopathology study was done to observe the tubular injury. Data of the report reveals that treatment with Cinnamtannin A2 ameliorates the altered level of creatinine, blood urea nitrogen (BUN), Neutrophil gelatinase-associated lipocalin (NGAL), Kidney Injury Molecule-1 (KIM-1) and cytokines in the serum and microalbuminurea in the urine of 5/6 nephrectomized rat. Oxidative stress level was reduced in Cinnamtannin A2 treated group than CRF group. Moreover treatment with Cinnamtannin A2 attenuates the altered expression of proteins involved in Nrf2-Keap1 pathway in the kidney tissue of 5/6 nephrectomized rat. Result of histopathology reveals that tubular injury score was reduced in the kidney tissue of Cinnamtannin A2 treated group than CRF group. In conclusion, data of the report suggest that treatment with Cinnamtannin A2 ameliorates the level of KIM1 and NAGL in 5/6 nephractomized rats by regulating Nrf2- Keap1 pathway.
Collapse
Affiliation(s)
- Na Li
- grid.64924.3d0000 0004 1760 5735Department of Nephrology, The third Hospital of Jilin University, No 126 Xiantai Street, Changchun, 130033 Jilin People’s Republic of China
| | - Mingzhu Xu
- grid.64924.3d0000 0004 1760 5735Department of Nephrology, The third Hospital of Jilin University, No 126 Xiantai Street, Changchun, 130033 Jilin People’s Republic of China
| | - Mei Wu
- grid.64924.3d0000 0004 1760 5735Central Laboratory, The third Hospital of Jilin University, Changchun, 130033 Jilin People’s Republic of China
| | - Guanjie Zhao
- grid.64924.3d0000 0004 1760 5735Department of Nephrology, The third Hospital of Jilin University, No 126 Xiantai Street, Changchun, 130033 Jilin People’s Republic of China
| |
Collapse
|
16
|
Pech-Pool S, Berumen LC, Martínez-Moreno CG, García-Alcocer G, Carranza M, Luna M, Arámburo C. Thyrotropin-Releasing Hormone (TRH) and Somatostatin (SST), but not Growth Hormone-Releasing Hormone (GHRH) nor Ghrelin (GHRL), Regulate Expression and Release of Immune Growth Hormone (GH) from Chicken Bursal B-Lymphocyte Cultures. Int J Mol Sci 2020; 21:ijms21041436. [PMID: 32093298 PMCID: PMC7073104 DOI: 10.3390/ijms21041436] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 12/31/2022] Open
Abstract
It is known that growth hormone (GH) is expressed in immune cells, where it exerts immunomodulatory effects. However, the mechanisms of expression and release of GH in the immune system remain unclear. We analyzed the effect of growth hormone-releasing hormone (GHRH), thyrotropin-releasing hormone (TRH), ghrelin (GHRL), and somatostatin (SST) upon GH mRNA expression, intracellular and released GH, Ser133-phosphorylation of CREB (pCREBS133), intracellular Ca2+ levels, as well as B-cell activating factor (BAFF) mRNA expression in bursal B-lymphocytes (BBLs) cell cultures since several GH secretagogues, as well as their corresponding receptors (-R), are expressed in B-lymphocytes of several species. The expression of TRH/TRH-R, ghrelin/GHS-R1a, and SST/SST-Rs (Subtypes 1 to 5) was observed in BBLs by RT-PCR and immunocytochemistry (ICC), whereas GHRH/GHRH-R were absent in these cells. We found that TRH treatment significantly increased local GH mRNA expression and CREB phosphorylation. Conversely, SST decreased GH mRNA expression. Additionally, when added together, SST prevented TRH-induced GH mRNA expression, but no changes were observed in pCREBS133 levels. Furthermore, TRH stimulated GH release to the culture media, while SST increased the intracellular content of this hormone. Interestingly, SST inhibited TRH-induced GH release in a dose-dependent manner. The coaddition of TRH and SST decreased the intracellular content of GH. After 10 min. of incubation with either TRH or SST, the intracellular calcium levels significantly decreased, but they were increased at 60 min. However, the combined treatment with both peptides maintained the Ca2+ levels reduced up to 60-min. of incubation. On the other hand, BAFF cytokine mRNA expression was significantly increased by TRH administration. Altogether, our results suggest that TRH and SST are implicated in the regulation of GH expression and release in BBL cultures, which also involve changes in pCREBS133 and intracellular Ca2+ concentration. It is likely that TRH, SST, and GH exert autocrine/paracrine immunomodulatory actions and participate in the maturation of chicken BBLs.
Collapse
Affiliation(s)
- Santiago Pech-Pool
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (S.P.-P.); (M.C.)
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico; (L.C.B.); (G.G.-A.)
| | - Laura C. Berumen
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico; (L.C.B.); (G.G.-A.)
| | - Carlos G. Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (S.P.-P.); (M.C.)
| | - Guadalupe García-Alcocer
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico; (L.C.B.); (G.G.-A.)
| | - Martha Carranza
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (S.P.-P.); (M.C.)
| | - Maricela Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico; (S.P.-P.); (M.C.)
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro 76230, Mexico
- 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; (S.P.-P.); (M.C.)
