1
|
Sivcev S, Constantin S, Smiljanic K, Sokanovic SJ, Fletcher PA, Sherman AS, Zemkova H, Stojilkovic SS. Distribution and calcium signaling function of somatostatin receptor subtypes in rat pituitary. Cell Calcium 2024; 124:102967. [PMID: 39522307 PMCID: PMC11624061 DOI: 10.1016/j.ceca.2024.102967] [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: 09/25/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The somatostatin (SST) receptor family controls pituitary hormone secretion, but the distribution and specific roles of these receptors on the excitability and voltage-gated calcium signaling of hormone producing pituitary cells have not been fully characterized. Here we show that the rat pituitary gland expressed Sstr1, Sstr2, Sstr3, and Sstr5 receptor genes in a cell type-specific manner: Sstr1 and Sstr2 in thyrotrophs, Sstr3 in gonadotrophs and lactotrophs, Sstr2, Sstr3, and Sstr5 in somatotrophs, and none in corticotrophs and melanotrophs. Most gonadotrophs and thyrotrophs spontaneously fired high-amplitude single action potentials, which were silenced by SST without affecting intracellular calcium concentrations. In contrast, lactotrophs and somatotrophs spontaneously fired low-amplitude plateau-bursting action potentials in conjunction with calcium transients, both of which were silenced by SST. Moreover, SST inhibited GPCR-induced voltage-gated calcium signaling and hormone secretion in all cell types expressing SST receptors, but the inhibition was more pronounced in somatotrophs. The pattern of inhibition of electrical activity and calcium signaling was consistent with both direct and indirect inhibition of voltage-gated calcium channels, the latter being driven by cell type-specific hyperpolarization. These results indicate that the action of SST in somatotrophs is enhanced by the expression of several types of SST receptors and their slow desensitization, that SST may play a role in the electrical resynchronization of gonadotrophs, thyrotrophs, and lactotrophs, and that the lack of SST receptors in corticotrophs and melanotrophs keeps them excitable and ready to responses to stress.
Collapse
Affiliation(s)
- Sonja Sivcev
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda. USA
| | - Stephanie Constantin
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda. USA
| | - Kosara Smiljanic
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda. USA
| | - Srdjan J Sokanovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Patrick A Fletcher
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, USA
| | - Arthur S Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, USA
| | - Hana Zemkova
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda. USA.
| |
Collapse
|
2
|
Lin TL, So EC, Wu SN. Exploring the Effects of Tomatidine ((3β, 5α, 22β, and 25β)-Spirosolan-3-ol) on Voltage-gated Na+ currents: Insights Into Its Ionic Mechanisms of Action on Current Magnitude, Gating, and Frequency Dependence. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:298-311. [PMID: 39641137 DOI: 10.4103/ejpi.ejpi-d-24-00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024]
Abstract
ABSTRACT Tomatidine, a major tomato glycoalkaloid, is effective for the prevention of skeletal muscle wasting and enhancing mitophagy. However, its effects on transmembrane ionic currents are not well explored. In this study, we explored the interactions between tomatidine and Na+ current. GH3 or Neuro-2a cells were used for recording the ion currents employing modified patch-clamp technique under whole-cell configuration. Tomatidine increased both the peak, (transient Na+ current [INa (T)]) and sustained (late Na+ current [INa (L)]) components of voltage-gated Na+ current (INa) in a concentration-dependent manner, with the concentration required for 50% stimulation values of 43.3 μM and 3.1 μM, respectively. The steady-state current-voltage relationship of INa (T) remained unchanged; however, the steady-state inactivation curve of INa (T) in the presence of 3 μM tomatidine was shifted to less depolarized potential by around 6 mV. Tomatidine enhanced the window INa (window Na+ current [INa (W)]), which were attenuated by the ranolazine (Ran) and carbamazepine (CBZ). During a train of depolarizing pulses, tomatidine slowed the exponential decay of INa (T), and this effect was reversed by Ran or dapagliflozin. Tomatidine increased both fast and slow recovery time constants from INa (T) block, affecting the recovery time course. Tomatidine increased the amplitude of persistent Na+ current in response to a sinusoidal waveform. In neuro-2a cells, tomatidine increased INa (T) amplitude and slowed its inactivation, with this effect being attenuated by Ran or CBZ. In conclusion, tomatidine enhanced magnitude and modified its gating behaviors.
Collapse
Affiliation(s)
- Tso-Lin Lin
- Department of Paediatrics, An Nan Hospital, China Medical University, Tainan, Taiwan
| | - Edmund Cheung So
- Department of Anaesthesia, An Nan Hospital, China Medical University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, National Cheng Kung University Medical College, Tainan, Taiwan
- Department of Anaesthesiology, University of Hong Kong, Hong Kong
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan, Taiwan
- Department of Medical Education and Research, An Nan Hospital, China Medical University, Tainan, Taiwan
| |
Collapse
|
3
|
Li W, Ye C, He M, Ko WKW, Cheng CHK, Chan YW, Wong AOL. Differential involvement of cAMP/PKA-, PLC/PKC- and Ca 2+/calmodulin-dependent pathways in GnRH-induced prolactin secretion and gene expression in grass carp pituitary cells. Front Endocrinol (Lausanne) 2024; 15:1399274. [PMID: 38894746 PMCID: PMC11183098 DOI: 10.3389/fendo.2024.1399274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is a key stimulator for gonadotropin secretion in the pituitary and its pivotal role in reproduction is well conserved in vertebrates. In fish models, GnRH can also induce prolactin (PRL) release, but little is known for the corresponding effect on PRL gene expression as well as the post-receptor signalling involved. Using grass carp as a model, the functional role of GnRH and its underlying signal transduction for PRL regulation were examined at the pituitary level. Using laser capture microdissection coupled with RT-PCR, GnRH receptor expression could be located in carp lactotrophs. In primary cell culture prepared from grass carp pituitaries, the native forms of GnRH, GnRH2 and GnRH3, as well as the GnRH agonist [D-Arg6, Pro9, NEt]-sGnRH were all effective in elevating PRL secretion, PRL mRNA level, PRL cell content and total production. In pituitary cells prepared from the rostral pars distalis, the region in the carp pituitary enriched with lactotrophs, GnRH not only increased cAMP synthesis with parallel CREB phosphorylation and nuclear translocation but also induced a rapid rise in cytosolic Ca2+ by Ca2+ influx via L-type voltage-sensitive Ca2+ channel (VSCC) with subsequent CaM expression and NFAT2 dephosphorylation. In carp pituitary cells prepared from whole pituitaries, GnRH-induced PRL secretion was reduced/negated by inhibiting cAMP/PKA, PLC/PKC and Ca2+/CaM/CaMK-II pathways but not the signalling events via IP3 and CaN/NFAT. The corresponding effect on PRL mRNA expression, however, was blocked by inhibiting cAMP/PKA/CREB/CBP and Ca2+/CaM/CaN/NFAT2 signalling but not PLC/IP3/PKC pathway. At the pituitary cell level, activation of cAMP/PKA pathway could also induce CaM expression and Ca2+ influx via VSCC with parallel rises in PRL release and gene expression in a Ca2+/CaM-dependent manner. These findings, as a whole, suggest that the cAMP/PKA-, PLC/PKC- and Ca2+/CaM-dependent cascades are differentially involved in GnRH-induced PRL secretion and PRL transcript expression in carp lactotrophs. During the process, a functional crosstalk between the cAMP/PKA- and Ca2+/CaM-dependent pathways may occur with PRL release linked with CaMK-II and PKC activation and PRL gene transcription caused by nuclear action of CREB/CBP and CaN/NFAT2 signalling.
Collapse
Affiliation(s)
- Wensheng Li
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Cheng Ye
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Mulan He
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wendy K. W. Ko
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Christopher H. K. Cheng
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ying Wai Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Anderson O. L. Wong
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
Wang L, Liu X, Zhao M, Li F, Liu J. Disruption of gonadotropin hormone biosynthesis by parabens: A potential development and reproduction-associated adverse outcome pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 347:123716. [PMID: 38458526 DOI: 10.1016/j.envpol.2024.123716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/24/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Parabens are widely used as antibacterial preservatives in foods and personal care products. The knowledge about the modes of toxic action of parabens on development and reproduction remain very limited. The present study attempted to establish a development and reproduction-associated adverse outcome pathway (AOP) by evaluating the effects of methylparaben (MP), ethylparaben (EP), propylparaben (PP) and butylparaben (BP) on the biosynthesis of gonadotropins, which are key hormones for development and reproduction. MP and BP significantly upregulated the mRNA and protein levels of follicle stimulating hormone (FSH) and luteinizing hormone (LH) in pituitary gonadotropic cells in a concentration-dependent manner. Activation of gonadotropin-releasing hormone receptor (GnRHR) was required for gonadotropin biosynthesis induced by BP, but not MP. Molecular docking data further demonstrated the higher binding efficiency of BP to human GnRHR than that of MP, suggesting GnRHR as a potential molecular initiative event (MIE) for BP-induced gonadotropin production. L-type voltage-gated calcium channels (VGCCs) were found to be another candidate for MIE in gonadotropic cells response to both MP and BP exposure. The calcium-dependent activation of extracellular signal-regulated kinase 1 (ERK1) and ERK2 was subsequently required for MP- and BP-induced activation of GnRHR and L-type VGCCs pathways. In summary, MP and BP promoted gonadotropin biosynthesis through their interactions with cellular macromolecules GnRHR, L-type VGCCs, and subsequent key event ERK1/2. This is the first study to report the direct interference of parabens with gonadotropin biosynthesis and establish a potential AOP based on pathway-specific mechanism, which contributes to the effective screening of environmental chemicals with developmental and reproductive health risks.
Collapse
Affiliation(s)
- Linping Wang
- MOE Key Lab of Environmental Remediation and Ecosystem Health, Research Center for Air Pollution and Health, Institute of Environmental Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaochen Liu
- MOE Key Lab of Environmental Remediation and Ecosystem Health, Research Center for Air Pollution and Health, Institute of Environmental Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Manqi Zhao
- MOE Key Lab of Environmental Remediation and Ecosystem Health, Research Center for Air Pollution and Health, Institute of Environmental Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feixue Li
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China
| | - Jing Liu
- MOE Key Lab of Environmental Remediation and Ecosystem Health, Research Center for Air Pollution and Health, Institute of Environmental Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
5
|
Santiago-Andres Y, Aquiles A, Taniguchi-Ponciano K, Salame L, Guinto G, Mercado M, Fiordelisio T. Association between Intracellular Calcium Signaling and Tumor Recurrence in Human Non-Functioning Pituitary Adenomas. Int J Mol Sci 2024; 25:3968. [PMID: 38612778 PMCID: PMC11011867 DOI: 10.3390/ijms25073968] [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: 02/24/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Clinically non-functioning pituitary adenomas (CNFPAs) are the second most frequent sellar tumor among studies on community-dwelling adults. They are characterized by the absence of hormonal hypersecretion syndrome, and patients present with compressive symptoms, such as a headache and visual field defects. Immunohistochemically, most CNFPAs are of gonadotrope differentiation, with only a few of them being truly null cell adenomas. Although these tumors express receptors for one or more hypothalamic releasing hormones, to what extent this has an impact on the biological and clinical behavior of these neoplasms remains to be defined. In this research, we evaluated the basal and hypothalamic secretagogue-stimulated intracellular calcium mobilization in 13 CNFPAs, trying to correlate this response to the phenotypic features of the patients. Our results indicate that the recurrence of a CNFPA correlates positively with cellular responsiveness, as measured by spontaneous intracellular calcium activity and the ability to respond to multiple hypothalamic secretagogues. We conclude that this finding may be a useful tool for predicting the clinicopathologic behavior of CNFPAs, by testing the variation of cellular responsiveness to hypothalamic secretagogues.
Collapse
Affiliation(s)
- Yorgui Santiago-Andres
- Laboratorio de Neuroendocrinología Comparada, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de Mexico 04510, Mexico;
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Ciudad de Mexico 04510, Mexico
| | - Ana Aquiles
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro 76230, Mexico;
| | - Keiko Taniguchi-Ponciano
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico 06720, Mexico; (K.T.-P.); (L.S.)
| | - Latife Salame
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico 06720, Mexico; (K.T.-P.); (L.S.)
| | - Gerardo Guinto
- Centro Neurológico, Centro Médico ABC, Ciudad de México 05370, Mexico;
| | - Moises Mercado
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico 06720, Mexico; (K.T.-P.); (L.S.)
| | - Tatiana Fiordelisio
- Laboratorio de Neuroendocrinología Comparada, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de Mexico 04510, Mexico;
| |
Collapse
|
6
|
Axiak CJ, Pleven A, Attard R, Borg Carbott F, Ebejer JP, Brincat I, Cassar K, Gruppetta M, Vassallo J, Bezzina Wettinger S, Farrugia R. High Population Frequency of GNRHR p.Q106R in Malta: An Evaluation of Fertility and Hormone Profiles in Heterozygotes. J Endocr Soc 2024; 8:bvad172. [PMID: 38196663 PMCID: PMC10775685 DOI: 10.1210/jendso/bvad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Indexed: 01/11/2024] Open
Abstract
Context The gonadotropin-releasing hormone receptor variant GNRHR p.Q106R (rs104893836) in homozygosity, compound heterozygosity, or single heterozygosity is often reported as the causative variant in idiopathic hypogonadotropic hypogonadism (IHH) patients with GnRH deficiency. Genotyping of a Maltese newborn cord-blood collection yielded a minor allele frequency (MAF) 10 times higher (MAF = 0.029; n = 493) than that of the global population (MAF = 0.003). Objective To determine whether GNRHR p.Q106R in heterozygosity influences profiles of endogenous hormones belonging to the hypothalamic-pituitary axis and the onset of puberty and fertility in adult men (n = 739) and women (n = 239). Design Setting and Participants Analysis of questionnaire data relating to puberty and fertility, genotyping of the GNRHR p.Q106R variant, and hormone profiling of a highly phenotyped Maltese adult cohort from the Maltese Acute Myocardial Infarction Study. Main Outcome and Results Out of 978 adults, 43 GNRHR p.Q106R heterozygotes (26 men and 17 women) were identified. Hormone levels and fertility for all heterozygotes are within normal parameters except for TSH, which was lower in men 50 years or older. Conclusion Hormone data and baseline fertility characteristics of GNRHR p.Q106R heterozygotes are comparable to those of homozygous wild-type individuals who have no reproductive problems. The heterozygous genotype alone does not impair the levels of investigated gonadotropins and sex steroid hormones or affect fertility. GNRHR p.Q106R heterozygotes who exhibit IHH characteristics must have at least another variant, probably in a different IHH gene, that drives pathogenicity. We also conclude that GNRHR p.Q106R is likely a founder variant due to its overrepresentation and prevalence in the island population of Malta.