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro 76230, Mexico
- 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
|
17
|
Díaz-Galindo MDC, Calderón-Vallejo D, Olvera-Sandoval C, Quintanar JL. Therapeutic approaches of trophic factors in animal models and in patients with spinal cord injury. Growth Factors 2020; 38:1-15. [PMID: 32299267 DOI: 10.1080/08977194.2020.1753724] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Trophic factors are naturally produced by different tissues that participate in several functions such as the intercellular communication, in the development, stability, differentiation and regeneration at the cellular level. Specifically, in the case of spinal injuries, these factors can stimulate neuronal recovery. They are applied both in experimental models and in clinical trials in patients. The trophic factors analysed in this review include gonadotropin-releasing hormone (GnRH), thyrotropin-releasing hormone (TRH), growth hormone (GH), melatonin, oestrogens, the family of fibroblast growth factors (FGFs), the family of neurotrophins and the glial cell-derived neurotrophic factor (GDNF). There are some trophic (neurotrophic) factors that already been tested in patients with spinal cord injury (SCI), but only shown partial recovery effect. It is possible that, the administration of these trophic factors together with physical rehabilitation, act synergistically and, therefore, significantly improve the quality of life of patients with SCI.
Collapse
Affiliation(s)
- María Del Carmen Díaz-Galindo
- Department of Physiology and Pharmacology, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes. Av, Aguascalientes, México
| | - Denisse Calderón-Vallejo
- Department of Physiology and Pharmacology, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes. Av, Aguascalientes, México
- Department of Morphology, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes. Av, Aguascalientes, México
| | - Carlos Olvera-Sandoval
- Facultad de Medicina-Mexicali, Universidad Autónoma de Baja California, México. Dr. Humberto Torres Sanginés S/N. Centro Cívico, Mexicali, México
| | - J Luis Quintanar
- Department of Physiology and Pharmacology, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes. Av, Aguascalientes, México
| |
Collapse
|
18
|
Faraj Tabrizi P, Mohebbi Tafrechi A, Peters I, Atschekzei F, Kuczyk MA, Serth J, Tezval H. Cancer-Specific Loss of Urocortin 3 in Human Renal Cancer. Adv Ther 2020; 37:288-299. [PMID: 31721113 DOI: 10.1007/s12325-019-01141-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The corticotropin-releasing hormone (CRH) system, its receptors corticotropin-releasing hormone receptor 1 (CRHR1) and 2 (CRHR2), and its corresponding binding protein corticotropin-releasing hormone-binding protein (CRHBP) as well as the urocortin proteins-structural homologues to CRH, which are included in this peptide family-have become interesting oncological targets recently. Carcinogenesis of various human tumors has been reported with an altered presence of members of this system. The aim of the present study was to examine the role of urocortin 3 (UCN3) in renal cell carcinoma (RCC). METHODS Therefore, tumoral tissues of 106 patients with RCC and available corresponding normal tissues were analyzed using qPCR for quantitative mRNA expression analysis. Tissue localization and protein signals of UCN3 in normal and tumoral renal specimens were evaluated using western blot and immunohistochemistry. In addition, correlation studies of UCN3 mRNA expression with clinicopathological parameters of patients with RCC and different histological subtypes were evaluated. RESULTS UCN3 mRNA was significantly downregulated in nearly all tumoral tissues (p = 7.92 × 10-13). The same effect was observed at protein level using immunohistochemistry. Level of UCN3 mRNA expression was not directly correlated with clinicopathological parameters. CONCLUSION We report for the first time the significant downregulation of UCN3 in RCC. These results demonstrate a possible involvement of the CRH system and its significance in carcinogenesis of RCC.
Collapse
|
19
|
Gonadotropin-Releasing Hormone in Regulation of Thymic Development in Rats: Profile of Thymic Cytokines. Int J Mol Sci 2019; 20:ijms20164033. [PMID: 31430847 PMCID: PMC6720952 DOI: 10.3390/ijms20164033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/13/2019] [Accepted: 08/17/2019] [Indexed: 01/17/2023] Open
Abstract
An increasing body of recent experimental data confirms the impact of neurohormones on fetal development and function of different body systems. The synthesis of many neurohormones starts in fetal tissues before the hypothalamic–pituitary–adrenal and hypothalamic–pituitary–gonadal systems are formed, and their high levels are detected in the bloodstream. Here, we studied the role of gonadotropin-releasing hormone (GnRH) in rat thymus development and tried to reveal possible mechanisms underlying the GnRH effects in early development. Western blotting and reverse transcription-polymerase chain reaction allowed us to identify receptor for GnRH in the fetal thymus with peak expression on embryonic days 17–18 (ED17–18). Blocking the receptors in utero on ED17 by a GnRH antagonist suppressed the concanavalin A-induced proliferative response of T cells in adults. GnRH (10−7 M) increased mRNA expression of interleukin (IL)-4, IL-10, IL-1β, interferon γ (IFNγ), and tumor necrosis factor α (TNFα) in the thymus of 18-day fetuses after an ex vivo culture for 24 h. The increased mRNA levels of the cytokines in the thymus were accompanied by increased numbers of CD4+ T helpers. Overall, the data obtained confirm the regulatory or morphogenetic effect of GnRH on fetal thymus development mediated by synthesis of thymic cytokines.