Collapse
Affiliation(s)
- Clayton John Axiak
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
| | - Adrian Pleven
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
- Clinical Chemistry Section, Department of Pathology, Mater Dei Hospital, Msida, MSD 2080, Malta
| | - Ritienne Attard
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
| | - Francesca Borg Carbott
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
| | - Jean-Paul Ebejer
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, MSD 2080, Malta
| | - Ian Brincat
- Clinical Chemistry Section, Department of Pathology, Mater Dei Hospital, Msida, MSD 2080, Malta
| | - Karen Cassar
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta
| | - Mark Gruppetta
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta
- Division of Endocrinology and Diabetes, Department of Medicine, Mater Dei Hospital, Msida, MSD 2080, Malta
| | - Josanne Vassallo
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, MSD 2080, Malta
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta
- Division of Endocrinology and Diabetes, Department of Medicine, Mater Dei Hospital, Msida, MSD 2080, Malta
| | - Stephanie Bezzina Wettinger
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, MSD 2080, Malta
| | - Rosienne Farrugia
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, MSD 2080, Malta
| |
Collapse
|
7
|
Constantin S, Sokanovic SJ, Mochimaru Y, Smiljanic K, Sivcev S, Prévide RM, Wray S, Balla T, Stojilkovic SS. Postnatal Development and Maintenance of Functional Pituitary Gonadotrophs Is Dependent on PI4-Kinase A. Endocrinology 2023; 164:bqad168. [PMID: 37935042 PMCID: PMC10652335 DOI: 10.1210/endocr/bqad168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
Postnatal development of functional pituitary gonadotrophs is necessary for maturation of the hypothalamic-pituitary-gonadal axis, puberty, and reproduction. Here we examined the role of PI4-kinase A, which catalyzes the biosynthesis of PI4P in mouse reproduction by knocking out this enzyme in cells expressing the gonadotropin-releasing hormone (GnRH) receptor. Knockout (KO) mice were infertile, reflecting underdeveloped gonads and reproductive tracts and lack of puberty. The number and distribution of hypothalamic GnRH neurons and Gnrh1 expression in postnatal KOs were not affected, whereas Kiss1/kisspeptin expression was increased. KO of PI4-kinase A also did not alter embryonic establishment and neonatal development and function of the gonadotroph population. However, during the postnatal period, there was a progressive loss of expression of gonadotroph-specific genes, including Fshb, Lhb, and Gnrhr, accompanied by low gonadotropin synthesis. The postnatal gonadotroph population also progressively declined, reaching approximately one-third of that observed in controls at 3 months of age. In these residual gonadotrophs, GnRH-dependent calcium signaling and calcium-dependent membrane potential changes were lost, but intracellular administration of inositol-14,5-trisphosphate rescued this signaling. These results indicate a key role for PI4-kinase A in the postnatal development and maintenance of a functional gonadotroph population.
Collapse
Affiliation(s)
- Stephanie Constantin
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Srdjan J Sokanovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yuta Mochimaru
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kosara Smiljanic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sonja Sivcev
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rafael M Prévide
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tamas Balla
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Inhibition of Voltage-Gated Na + Currents Exerted by KB-R7943 (2-[2-[4-(4-nitrobenzyloxy)phenyl]ethyl]isothiourea), an Inhibitor of Na +-Ca 2+ Exchanging Process. Int J Mol Sci 2023; 24:ijms24021805. [PMID: 36675319 PMCID: PMC9864174 DOI: 10.3390/ijms24021805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
KB-R7943, an isothiourea derivative, has been recognized as an inhibitor in the reverse mode of the Na+-Ca2+ exchanging process. This compound was demonstrated to prevent intracellular Na+-dependent Ca2+ uptake in intact cells; however, it is much less effective at preventing extracellular Na+-dependent Ca2+ efflux. Therefore, whether or how this compound may produce any perturbations on other types of ionic currents, particularly on voltage-gated Na+ current (INa), needs to be further studied. In this study, the whole-cell current recordings demonstrated that upon abrupt depolarization in pituitary GH3 cells, the exposure to KB-R7943 concentration-dependently depressed the transient (INa(T)) or late component (INa(L)) of INa with an IC50 value of 11 or 0.9 μM, respectively. Likewise, the dissociation constant for the KB-R7943-mediated block of INa on the basis of a minimum reaction scheme was estimated to be 0.97 μM. The presence of benzamil or amiloride could suppress the INa(L) magnitude. The instantaneous window Na+ current (INa(W)) activated by abrupt ascending ramp voltage (Vramp) was suppressed by adding KB-R7943; however, subsequent addition of deltamethrin or tefluthrin (Tef) effectively reversed KB-R7943-inhibted INa(W). With prolonged duration of depolarizing pulses, the INa(L) amplitude became exponentially decreased; moreover, KB-R7943 diminished INa(L) magnitude. The resurgent Na+ current (INa(R)) evoked by a repolarizing Vramp was also suppressed by adding this compound; moreover, subsequent addition of ranolazine or Tef further diminished or reversed, respectively, its reduction in INa(R) magnitude. The persistent Na+ current (INa(P)) activated by sinusoidal voltage waveform became enhanced by Tef; however, subsequent application of KB-R7943 counteracted Tef-stimulated INa(P). The docking prediction reflected that there seem to be molecular interactions of this molecule with the hNaV1.2 or hNaV1.7 channels. Collectively, this study highlights evidence showing that KB-R7943 has the propensity to perturb the magnitude and gating kinetics of INa (e.g., INa(T), INa(L), INa(W), INa(R), and INa(P)) and that the NaV channels appear to be important targets for the in vivo actions of KB-R7943 or other relevant compounds.
Collapse
|
9
|
Stojilkovic SS, Balla T. PI(4,5)P2-dependent and -independent roles of PI4P in the control of hormone secretion by pituitary cells. Front Endocrinol (Lausanne) 2023; 14:1118744. [PMID: 36777340 PMCID: PMC9911653 DOI: 10.3389/fendo.2023.1118744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 01/28/2023] Open
Abstract
Plasma membrane and organelle membranes are home to seven phosphoinositides, an important class of low-abundance anionic signaling lipids that contribute to cellular functions by recruiting cytoplasmic proteins or interacting with the cytoplasmic domains of membrane proteins. Here, we briefly review the functions of three phosphoinositides, PI4P, PI(4,5)P2, and PI(3,4,5)P3, in cellular signaling and exocytosis, focusing on hormone-producing pituitary cells. PI(4,5)P2, acting as a substrate for phospholipase C, plays a key role in the control of pituitary cell functions, including hormone synthesis and secretion. PI(4,5)P2 also acts as a substrate for class I PI3-kinases, leading to the generation of two intracellular messengers, PI(3,4,5)P3 and PI(3,4)P2, which act through their intracellular effectors, including Akt. PI(4,5)P2 can also influence the release of pituitary hormones acting as an intact lipid to regulate ion channel gating and concomitant calcium signaling, as well as the exocytic pathway. Recent findings also show that PI4P is not only a precursor of PI(4,5)P2, but also a key signaling molecule in many cell types, including pituitary cells, where it controls hormone secretion in a PI(4,5)P2-independent manner.