Collapse
|
20
|
Liu XH, Khansari AR, Teles M, Martínez-Rodríguez G, Zhang YG, Mancera JM, Reyes-López FE, Tort L. Brain and Pituitary Response to Vaccination in Gilthead Seabream ( Sparus aurata L.). Front Physiol 2019; 10:717. [PMID: 31275156 PMCID: PMC6591443 DOI: 10.3389/fphys.2019.00717] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
Vaccination is a widely used therapeutical strategy in aquaculture, but whether vaccination elicits stress responses in the central neuroendocrine system and enhances the crosstalk between the immune and endocrine systems in the brain or pituitary after vaccination is unclear. To answer this question two experiments using two different vaccine exposure routes, i.e., bath or intraperitoneal (i.p.) injection, were carried out on gilthead seabream (Sparus aurata L.). In the first one, the stress responses of fish subjected to waterborne Vibrio anguillarum bacterin were compared with responses after air exposure or their combination. In the second experiment, fish were subjected to an intraperitoneal injection of Lactococcus garvieae bacterin and we assessed the central stress response and also whether or not a significant immune response was induced in brain and pituitary. In both experiments, blood, brain and pituitary tissues were collected at 1, 6, and 24 h post stress for plasma hormone determination and gene expression analysis, respectively. Results indicated that bath vaccination induced a decreased central stress response compared to air exposure which stimulated both brain and pituitary stress genes. In the second experiment, injection vaccination kept unchanged plasma stress hormones except cortisol that raised at 6 and 24 h. In agreement, non-significant or slight changes on the transcription of stress-related genes were recorded, including the hormone genes of the hypothalamic pituitary interrenal (HPI) axis and other stress markers such as hsp70, hsp90, and mt genes in either brain or pituitary. Significant changes were observed, however, in crhbp and gr. In this second experiment the immune genes il1β, cox2, and lys, showed a strong expression in both brain and pituitary after vaccination, notably il1β which showed more than 10 fold raise. Overall, vaccination procedures, although showing a cortisol response, did not induce other major stress response in brain or pituitary, regardless the administration route. Other than main changes, the alteration of crhbp and gr suggests that these genes could play a relevant role in the feedback regulation of HPI axis after vaccination. In addition, from the results obtained in this work, it is also demonstrated that the immune system maintains a high activity in both brain and pituitary after vaccine injection.
Collapse
Affiliation(s)
- X H Liu
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain.,Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, China
| | - A R Khansari
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - M Teles
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Y G Zhang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, China
| | - J M Mancera
- Department of Biology, Faculty of Marine and Environmental Sciences, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cádiz, Cádiz, Spain
| | - F E Reyes-López
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - L Tort
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Liu X, Khansari AR, Tort L. Fish pituitary show an active immune response after in vitro stimulation with Vibrio bacterin. Gen Comp Endocrinol 2019; 275:65-72. [PMID: 30769010 DOI: 10.1016/j.ygcen.2019.02.011] [Citation(s) in RCA: 4] [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: 10/29/2018] [Revised: 01/25/2019] [Accepted: 02/08/2019] [Indexed: 11/21/2022]
Abstract
The pituitary is a central organ of the neuro-endocrine system in fish that plays critical roles in various physiological processes, including stress response and behavior. Although it is known that pituitary hormones can have a direct or indirect influence stimulating or suppressing the immune responses, whether there is a local immune response in the pituitary or what is the effect of the immune stimulus on the pituitary function in fish is unknown. With the aim to understand the interaction between the immune responses and the endocrine axes at the pituitary level, particularly the Hypothalamus-Pituitary-Interrenal (HPI) axis, pituitaries of rainbow trout (Oncorhynchus mykiss) were cultured in vitro, incubated with bacterin, or bacterin plus CRH, cortisol, human recombinant IL1β, or spleen medium for 3 h, and then genes involved in pro-inflammation (il1β, il8, tnfα1, ifnγ), anti-inflammation (tgfβ1b, il10), immune modulation (mhcIIa, c3, mif) and stress response (crhbp, pomca, pomcb, gr1) were tested. Data showed that, incubation with bacterin alone and bacterin plus recombinant IL1β or CRH, as well as medium from bacterin treated spleen caused significant up-regulation of pro-inflammatory genes il1β and il8, while down-regulated the anti-inflammatory gene tgfβ1b. Besides, recombinant IL1β plus bacterin or alone caused raise of mhcIIa and tnfa, respectively. On the contrary, just a slight or even no alteration was recorded in the expression of stress response genes including crhbp, pomca, pomcb and gr1 in the in vitro cultured trout pituitary following this stimulation. These results suggest a local immune gene equipment in the pituitary of fish, and the potential for fish pituitary to develop both innate and adaptive immune responses, whereas that immune stimulation was not able to evoke a significant endocrine stress response in vitro.
Collapse
Affiliation(s)
- Xiaohong Liu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China; Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, 08193 Barcelona, Spain
| | - Ali R Khansari
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, 08193 Barcelona, Spain
| | - Lluis Tort
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, 08193 Barcelona, Spain.
| |
Collapse
|
22
|
White MR, Graziano MJ, Sanderson TP. Toxicity of Pexacerfont, a Corticotropin-Releasing Factor Type 1 Receptor Antagonist, in Rats and Dogs. Int J Toxicol 2019; 38:110-120. [DOI: 10.1177/1091581819827501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pexacerfont is a corticotropin-releasing factor subtype 1 receptor antagonist that was developed for the treatment of anxiety- and stress-related disorders. This report describes the results of repeat-dose oral toxicity studies in rats (3 and 6 months) and dogs (3 months and 1 year). Pexacerfont was well tolerated in all of these studies at exposures equal to or greater than areas under the curve in humans (clinical dose of 100 mg). Microscopic changes in the liver (hepatocellular hypertrophy), thyroid glands (hypertrophy/hyperplasia and adenomas of follicular cells), and pituitary (hypertrophy/hyperplasia and vacuolation of thyrotrophs) were only observed in rats and were considered adaptive changes in response to hepatic enzyme induction and subsequent alterations in serum thyroid hormone levels. Evidence for hepatic enzyme induction in dogs was limited to increased liver weights and reduced thyroxine (T4) levels. Mammary gland hyperplasia and altered female estrous cycling were only observed in rats, whereas adverse testicular effects (consistent with minimal to moderate degeneration of the germinal epithelium) were only noted following chronic dosing in dogs. The testicular effects were reversible changes with exposure margins of 8× at the no observed adverse effect level. It is not clear whether the changes in mammary gland, estrous cycling, and testes represent secondary hormonal changes due to perturbation of the hypothalamic–pituitary–adrenal axis or are off-target effects. In conclusion, the results of chronic toxicity studies in rats and dogs show that pexacerfont has an acceptable safety profile to support further clinical testing.
Collapse
|
23
|
Fröhlich E, Wahl R. The forgotten effects of thyrotropin-releasing hormone: Metabolic functions and medical applications. Front Neuroendocrinol 2019; 52:29-43. [PMID: 29935915 DOI: 10.1016/j.yfrne.2018.06.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/07/2018] [Accepted: 06/20/2018] [Indexed: 11/18/2022]
Abstract
Thyrotropin-releasing hormone (TRH) causes a variety of thyroidal and non-thyroidal effects, the best known being the feedback regulation of thyroid hormone levels. This was employed in the TRH stimulation test, which is currently little used. The role of TRH as a cancer biomarker is minor, but exaggerated responses to TSH and prolactin levels in breast cancer led to the hypothesis of a potential role for TRH in the pathogenesis of this disease. TRH is a rapidly degraded peptide with multiple targets, limiting its suitability as a biomarker and drug candidate. Although some studies reported efficacy in neural diseases (depression, spinal cord injury, amyotrophic lateral sclerosis, etc.), therapeutic use of TRH is presently restricted to spinocerebellar degenerative disease. Regulation of TRH production in the hypothalamus, patterns of expression of TRH and its receptor in the body, its role in energy metabolism and in prolactin secretion are addressed in this review.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Internal Medicine (Dept. of Endocrinology and Diabetology, Angiology, Nephrology and Clinical Chemistry), University of Tuebingen, Otfried-Muellerstrasse 10, 72076 Tuebingen, Germany; Center for Medical Research, Medical University Graz, Stiftingtalstr. 24, 8010 Graz, Austria
| | - Richard Wahl
- Internal Medicine (Dept. of Endocrinology and Diabetology, Angiology, Nephrology and Clinical Chemistry), University of Tuebingen, Otfried-Muellerstrasse 10, 72076 Tuebingen, Germany.
| |
Collapse
|
24
|
Leistner C, Menke A. How to measure glucocorticoid receptor's sensitivity in patients with stress-related psychiatric disorders. Psychoneuroendocrinology 2018; 91:235-260. [PMID: 29449045 DOI: 10.1016/j.psyneuen.2018.01.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 12/07/2017] [Accepted: 01/28/2018] [Indexed: 12/31/2022]
Abstract
Stress is a state of derailed homeostasis and a main environmental risk factor for psychiatric diseases. Chronic or uncontrollable stress may lead to a dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, which is a common feature of stress-related psychiatric disorders. One of the key mechanisms underlying a disturbed HPA axis is an impaired function of the glucocorticoid receptor (GR) with an enhanced or reduced feedback sensitivity for glucocorticoids and subsequently altered concentrations of peripheral cortisol. GR function is regulated by a multiprotein complex including the different expression of the hsp90 co-chaperone FK 506 binding protein 51 (FKBP5) that may be genetically determined or acquired in response to stressful stimuli. Specific patterns of a dysregulation of the HPA axis and GR function are found in different stress-related psychiatric entities e.g. major depression, job-related exhaustion or posttraumatic stress disorder. GR challenge tests like the dexamethasone-suppression test (DST), the dexamethasone-corticotropin-releasing hormone (dex-CRH) test or most recently the analysis of the dexamethasone-induced gene expression are employed to sensitively measure HPA axis activity in these disorders. They provide information for a stratification of phenotypic similar but neurobiological diverse psychiatric disorders. In this review we present a synopsis of GR challenge tests with a focus on the application of the DST, the CRH test and the dex-CRH test as well as the dexamethasone-induced gene expression in stress-related psychiatric entities.