Collapse
Affiliation(s)
- Stanko S. Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Stanko S. Stojilkovic,
| | - Tamas Balla
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
10
|
Lin MH, Lin JF, Yu MC, Wu SN, Wu CL, Cho HY. Characterization in Potent Modulation on Voltage-Gated Na + Current Exerted by Deltamethrin, a Pyrethroid Insecticide. Int J Mol Sci 2022; 23:ijms232314733. [PMID: 36499059 PMCID: PMC9737322 DOI: 10.3390/ijms232314733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Deltamethrin (DLT) is a type-II pyrethroid ester insecticide used in agricultural and domestic applications as well as in public health. However, transmembrane ionic channels perturbed by this compound remain largely unclear, although the agent is thought to alter the gating characteristics of voltage-gated Na+ (NaV) channel current. In this study, we reappraised whether and how it and other related compounds can make any further modifications on voltage-gated Na+ current (INa) in pituitary tumor (GH3) cells. Cell exposure to DLT produced a differential and dose-dependent stimulation of peak (transient, INa(T)) or sustained (late, INa(L)) INa; consequently, the EC50 value required for DLT-stimulated INa(T) or INa(L) was determined to be 11.2 or 2.5 μM, respectively. However, neither the fast nor slow component in the inactivation time constant of INa(T) activated by short depolarizing pulse was changed with the DLT presence; conversely, tefluthrin (Tef), a type-I pyrethroid insecticide, can accentuate INa with a slowing in inactivation time course of the current. The INa(L) augmented by DLT was attenuated by further application of either dapagliflozin (Dapa) or amiloride, but not by chlorotoxin. During pulse train (PT) stimulation, with the Tef or DLT presence, the cumulative inhibition of INa(T) became slowed; moreover, following PT stimuli, a large tail current with a slowly recovering process was observed. Alternatively, during rapid depolarizing pulse, the amplitude of INa(L) and tail INa (INa(Tail)) for each depolarizing pulse became progressively increased by adding DLT, not by Tef. The recovery time constant following PT stimulation with continued presence of Tef or DLT was shortened by further addition of Dapa. The voltage-dependent hysteresis (Hys(V)) of persistent INa was differentially augmented by Tef or DLT. Taken together, the magnitude, gating, frequency dependence, as well as Hys(V) behavior of INa exerted by the presence of DLT or Tef might exert a synergistic impact on varying functional activities of excitable cells in culture or in vivo.
Collapse
Affiliation(s)
- Mao-Hsun Lin
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 600, Taiwan
| | - Jen-Feng Lin
- Department of Emergency Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 600, Taiwan
| | - Meng-Cheng Yu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 701, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 701, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 701, Taiwan
- Department of Post-Baccalaureate Medicine, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Correspondence: ; Tel.: +886-6-2353535-5334; Fax: 886-6-2362780
| | - Chao-Liang Wu
- Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 600, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan 701, Taiwan
| |
Collapse
|
11
|
Hou H, Chan C, Yuki KE, Sokolowski D, Roy A, Qu R, Uusküla-Reimand L, Faykoo-Martinez M, Hudson M, Corre C, Goldenberg A, Zhang Z, Palmert MR, Wilson MD. Postnatal developmental trajectory of sex-biased gene expression in the mouse pituitary gland. Biol Sex Differ 2022; 13:57. [PMID: 36221127 PMCID: PMC9552479 DOI: 10.1186/s13293-022-00467-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The pituitary gland regulates essential physiological processes such as growth, pubertal onset, stress response, metabolism, reproduction, and lactation. While sex biases in these functions and hormone production have been described, the underlying identity, temporal deployment, and cell-type specificity of sex-biased pituitary gene regulatory networks are not fully understood. METHODS To capture sex differences in pituitary gene regulation dynamics during postnatal development, we performed 3' untranslated region sequencing and small RNA sequencing to ascertain gene and microRNA expression, respectively, across five postnatal ages (postnatal days 12, 22, 27, 32, 37) that span the pubertal transition in female and male C57BL/6J mouse pituitaries (n = 5-6 biological replicates for each sex at each age). RESULTS We observed over 900 instances of sex-biased gene expression and 17 sex-biased microRNAs, with the majority of sex differences occurring with puberty. Using miRNA-gene target interaction databases, we identified 18 sex-biased genes that were putative targets of 5 sex-biased microRNAs. In addition, by combining our bulk RNA-seq with publicly available male and female mouse pituitary single-nuclei RNA-seq data, we obtained evidence that cell-type proportion sex differences exist prior to puberty and persist post-puberty for three major hormone-producing cell types: somatotropes, lactotropes, and gonadotropes. Finally, we identified sex-biased genes in these three pituitary cell types after accounting for cell-type proportion differences between sexes. CONCLUSION Our study reveals the identity and postnatal developmental trajectory of sex-biased gene expression in the mouse pituitary. This work also highlights the importance of considering sex biases in cell-type composition when understanding sex differences in the processes regulated by the pituitary gland.
Collapse
Affiliation(s)
- Huayun Hou
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Cadia Chan
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada
| | - Kyoko E Yuki
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Dustin Sokolowski
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Anna Roy
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Rihao Qu
- Interdepartmental Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.,Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | | | - Mariela Faykoo-Martinez
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Matt Hudson
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Christina Corre
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada.,Division of Endocrinology, The Hospital for Sick Children, Toronto, ON, Canada.,Departments of Pediatrics and Physiology, University of Toronto, Toronto, ON, Canada
| | - Anna Goldenberg
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada.,Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Zhaolei Zhang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Mark R Palmert
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada. .,Division of Endocrinology, The Hospital for Sick Children, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Departments of Pediatrics and Physiology, University of Toronto, Toronto, ON, Canada.
| | - Michael D Wilson
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
12
|
Shiau AL, Liao CS, Tu CW, Wu SN, Cho HY, Yu MC. Characterization in Effective Stimulation on the Magnitude, Gating, Frequency Dependence, and Hysteresis of INa Exerted by Picaridin (or Icaridin), a Known Insect Repellent. Int J Mol Sci 2022; 23:ijms23179696. [PMID: 36077093 PMCID: PMC9456182 DOI: 10.3390/ijms23179696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Picaridin (icaridin), a member of the piperidine chemical family, is a broad-spectrum arthropod repellent. Its actions have been largely thought to be due to its interaction with odorant receptor proteins. However, to our knowledge, to what extent the presence of picaridin can modify the magnitude, gating, and/or the strength of voltage-dependent hysteresis (Hys(V)) of plasmalemmal ionic currents, such as, voltage-gated Na+ current [INa], has not been entirely explored. In GH3 pituitary tumor cells, we demonstrated that with exposure to picaridin the transient (INa(T)) and late (INa(L)) components of voltage-gated Na+ current (INa) were differentially stimulated with effective EC50’s of 32.7 and 2.8 μM, respectively. Upon cell exposure to it, the steady-state current versus voltage relationship INa(T) was shifted to more hyperpolarized potentials. Moreover, its presence caused a rightward shift in the midpoint for the steady-state inactivate curve of the current. The cumulative inhibition of INa(T) induced during repetitive stimuli became retarded during its exposure. The recovery time course from the INa block elicited, following the conditioning pulse stimulation, was satisfactorily fitted by two exponential processes. Moreover, the fast and slow time constants of recovery from the INa block by the same conditioning protocol were noticeably increased in the presence of picaridin. However, the fraction in fast or slow component of recovery time course was, respectively, increased or decreased with an increase in picaridin concentrations. The Hys(V)’s strength of persistent INa (INa(P)), responding to triangular ramp voltage, was also enhanced during cell exposure to picaridin. The magnitude of resurgent INa (INa(R)) was raised in its presence. Picaritin-induced increases of INa(P) or INa(R) intrinsically in GH3 cells could be attenuated by further addition of ranolazine. The predictions of molecular docking also disclosed that there are possible interactions of the picaridin molecule with the hNaV1.7 channel. Taken literally, the stimulation of INa exerted by the exposure to picaridin is expected to exert impacts on the functional activities residing in electrically excitable cells.