Collapse
Affiliation(s)
- Carolin Leistner
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Margarete-Hoeppel-Platz 1, Wuerzburg, 97080, Germany
| | - Andreas Menke
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Margarete-Hoeppel-Platz 1, Wuerzburg, 97080, Germany; Comprehensive Heart Failure Center, University Hospital of Wuerzburg, Am Schwarzenberg 15, Wuerzburg, 97080, Germany.
| |
Collapse
|
25
|
Loggins Clay S, Griffin M, Averhart W. Black/White disparities in pregnant women in the United States: An examination of risk factors associated with Black/White racial identity. HEALTH & SOCIAL CARE IN THE COMMUNITY 2018; 26:654-663. [PMID: 29488271 DOI: 10.1111/hsc.12565] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/24/2018] [Indexed: 06/08/2023]
Abstract
This paper explores racial disparities and risk factors of adverse pregnancy outcomes in Black and White pregnant women in the US. The study uses a cross-sectional approach to explore Black and White disparities using data from the 2012 National Survey on Drug Use and Health (NSDUH), which collects interview data from approximately 70,000 randomly selected participants. We included several self-reported conditions that we categorised as individual and social stressors (e.g. measures of institutionalised racism, individual health behaviours, access to quality care and social context factors). We used descriptive statistics, univariate and multivariate analyses in risk factors of adverse pregnancy outcomes between Black and White women. Black women who were pregnant had a lower socioeconomic status and experienced more measures of institutionalised racism compared to White women who were pregnant. More white women who were pregnant were married, had higher levels of educational attainment, higher income levels, and greater employment opportunities. White pregnant women also had higher levels of private health insurance and less dependency on government programmes for access to healthcare. Results from the regressions indicated that Black pregnant women were less likely to be married (OR = 0.01), less likely to have higher income levels (OR = 0.31) and less likely to be employed (OR = 0.52). However, Black pregnant women were more likely to be younger (OR = 1.82). For the health-eroding behaviours, Black pregnant women were less likely to smoke (OR = 0.53) and use alcohol (0.52). After assessing the SES Household-level stressors (access to healthcare), Black pregnant women were more likely to have Medicaid/CHIP (OR = 3.21) and health coverage through government assistant programmes (OR = 3.80); however, less likely to have private health insurance (OR = 0.38). There are differences in risk factors of adverse pregnancy outcomes between White and Black pregnant women based on measures of individual level/social stressors, institutionalised racism, health behaviours and access to care.
Collapse
Affiliation(s)
- Shondra Loggins Clay
- Counseling Center, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Marquianna Griffin
- Chapin Hall Center for Children, Data and Research Technology Liason, Oak Park, IL, USA
| | - Wanda Averhart
- Department of Pediatrics' General Pediatrics Division, University of Florida, Gainesville, FL, USA
| |
Collapse
|
26
|
Fang X, Hong Y, Dai L, Qian Y, Zhu C, Wu B, Li S. CRH promotes human colon cancer cell proliferation via IL-6/JAK2/STAT3 signaling pathway and VEGF-induced tumor angiogenesis. Mol Carcinog 2017; 56:2434-2445. [PMID: 28618089 DOI: 10.1002/mc.22691] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/01/2017] [Accepted: 06/13/2017] [Indexed: 12/14/2022]
Abstract
Corticotrophin-releasing hormone (CRH) has been demonstrated to participate in various diseases. Our previous study showed that its receptor CRHR1 mediated the development of colitis-associated cancer in mouse model. However, the detailed mechanisms remain unclear. In this study, we explored the oncogenetic role of CRH/CRHR1 signaling in colon cancer cells. Cell proliferation and colony formation assays revealed that CRH contributed to cell proliferation. Moreover, tube formation assay showed that CRH-treated colon cancer cell supernatant significantly promoted tube formation of human umbilical vein endothelial cells (HUVECs). And these effects could be reversed by the CRHR1 specific antagonist Antalarmin. Further investigation showed that CRH significantly upregulated the expressions of interlukin-6 (IL-6) and vascular endothelial growth factor (VEGF) through activating nuclear factor-kappa B (NF-κB). The CRH-induced IL-6 promoted phosphorylation of janus kinase 2 (JAK2) and signal transducers and activators of transcription 3 (STAT3). STAT3 inhibition by Stattic significantly inhibited the CRH-induced cell proliferation. In addition, silence of VEGF resulted in declined tube formation induced by CRH. Taken together, CRH/CRHR1 signaling promoted human colon cancer cell proliferation via NF-κB/IL-6/JAK2/STAT3 signaling pathway and tumor angiogenesis via NF-κB/VEGF signaling pathway. Our results provide evidence to support a critical role for the CRH/CRHR1 signaling in colon cancer progression and suggest its potential utility as a new therapeutic target for colon cancer.
Collapse
Affiliation(s)
- Xianjun Fang
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Yali Hong
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Li Dai
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Yuanyuan Qian
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Chao Zhu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Biao Wu
- Department of Surgery, The first affiliated hospital, Nanchang University, Nanchang, China
| | - Shengnan Li
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
27
|
The syndrome of central hypothyroidism and macroorchidism: IGSF1 controls TRHR and FSHB expression by differential modulation of pituitary TGFβ and Activin pathways. Sci Rep 2017; 7:42937. [PMID: 28262687 PMCID: PMC5338029 DOI: 10.1038/srep42937] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 01/18/2017] [Indexed: 12/22/2022] Open
Abstract
IGSF1 (Immunoglobulin Superfamily 1) gene defects cause central hypothyroidism and macroorchidism. However, the pathogenic mechanisms of the disease remain unclear. Based on a patient with a full deletion of IGSF1 clinically followed from neonate to adulthood, we investigated a common pituitary origin for hypothyroidism and macroorchidism, and the role of IGSF1 as regulator of pituitary hormone secretion. The patient showed congenital central hypothyroidism with reduced TSH biopotency, over-secretion of FSH at neonatal minipuberty and macroorchidism from 3 years of age. His markedly elevated inhibin B was unable to inhibit FSH secretion, indicating a status of pituitary inhibin B resistance. We show here that IGSF1 is expressed both in thyrotropes and gonadotropes of the pituitary and in Leydig and germ cells in the testes, but at very low levels in Sertoli cells. Furthermore, IGSF1 stimulates transcription of the thyrotropin-releasing hormone receptor (TRHR) by negative modulation of the TGFβ1-Smad signaling pathway, and enhances the synthesis and biopotency of TSH, the hormone secreted by thyrotropes. By contrast, IGSF1 strongly down-regulates the activin-Smad pathway, leading to reduced expression of FSHB, the hormone secreted by gonadotropes. In conclusion, two relevant molecular mechanisms linked to central hypothyroidism and macroorchidism in IGSF1 deficiency are identified, revealing IGSF1 as an important regulator of TGFβ/Activin pathways in the pituitary.
Collapse
|
28
|
Li T, Wang P, Wang SC, Wang YF. Approaches Mediating Oxytocin Regulation of the Immune System. Front Immunol 2017; 7:693. [PMID: 28119696 PMCID: PMC5223438 DOI: 10.3389/fimmu.2016.00693] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 12/28/2016] [Indexed: 12/02/2022] Open
Abstract
The hypothalamic neuroendocrine system is mainly composed of the neural structures regulating hormone secretion from the pituitary gland and has been considered as the higher regulatory center of the immune system. Recently, the hypothalamo-neurohypophysial system (HNS) emerged as an important component of neuroendocrine–immune network, wherein the oxytocin (OT)-secreting system (OSS) plays an essential role. The OSS, consisting of OT neurons in the supraoptic nucleus, paraventricular nucleus, their several accessory nuclei and associated structures, can integrate neural, endocrine, metabolic, and immune information and plays a pivotal role in the development and functions of the immune system. The OSS can promote the development of thymus and bone marrow, perform immune surveillance, strengthen immune defense, and maintain immune homeostasis. Correspondingly, OT can inhibit inflammation, exert antibiotic-like effect, promote wound healing and regeneration, and suppress stress-associated immune disorders. In this process, the OSS can release OT to act on immune system directly by activating OT receptors or through modulating activities of other hypothalamic–pituitary–immune axes and autonomic nervous system indirectly. However, our understandings of the role of the OSS in neuroendocrine regulation of immune system are largely incomplete, particularly its relationship with other hypothalamic–pituitary–immune axes and the vasopressin-secreting system that coexists with the OSS in the HNS. In addition, it remains unclear about the relationship between the OSS and peripherally produced OT in immune regulation, particularly intrathymic OT that is known to elicit central immunological self-tolerance of T-cells to hypophysial hormones. In this work, we provide a brief review of current knowledge of the features of OSS regulation of the immune system and of potential approaches that mediate OSS coordination of the activities of entire neuroendocrine–immune network.