Collapse
Affiliation(s)
- Ai-Li Shiau
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Chih-Szu Liao
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Chi-Wen Tu
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan
- Correspondence: ; Tel.: +886-6-2353535-5334; Fax: +886-6-2362780
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Meng-Cheng Yu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| |
Collapse
|
13
|
Pharmacological Dissection of the Crosstalk between Na V and Ca V Channels in GH3b6 Cells. Int J Mol Sci 2022; 23:ijms23020827. [PMID: 35055012 PMCID: PMC8775721 DOI: 10.3390/ijms23020827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 02/01/2023] Open
Abstract
Thanks to the crosstalk between Na+ and Ca2+ channels, Na+ and Ca2+ homeostasis interplay in so-called excitable cells enables the generation of action potential in response to electrical stimulation. Here, we investigated the impact of persistent activation of voltage-gated Na+ (NaV) channels by neurotoxins, such as veratridine (VTD), on intracellular Ca2+ concentration ([Ca2+]i) in a model of excitable cells, the rat pituitary GH3b6 cells, in order to identify the molecular actors involved in Na+-Ca2+ homeostasis crosstalk. By combining RT-qPCR, immunoblotting, immunocytochemistry, and patch-clamp techniques, we showed that GH3b6 cells predominantly express the NaV1.3 channel subtype, which likely endorses their voltage-activated Na+ currents. Notably, these Na+ currents were blocked by ICA-121431 and activated by the β-scorpion toxin Tf2, two selective NaV1.3 channel ligands. Using Fura-2, we showed that VTD induced a [Ca2+]i increase. This effect was suppressed by the selective NaV channel blocker tetrodotoxin, as well by the selective L-type CaV channel (LTCC) blocker nifedipine. We also evidenced that crobenetine, a NaV channel blocker, abolished VTD-induced [Ca2+]i elevation, while it had no effects on LTCC. Altogether, our findings highlight a crosstalk between NaV and LTCC in GH3b6 cells, providing a new insight into the mode of action of neurotoxins.
Collapse
|
14
|
Li X, Zhou L, Peng G, Liao M, Zhang L, Hu H, Long L, Tang X, Qu H, Shao J, Zheng H, Long M. Pituitary P62 deficiency leads to female infertility by impairing luteinizing hormone production. Exp Mol Med 2021; 53:1238-1249. [PMID: 34453106 PMCID: PMC8417229 DOI: 10.1038/s12276-021-00661-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
P62 is a protein adaptor for various metabolic processes. Mice that lack p62 develop adult-onset obesity. However, investigations on p62 in reproductive dysfunction are rare. In the present study, we explored the effect of p62 on the reproductive system. P62 deficiency-induced reproductive dysfunction occurred at a young age (8 week old). Young systemic p62 knockout (p62-/-) and pituitary-specific p62 knockout (p62flox/flox αGSUcre) mice both presented a normal metabolic state, whereas they displayed infertility phenotypes (attenuated breeding success rates, impaired folliculogenesis and ovulation, etc.) with decreased luteinizing hormone (LH) expression and production. Consistently, in an infertility model of polycystic ovary syndrome (PCOS), pituitary p62 mRNA was positively correlated with LH levels. Mechanistically, p62-/- pituitary RNA sequencing showed a significant downregulation of the mitochondrial oxidative phosphorylation (OXPHOS) pathway. In vitro experiments using the pituitary gonadotroph cell line LβT2 and siRNA/shRNA/plasmid confirmed that p62 modulated LH synthesis and secretion via mitochondrial OXPHOS function, especially Ndufa2, a component molecule of mitochondrial complex I, as verified by Seahorse and rescue tests. After screening OXPHOS markers, Ndufa2 was found to positively regulate LH production in LβT2 cells. Furthermore, the gonadotropin-releasing hormone (GnRH)-stimulating test in p62flox/flox αGSUcre mice and LβT2 cells illustrated that p62 is a modulator of the GnRH-LH axis, which is dependent on intracellular calcium and ATP. These findings demonstrated that p62 deficiency in the pituitary impaired LH production via mitochondrial OXPHOS signaling and led to female infertility, thus providing the GnRH-p62-OXPHOS(Ndufa2)-Ca2+/ATP-LH pathway in gonadotropic cells as a new theoretical basis for investigating female reproductive dysfunction.
Collapse
Affiliation(s)
- Xing Li
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China
- Department of Endocrinology, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Street No. 305, Xuanwu, Nanjing, China
- Department of Endocrinology, Jinling Hospital, Nanjing Medical University, Zhongshan East Street No. 305, Xuanwu, Nanjing, China
| | - Ling Zhou
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China
| | - Guiliang Peng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China
| | - Mingyu Liao
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China
| | - Linlin Zhang
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China
| | - Hua Hu
- Department of Gynaecology and Obstetrics, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China
| | - Ling Long
- Department of Gynaecology and Obstetrics, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China
| | - Xuefeng Tang
- Department of Pathology, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China
| | - Hua Qu
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China
| | - Jiaqing Shao
- Department of Endocrinology, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Street No. 305, Xuanwu, Nanjing, China
- Department of Endocrinology, Jinling Hospital, Nanjing Medical University, Zhongshan East Street No. 305, Xuanwu, Nanjing, China
| | - Hongting Zheng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China.
| | - Min Long
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Xinqiao Main Street No. 183, Shapingba, Chongqing, China.
| |
Collapse
|
15
|
Differential Regulation of Gonadotropins as Revealed by Transcriptomes of Distinct LH and FSH Cells of Fish Pituitary. Int J Mol Sci 2021; 22:ijms22126478. [PMID: 34204216 PMCID: PMC8234412 DOI: 10.3390/ijms22126478] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/08/2023] Open
Abstract
From mammals to fish, reproduction is driven by luteinizing hormone (LH) and follicle-stimulating hormone (FSH) temporally secreted from the pituitary gland. Teleost fish are an excellent model for addressing the unique regulation and function of each gonadotropin cell since, unlike mammals, they synthesize and secrete LH and FSH from distinct cells. Only very distant vertebrate classes (such as fish and birds) demonstrate the mono-hormonal strategy, suggesting a potential convergent evolution. Cell-specific transcriptome analysis of double-labeled transgenic tilapia expressing GFP and RFP in LH or FSH cells, respectively, yielded genes specifically enriched in each cell type, revealing differences in hormone regulation, receptor expression, cell signaling, and electrical properties. Each cell type expresses a unique GPCR signature that reveals the direct regulation of metabolic and homeostatic hormones. Comparing these novel transcriptomes to that of rat gonadotrophs revealed conserved genes that might specifically contribute to each gonadotropin activity in mammals, suggesting conserved mechanisms controlling the differential regulation of gonadotropins in vertebrates.
Collapse
|
16
|
Santiago-Andres Y, Golan M, Fiordelisio T. Functional Pituitary Networks in Vertebrates. Front Endocrinol (Lausanne) 2021; 11:619352. [PMID: 33584547 PMCID: PMC7873642 DOI: 10.3389/fendo.2020.619352] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
The pituitary is a master endocrine gland that developed early in vertebrate evolution and therefore exists in all modern vertebrate classes. The last decade has transformed our view of this key organ. Traditionally, the pituitary has been viewed as a randomly organized collection of cells that respond to hypothalamic stimuli by secreting their content. However, recent studies have established that pituitary cells are organized in tightly wired large-scale networks that communicate with each other in both homo and heterotypic manners, allowing the gland to quickly adapt to changing physiological demands. These networks functionally decode and integrate the hypothalamic and systemic stimuli and serve to optimize the pituitary output into the generation of physiologically meaningful hormone pulses. The development of 3D imaging methods and transgenic models have allowed us to expand the research of functional pituitary networks into several vertebrate classes. Here we review the establishment of pituitary cell networks throughout vertebrate evolution and highlight the main perspectives and future directions needed to decipher the way by which pituitary networks serve to generate hormone pulses in vertebrates.