Collapse
Affiliation(s)
- Tong Li
- School of Basic Medical Sciences, Harbin Medical University , Harbin , China
| | - Ping Wang
- School of Basic Medical Sciences, Harbin Medical University , Harbin , China
| | - Stephani C Wang
- Department of Internal Medicine, Albany Medical Center , Albany, NY , USA
| | - Yu-Feng Wang
- School of Basic Medical Sciences, Harbin Medical University , Harbin , China
| |
Collapse
|
29
|
Segner H, Verburg-van Kemenade BML, Chadzinska M. The immunomodulatory role of the hypothalamus-pituitary-gonad axis: Proximate mechanism for reproduction-immune trade offs? DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:43-60. [PMID: 27404794 DOI: 10.1016/j.dci.2016.07.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/06/2016] [Accepted: 07/07/2016] [Indexed: 06/06/2023]
Abstract
The present review discusses the communication between the hypothalamic-pituitary-gonad (HPG) axis and the immune system of vertebrates, attempting to situate the HPG-immune interaction into the context of life history trade-offs between reproductive and immune functions. More specifically, (i) we review molecular and cellular interactions between hormones of the HPG axis, and, as far as known, the involved mechanisms on immune functions, (ii) we evaluate whether the HPG-immune crosstalk serves as proximate mechanism mediating reproductive-immune trade-offs, and (iii) we ask whether the nature of the HPG-immune interaction is conserved throughout vertebrate evolution, despite the changes in immune functions, reproductive modes, and life histories. In all vertebrate classes studied so far, HPG hormones have immunomodulatory functions, and indications exist that they contribute to reproduction-immunity resource trade-offs, although the very limited information available for most non-mammalian vertebrates makes it difficult to judge how comparable or different the interactions are. There is good evidence that the HPG-immune crosstalk is part of the proximate mechanisms underlying the reproductive-immune trade-offs of vertebrates, but it is only one factor in a complex network of factors and processes. The fact that the HPG-immune interaction is flexible and can adapt to the functional and physiological requirements of specific life histories. Moreover, the assumption of a relatively fixed pattern of HPG influence on immune functions, with, for example, androgens always leading to immunosuppression and estrogens always being immunoprotective, is probably oversimplified, but the HPG-immune interaction can vary depending on the physiological and envoironmental context. Finally, the HPG-immune interaction is not only driven by resource trade-offs, but additional factors such as, for instance, the evolution of viviparity shape this neuroendocrine-immune relationship.
Collapse
Affiliation(s)
- Helmut Segner
- Centre for Fish and Wildlife Health, Dept of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, P.O. Box, CH-3001, Bern, Switzerland.
| | - B M Lidy Verburg-van Kemenade
- Cell Biology and Immunology Group, Dept. of Animal Sciences, Wageningen University, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| |
Collapse
|
30
|
Guzmán-Soto I, Salinas E, Quintanar JL. Leuprolide Acetate Inhibits Spinal Cord Inflammatory Response in Experimental Autoimmune Encephalomyelitis by Suppressing NF-κB Activation. Neuroimmunomodulation 2016; 23:33-40. [PMID: 26445405 DOI: 10.1159/000438927] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/17/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Recent findings have shown that gonadotropin-releasing hormone (GnRH) administration in an animal model of multiple sclerosis (experimental autoimmune encephalomyelitis, EAE) improves clinical signs of locomotion. The present study was designed to determine whether the administration of the synthetic analog of GnRH, leuprolide acetate (LA) - besides its effects on clinical signs of locomotion - also has an effect on the activation/expression levels of molecular markers of EAE, namely transcription nuclear factor (NF)-κB and the proinflammatory cytokines IL-1β, IL-17A, IL-23 and TNF-α. METHODS EAE spinal cords were collected from control and LA-administered rats. Lumbar sections were processed at four different time points during the course of the disease to analyze NF-κB activation by chemiluminescent Western blot, and during the EAE recovery phase to evaluate proinflammatory cytokine levels by quantitative real-time PCR. RESULTS It was found that LA administration to EAE rats promoted a significant reduction of NF-κB activation during the course of the disease and also decreased the mRNA expression levels of the proinflammatory cytokines IL-1β, IL-17A and TNF-α in the EAE recovery phase; both effects are consistent with the decrease in the severity of clinical signs of locomotion induced by the treatment. CONCLUSION LA causes a reduction in the severity of locomotor activity, as well as in the activation of NF-κB and the number of proinflammatory markers in rats with EAE. These results suggest the use of this agonist as a potential therapeutic approach for multiple sclerosis.
Collapse
Affiliation(s)
- Irene Guzmán-Soto
- Laboratory of Neurophysiology, Department of Physiology and Pharmacology, Centro de Ciencias Bx00E1;sicas, Universidad Autx00F3;noma de Aguascalientes, Aguascalientes, Mexico
| | | | | |
Collapse
|
31
|
del Pino J, Moyano-Cires PV, Anadon MJ, Díaz MJ, Lobo M, Capo MA, Frejo MT. Molecular Mechanisms of Amitraz Mammalian Toxicity: A Comprehensive Review of Existing Data. Chem Res Toxicol 2015; 28:1073-94. [DOI: 10.1021/tx500534x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Javier del Pino
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Paula Viviana Moyano-Cires
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Maria Jose Anadon
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - María Jesús Díaz
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Margarita Lobo
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Miguel Andrés Capo
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - María Teresa Frejo
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| |
Collapse
|
32
|
Ndjom CGN, Jones HP. CRH promotes S. pneumoniae growth in vitro and increases lung carriage in mice. Front Microbiol 2015; 6:279. [PMID: 25904910 PMCID: PMC4389549 DOI: 10.3389/fmicb.2015.00279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 03/19/2015] [Indexed: 11/13/2022] Open
Abstract
Streptococcus pneumoniae (S. pneumoniae), a commensal across the nasal passages, is responsible for the majority of infectious pneumonia cases worldwide. Previous studies have shown that hormonal factors may be influential in regulating S. pneumoniae’s transition from a non-pathogen to a pathogenic state. The current study investigated the effects of corticotropin-releasing hormone (CRH), a peptide hormone involved in stress, on the pathogenicity of S. pneumoniae. Mice were infected with CRH-treated S. pneumoniae via intranasal route, showing an increase in pulmonary bacterial burden. We also quantified S. pneumoniae’s response to CRH through limited serial dilutions and growth curve analysis. We demonstrated that CRH promotes S. pneumoniae titer-dependent proliferation, as well as accelerates log-phase growth. Results also showed an increase in pneumococcal-associated virulence protein A virulence gene expression in response to CRH. These results demonstrate a role for CRH in S. pneumoniae pathogenicity, thus implicating CRH in mediating the transition of S. pneumoniae into a pathogenic state.
Collapse
Affiliation(s)
- Colette G Ngo Ndjom
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Fort Worth, TX USA ; Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD USA
| | - Harlan P Jones
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Fort Worth, TX USA
| |
Collapse
|
33
|
Cao BB, Huang Y, Jiang YY, Qiu YH, Peng YP. Cerebellar fastigial nuclear glutamatergic neurons regulate immune function via hypothalamic and sympathetic pathways. J Neuroimmune Pharmacol 2015; 10:162-78. [PMID: 25649846 DOI: 10.1007/s11481-014-9572-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 11/18/2014] [Indexed: 10/24/2022]
Abstract
We previously have shown that cerebellar fastigial nucleus (FN) modulates immune function, but pathways or mechanisms underlying this immunomodulation require clarification. Herein, an anterograde and retrograde tracing of nerve tracts between the cerebellar FN and hypothalamus/thalamus was performed in rats. After demonstrating a direct cerebellar FN-hypothalamic/thalamic glutamatergic projection, 6-diazo-5-oxo-L-norleucine (DON), an inhibitor of glutaminase that catalyzes glutamate synthesis, was injected bilaterally in the cerebellar FN and simultaneously, D,L-threo-β-hydroxyaspartic acid (THA), an inhibitor of glutamate transporters on cell membrane, was bilaterally injected in the lateral hypothalamic area (LHA) or the ventrolateral (VL) thalamic nucleus. DON treatment in the FN alone decreased number of glutamatergic neurons that projected axons to the LHA and also diminished glutamate content in both the hypothalamus and the thalamus. These effects of DON were reduced by combined treatment with THA in the LHA or in the VL. Importantly, DON treatment in the FN alone attenuated percentage and cytotoxicity of natural killer (NK) cells and also lowered percentage and cytokine production of T lymphocytes. These DON-caused immune effects were reduced or abolished by combined treatment with THA in the LHA, but not in the VL. Simultaneously, DON treatment elevated level of norepinephrine (NE) in the spleen and mesenteric lymphoid nodes, and THA treatment in the LHA, rather than in the VL, antagonized the DON-caused NE elevation. These findings suggest that glutamatergic neurons in the cerebellar FN regulate innate and adaptive immune functions and the immunomodulation is conveyed by FN-hypothalamic glutamatergic projections and sympathetic nerves that innervate lymphoid tissues.
Collapse
Affiliation(s)
- Bei-Bei Cao
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | | | | | | | | |
Collapse
|
34
|
Klein JR. Biological Impact of the TSHβ Splice Variant in Health and Disease. Front Immunol 2014; 5:155. [PMID: 24778635 PMCID: PMC3985016 DOI: 10.3389/fimmu.2014.00155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/24/2014] [Indexed: 12/18/2022] Open
Abstract
Thyroid stimulating hormone (TSH), a glycoprotein hormone composed of α and β chains, is produced by thyrotrope cells of the anterior pituitary. Within the conventional endocrine loop, pituitary-derived TSH binds to receptors in the thyroid, resulting in the release of the thyroid hormones thyroxine (T4) and triiodothyronine (T3). T4 and T3 in turn regulate nearly every aspect of mammalian physiology, including basal metabolism, growth and development, and mood and cognition. Although TSHβ has been known for years to be produced by cells of the immune system, the significance of that has remained largely unclear. Recently, a splice variant of TSHβ (TSHβv), which consists of a truncated but biologically functional portion of the native form of TSHβ, was shown to be produced by bone marrow cells and peripheral blood leukocytes, particularly cells of the myeloid/monocyte lineage. In contrast, full-length native TSHβ is minimally produced by cells of the immune system. The present article will describe the discovery of the TSHβv and will discuss its potential role in immunity and autoimmunity, inflammation, and bone remodeling.
Collapse
Affiliation(s)
- John R Klein
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston , Houston, TX , USA
| |
Collapse
|
35
|
Velázquez-Moctezuma J, Domínguez-Salazar E, Gómez-González B. Beyond the borders: the gates and fences of neuroimmune interaction. Front Integr Neurosci 2014; 8:26. [PMID: 24659958 PMCID: PMC3952191 DOI: 10.3389/fnint.2014.00026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 02/25/2014] [Indexed: 12/25/2022] Open
Affiliation(s)
- Javier Velázquez-Moctezuma
- Area of Neurosciences, Biology of Reproduction Department, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa Mexico City, Mexico
| | - Emilio Domínguez-Salazar
- Area of Neurosciences, Biology of Reproduction Department, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa Mexico City, Mexico
| | - Beatriz Gómez-González
- Area of Neurosciences, Biology of Reproduction Department, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa Mexico City, Mexico
| |
Collapse
|