Collapse
Affiliation(s)
- Yorgui Santiago-Andres
- Laboratorio de Neuroendocrinología Comparada, Departamento de Ecología y Recursos Naturales, Biología, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Matan Golan
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Rishon Lezion, Israel
| | - Tatiana Fiordelisio
- Laboratorio de Neuroendocrinología Comparada, Departamento de Ecología y Recursos Naturales, Biología, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| |
Collapse
|
17
|
Chang WT, Ragazzi E, Liu PY, Wu SN. Effective block by pirfenidone, an antifibrotic pyridone compound (5-methyl-1-phenylpyridin-2[H-1]-one), on hyperpolarization-activated cation current: An additional but distinctive target. Eur J Pharmacol 2020; 882:173237. [PMID: 32525005 PMCID: PMC7276140 DOI: 10.1016/j.ejphar.2020.173237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 12/26/2022]
Abstract
Pirfenidone (PFD), a pyridone compound, is well recognized as an antifibrotic agent tailored for the treatment of idiopathic pulmonary fibrosis. Recently, through its anti-inflammatory and anti-oxidant effects, PFD based clinical trial has also been launched for the treatment of coronavirus disease (COVID-19). To what extent this drug can perturb membrane ion currents remains largely unknown. Herein, the exposure to PFD was observed to depress the amplitude of hyperpolarization-activated cation current (Ih) in combination with a considerable slowing in the activation time of the current in pituitary GH3 cells. In the continued presence of ivabradine or zatebradine, subsequent application of PFD decreased Ih amplitude further. The presence of PFD resulted in a leftward shift in Ih activation curve without changes in the gating charge. The addition of this compound also led to a reduction in area of voltage-dependent hysteresis evoked by long-lasting inverted triangular (downsloping and upsloping) ramp pulse. Neither the amplitude of M-type nor erg-mediated K+ current was altered by its presence. In whole-cell potential recordings, addition of PFD reduced the firing frequency, and this effect was accompanied by the depression in the amplitude of sag voltage elicited by hyperpolarizing current stimulus. Overall, this study highlights evidence that PFD is capable of perturbing specific ionic currents, revealing a potential additional impact on functional activities of different excitable cells.
Collapse
Affiliation(s)
- Wei-Ting Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan; Division of Cardiology, Internal Medicine, Chi-Mei Medical Center, Tainan, 71004, Taiwan; Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, 71004, Taiwan.
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Ping-Yen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan; Division of Cardiology, Internal Medicine, College of Medicine, National Cheng Kung University Hospital, Tainan, 70401, Taiwan.
| | - Sheng-Nan Wu
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan, Taiwan; Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
18
|
Zhang J, Ma J, Zhou X, Hu S, Ge L, Sun J, Li P, Long K, Jin L, Tang Q, Liu L, Li X, Shuai S, Li M. Comprehensive Analysis of mRNA and lncRNA Transcriptomes Reveals the Differentially Hypoxic Response of Preadipocytes During Adipogenesis. Front Genet 2020; 11:845. [PMID: 32849828 PMCID: PMC7425071 DOI: 10.3389/fgene.2020.00845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/13/2020] [Indexed: 11/28/2022] Open
Abstract
Local hypoxia has recently been reported to occur in the white adipose tissue (WAT) microenvironment during obesity. Adipocytes have a unique life cycle that reflects the different stages of adipogenesis in the WAT niche. Long non-coding RNAs (lncRNAs) play an important role in the cellular response to hypoxia. However, the differentially hypoxic responses of preadipocytes during adipogenesis and the potential role of lncRNAs in this process remain to be elucidated. Here, we evaluated the differentially hypoxic responses of primary hamster preadipocytes during adipogenesis and analyzed mRNA and lncRNA expression in same Ribo-Zero RNA-seq libraries. Hypoxia induced HIF-1α protein during adipogenesis and caused divergent changes of cell phenotypes. A total of 10,318 mRNAs were identified to be expressed in twenty libraries (five timepoints), and 3,198 differentially expressed mRNAs (DE mRNAs) were detected at five timepoints (hypoxia vs. normoxia). Functional enrichment analysis revealed the shared and specific hypoxia response pathways in the different stages of adipogenesis. Hypoxia differentially modulated the expression profile of adipose-associated genes, including adipokines, lipogenesis, lipolysis, hyperplasia, hypertrophy, inflammatory, and extracellular matrix. We also identified 4,296 lncRNAs that were expressed substantially and detected 1,431 DE lncRNAs at five timepoints. Two, 3, 5, 13, and 50 DE mRNAs at D0, D1, D3, D7, and D11, respectively, were highly correlated and locus-nearby DE lncRNAs and mainly involved in the cell cycle, vesicle-mediated transport, and mitochondrion organization. We identified 28 one-to-one lncRNA-mRNA pairs that might be closely related to adipocyte functions, such as ENSCGRT00015041780-Hilpda, TU2105-Cdsn, and TU17588-Ltbp3. These lncRNAs may represent the crucial regulation axis in the cellular response to hypoxia during adipogenesis. This study dissected the effects of hypoxia in the cell during adipogenesis, uncovered novel regulators potentially associated with WAT function, and may provide a new viewpoint for interpretation and treatment of obesity.
Collapse
Affiliation(s)
- Jinwei Zhang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jideng Ma
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xiankun Zhou
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Silu Hu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China.,Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, China
| | - Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China.,Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, China
| | - Penghao Li
- Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu Xi Nan Gynecological Hospital, Chengdu, China
| | - Keren Long
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Long Jin
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Qianzi Tang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Lingyan Liu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xuewei Li
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Surong Shuai
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Mingzhou Li
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
19
|
Gregory LC, Dattani MT. The Molecular Basis of Congenital Hypopituitarism and Related Disorders. J Clin Endocrinol Metab 2020; 105:5614788. [PMID: 31702014 DOI: 10.1210/clinem/dgz184] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/07/2019] [Indexed: 12/23/2022]
Abstract
CONTEXT Congenital hypopituitarism (CH) is characterized by the presence of deficiencies in one or more of the 6 anterior pituitary (AP) hormones secreted from the 5 different specialized cell types of the AP. During human embryogenesis, hypothalamo-pituitary (HP) development is controlled by a complex spatio-temporal genetic cascade of transcription factors and signaling molecules within the hypothalamus and Rathke's pouch, the primordium of the AP. EVIDENCE ACQUISITION This mini-review discusses the genes and pathways involved in HP development and how mutations of these give rise to CH. This may present in the neonatal period or later on in childhood and may be associated with craniofacial midline structural abnormalities such as cleft lip/palate, visual impairment due to eye abnormalities such as optic nerve hypoplasia (ONH) and microphthalmia or anophthalmia, or midline forebrain neuroradiological defects including agenesis of the septum pellucidum or corpus callosum or the more severe holoprosencephaly. EVIDENCE SYNTHESIS Mutations give rise to an array of highly variable disorders ranging in severity. There are many known causative genes in HP developmental pathways that are routinely screened in CH patients; however, over the last 5 years this list has rapidly increased due to the identification of variants in new genes and pathways of interest by next-generation sequencing. CONCLUSION The majority of patients with these disorders do not have an identified molecular basis, often making management challenging. This mini-review aims to guide clinicians in making a genetic diagnosis based on patient phenotype, which in turn may impact on clinical management.
Collapse
Affiliation(s)
- Louise Cheryl Gregory
- Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Mehul Tulsidas Dattani
- Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
20
|
Review: Mathematical Modeling of Prostate Cancer and Clinical Application. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10082721] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We review and synthesize key findings and limitations of mathematical models for prostate cancer, both from theoretical work and data-validated approaches, especially concerning clinical applications. Our focus is on models of prostate cancer dynamics under treatment, particularly with a view toward optimizing hormone-based treatment schedules and estimating the onset of treatment resistance under various assumptions. Population models suggest that intermittent or adaptive therapy is more beneficial to delay cancer relapse as compared to the standard continuous therapy if treatment resistance comes at a competitive cost for cancer cells. Another consensus among existing work is that the standard biomarker for cancer growth, prostate-specific antigen, may not always correlate well with cancer progression. Instead, its doubling rate appears to be a better indicator of tumor growth. Much of the existing work utilizes simple ordinary differential equations due to difficulty in collecting spatial data and due to the early success of using prostate-specific antigen in mathematical modeling. However, a shift toward more complex and realistic models is taking place, which leaves many of the theoretical and mathematical questions unexplored. Furthermore, as adaptive therapy displays better potential than existing treatment protocols, an increasing number of studies incorporate this treatment into modeling efforts. Although existing modeling work has explored and yielded useful insights on the treatment of prostate cancer, the road to clinical application is still elusive. Among the pertinent issues needed to be addressed to bridge the gap from modeling work to clinical application are (1) real-time data validation and model identification, (2) sensitivity analysis and uncertainty quantification for model prediction, and (3) optimal treatment/schedule while considering drug properties, interactions, and toxicity. To address these issues, we suggest in-depth studies on various aspects of the parameters in dynamical models such as the evolution of parameters over time. We hope this review will assist future attempts at studying prostate cancer.
Collapse
|
21
|
Abstract
The development of the anterior pituitary gland occurs in distinct sequential developmental steps, leading to the formation of a complex organ containing five different cell types secreting six different hormones. During this process, the temporal and spatial expression of a cascade of signaling molecules and transcription factors plays a crucial role in organ commitment, cell proliferation, patterning, and terminal differentiation. The morphogenesis of the gland and the emergence of distinct cell types from a common primordium are governed by complex regulatory networks involving transcription factors and signaling molecules that may be either intrinsic to the developing pituitary or extrinsic, originating from the ventral diencephalon, the oral ectoderm, and the surrounding mesenchyme. Endocrine cells of the pituitary gland are organized into structural and functional networks that contribute to the coordinated response of endocrine cells to stimuli; these cellular networks are formed during embryonic development and are maintained or may be modified in adulthood, contributing to the plasticity of the gland. Abnormalities in any of the steps of pituitary development may lead to congenital hypopituitarism that includes a spectrum of disorders from isolated to combined hormone deficiencies including syndromic disorders such as septo-optic dysplasia. Over the past decade, the acceleration of next-generation sequencing has allowed for rapid analysis of the patient genome to identify novel mutations and novel candidate genes associated with hypothalmo-pituitary development. Subsequent functional analysis using patient fibroblast cells, and the generation of stem cells derived from patient cells, is fast replacing the need for animal models while providing a more physiologically relevant characterization of novel mutations. Furthermore, CRISPR-Cas9 as the method for gene editing is replacing previous laborious and time-consuming gene editing methods that were commonly used, thus yielding knockout cell lines in a fraction of the time. © 2020 American Physiological Society. Compr Physiol 10:389-413, 2020.
Collapse
Affiliation(s)
- Kyriaki S Alatzoglou
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London (UCL), London, UK
| | - Louise C Gregory
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London (UCL), London, UK
| | - Mehul T Dattani
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London (UCL), London, UK
| |
Collapse
|
22
|
GnRH Antagonists Produce Differential Modulation of the Signaling Pathways Mediated by GnRH Receptors. Int J Mol Sci 2019; 20:ijms20225548. [PMID: 31703269 PMCID: PMC6888270 DOI: 10.3390/ijms20225548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Commercial gonadotropin-releasing hormone (GnRH) antagonists differ by 1-2 amino acids and are used to inhibit gonadotropin production during assisted reproduction technologies (ART). In this study, potencies of three GnRH antagonists, Cetrorelix, Ganirelix and Teverelix, in inhibiting GnRH-mediated intracellular signaling, were compared in vitro. GnRH receptor (GnRHR)-transfected HEK293 and neuroblastoma-derived SH-SY5Y cell lines, as well as mouse pituitary LβT2 cells endogenously expressing the murine GnRHR, were treated with GnRH in the presence or absence of the antagonist. We evaluated intracellular calcium (Ca2+) and cAMP increases, cAMP-responsive element binding-protein (CREB) and extracellular-regulated kinase 1 and 2 (ERK1/2) phosphorylation, β-catenin activation and mouse luteinizing-hormone β-encoding gene (Lhb) transcription by bioluminescence resonance energy transfer (BRET), Western blotting, immunostaining and real-time PCR as appropriate. The kinetics of GnRH-induced Ca2+ rapid increase revealed dose-response accumulation with potency (EC50) of 23 nM in transfected HEK293 cells, transfected SH-SY5Y and LβT2 cells. Cetrorelix inhibited the 3 × EC50 GnRH-activated calcium signaling at concentrations of 1 nM-1 µM, demonstrating higher potency than Ganirelix and Teverelix, whose inhibitory doses fell within the 100 nM-1 µM range in both transfected HEK293 and SH-SY5Y cells in vitro. In transfected SH-SY5Y, Cetrorelix was also significantly more potent than other antagonists in reducing GnRH-mediated cAMP accumulation. All antagonists inhibited pERK1/2 and pCREB activation at similar doses, in LβT2 and transfected HEK293 cells treated with 100 nM GnRH. Although immunostainings suggested that Teverelix could be less effective than Cetrorelix and Ganirelix in inhibiting 1 µM GnRH-induced β-catenin activation, Lhb gene expression increase occurring upon LβT2 cell treatment by 1 µM GnRH was similarly inhibited by all antagonists. To conclude, this study has demonstrated Cetrorelix-, Ganirelix- and Teverelix-specific biased effects at the intracellular level, not affecting the efficacy of antagonists in inhibiting Lhb gene transcription.
Collapse
|
23
|
A computational model for gonadotropin releasing cells in the teleost fish medaka. PLoS Comput Biol 2019; 15:e1006662. [PMID: 31437161 PMCID: PMC6726249 DOI: 10.1371/journal.pcbi.1006662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 09/04/2019] [Accepted: 08/01/2019] [Indexed: 01/16/2023] Open
Abstract
Pituitary endocrine cells fire action potentials (APs) to regulate their cytosolic Ca2+ concentration and hormone secretion rate. Depending on animal species, cell type, and biological conditions, pituitary APs are generated either by TTX-sensitive Na+ currents (INa), high-voltage activated Ca2+ currents (ICa), or by a combination of the two. Previous computational models of pituitary cells have mainly been based on data from rats, where INa is largely inactivated at the resting potential, and spontaneous APs are predominantly mediated by ICa. Unlike in rats, spontaneous INa-mediated APs are consistently seen in pituitary cells of several other animal species, including several species of fish. In the current work we develop a computational model of gonadotropin releasing cells in the teleost fish medaka (Oryzias latipes). The model stands out from previous modeling efforts by being (1) the first model of a pituitary cell in teleosts, (2) the first pituitary cell model that fires sponateous APs that are predominantly mediated by INa, and (3) the first pituitary cell model where the kinetics of the depolarizing currents, INa and ICa, are directly fitted to voltage-clamp data. We explore the firing properties of the model, and compare it to the properties of previous models that fire ICa-based APs. We put a particular focus on how the big conductance K+ current (IBK) modulates the AP shape. Interestingly, we find that IBK can prolong AP duration in models that fire ICa-based APs, while it consistently shortens the duration of the predominantly INa-mediated APs in the medaka gonadotroph model. Although the model is constrained to experimental data from gonadotroph cells in medaka, it may likely provide insights also into other pituitary cell types that fire INa-mediated APs. Excitable cells elicit electrical pulses called action potentials (APs), which are generated and shaped by a combination of ion channels in the cell membrane. Since one type of ion channels is permeable to Ca2+ ions, there is typically an influx of Ca2+ during an AP. Pituitary cells therefore use AP firing to regulate their cytosolic Ca2+ concentration, which in turn controls their hormone secretion rate. The amount of Ca2+ that enters during an AP depends strongly on how long it lasts, and it is therefore important to understand the mechanisms that control this. Pituitary APs are generally mediated by a combination of Ca2+ channels and Na+ channels, and the relative contributions of from the two vary between cell types, animal species and biological conditions. Previous computer models have predominantly been adapted to data from pituitary cells which tend to fire Ca2+-based APs. Here we develop a new model, adapted to data from pituitary cells in the fish medaka, which APs that are predominantly Na+-based, and compare its dynamical properties to the previous models that fire Ca2+-based APs.
Collapse
|
24
|
Wang X, Ali MS, Lacerda CMR. Osteogenesis inducers promote distinct biological responses in aortic and mitral valve interstitial cells. J Cell Biochem 2019; 120:11158-11171. [PMID: 30746757 DOI: 10.1002/jcb.28392] [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: 10/04/2018] [Revised: 12/27/2018] [Accepted: 01/09/2019] [Indexed: 01/24/2023]
Abstract
Both aortic and mitral valves calcify in pathological conditions; however, the prevalence of aortic valve calcification is high whereas mitral valve leaflet calcification is somewhat rare. Patterns of valvular calcification may differ due to valvular architecture, but little is known to that effect. In this study, we investigated the intrinsic osteogenic differentiation potential of aortic versus mitral valve interstitial cells provided minimal differentiation conditions. For the assessment of calcification at the cellular level, we used classic inducers of osteogenesis in stem cells: β-glycerophosphate (β-Gly), dexamethasone (Dex), and ascorbate (Asc). In addition to proteomic analyses, osteogenic markers and calcium precipitates were evaluated across treatments of aortic and mitral valve cells. The combination of β-Gly, Asc, and Dex induced aortic valve interstitial cells to synthesize extracellular matrix, overexpress osteoblastic markers, and deposit calcium. However, no strong evidence showed the calcification of mitral valve interstitial cells. Mitral cells mainly responded to Asc and Dex by cell activation. These findings provide a deeper understanding of the physiological properties of aortic and mitral valves and tendencies for calcific changes within each valve type, contributing to the development of future therapeutics for heart valve diseases.
Collapse
Affiliation(s)
- Xinmei Wang
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas
| | - Mir S Ali
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas
| | - Carla M R Lacerda
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas
| |
Collapse
|
25
|
Store-operated Ca2+ entry and Ca2+ responses to hypothalamic releasing hormones in anterior pituitary cells from Orai1−/− and heptaTRPC knockout mice. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1124-1136. [DOI: 10.1016/j.bbamcr.2018.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 01/25/2023]
|
26
|
Abstract
The hypothalamic decapeptide, GnRH, is the gatekeeper of mammalian reproductive development and function. Activation of specific, high-affinity cell surface receptors (GnRH receptors) on gonadotropes by GnRH triggers signal transduction cascades to stimulate the coordinated synthesis and secretion of the pituitary gonadotropins FSH and LH. These hormones direct gonadal steroidogenesis and gametogenesis, making their tightly regulated production and secretion essential for normal sexual maturation and reproductive health. FSH and LH are glycoprotein heterodimers comprised of a common α-subunit and a unique β-subunit (FSHβ and LHβ, respectively), which determines the biological specificity of the gonadotropins. The unique β-subunit is the rate-limiting step for the production of the mature gonadotropins. Therefore, FSH synthesis is regulated at the transcriptional level by Fshb gene expression. The overarching goal of this review is to expand our understanding of the mechanisms and pathways underlying the carefully orchestrated control of FSH synthesis and secretion by GnRH, focusing on the transcriptional regulation of the Fshb gene. Identification of these regulatory mechanisms is not only fundamental to our understanding of normal reproductive function but will also provide a context for the elucidation of the pathophysiology of reproductive disorders and infertility to lead to potential new therapeutic approaches.
Collapse
Affiliation(s)
- George A Stamatiades
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Yale New Haven Health, Bridgeport Hospital, Bridgeport, Connecticut
- School of Medicine, University of Crete, Heraklion, Greece
| | - Rona S Carroll
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
27
|
Gregory LC, Dattani MT. Embryologic and Genetic Disorders of the Pituitary Gland. CONTEMPORARY ENDOCRINOLOGY 2019:3-27. [DOI: 10.1007/978-3-030-11339-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
28
|
Guo S, Li Z, Yan L, Sun Y, Feng Y. GnRH agonist improves pregnancy outcome in mice with induced adenomyosis by restoring endometrial receptivity. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:1621-1631. [PMID: 29922037 PMCID: PMC5995291 DOI: 10.2147/dddt.s162541] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Purpose Adenomyosis has a negative impact on female fertility. GnRH agonist treatment can improve pregnancy outcomes in women with adenomyosis. However, the impact of GnRH agonist upon endometrium receptivity of patients with adenomyosis remains unclear. In this study, endometrial receptivity and pregnancy outcome were investigated using a mouse model of adenomyosis. Materials and methods Adenomyosis was induced in 12 female ICR mice, neonatally treated with tamoxifen, while another six female mice (control group) received solvent only. At 75 days, the induced adenomyosis group was randomly divided into two groups: an untreated group and a group treated with GnRH agonist (n = 6 each). Sixty days later, the mice were mated and pregnancy outcomes were observed and compared among the three groups (n = 6 each). In a parallel experiment using the same treatment regimes, uterus samples were collected on day 4 of pregnancy for immunohistochemistry, gene (quantitative polymerase chain reaction) and protein expression (Western blot), and scanning electron microscopy analyses. Results We found that the average live litter size was reduced in the adenomyosis compared with control group (8 ± 0.56 versus 13 ± 0.71; P = 0.03). However, the litter size was significantly increased in the treated with GnRH agonist group compared with the untreated group (12 ± 0.35 versus 8 ± 0.56; P = 0.04). The uterine expression levels of Hoxa10, Hoxa11, Lif and integrin b3 mRNA and protein were decreased in the adenomyosis group, and were significantly increased after GnRH agonist treatment. Additionally, pinopodes were reduced in number and poorly developed in mice with induced adenomyosis. However, pinopodes were abundant and well-developed in the GnRH agonist treatment group. Conclusion Adenomyosis may have an adverse impact on endometrial receptivity and reduce pregnancy outcomes in mice. However, GnRH agonist may improve the pregnancy outcome by partially restoring endometrial receptivity.
Collapse
Affiliation(s)
- Song Guo
- Department of Gynaecology and Obstetrics, Qianfoshan Hospital, Shandong University, Jinan, China
| | - Zhichao Li
- Gynecology, Shanghai Ji Ai Genetics & In Vitro Fertilization Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Li Yan
- Department of Gynaecology and Obstetrics, Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yijuan Sun
- Gynecology, Shanghai Ji Ai Genetics & In Vitro Fertilization Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yun Feng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Stamatiades GA, Kaiser UB. Gonadotropin regulation by pulsatile GnRH: Signaling and gene expression. Mol Cell Endocrinol 2018; 463:131-141. [PMID: 29102564 PMCID: PMC5812824 DOI: 10.1016/j.mce.2017.10.015] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/27/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022]
Abstract
The precise orchestration of hormonal regulation at all levels of the hypothalamic-pituitary-gonadal axis is essential for normal reproductive function and fertility. The pulsatile secretion of hypothalamic gonadotropin-releasing hormone (GnRH) stimulates the synthesis and release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) by pituitary gonadotropes. GnRH acts by binding to its high affinity seven-transmembrane receptor (GnRHR) on the cell surface of anterior pituitary gonadotropes. Different signaling cascades and transcriptional mechanisms are activated, depending on the variation in GnRH pulse frequency, to stimulate the synthesis and release of FSH and LH. While changes in GnRH pulse frequency may explain some of the differential regulation of FSH and LH, other factors, such as activin, inhibin and sex steroids, also contribute to gonadotropin production. In this review, we focus on the transcriptional regulation of the gonadotropin subunit genes and the signaling pathways activated by pulsatile GnRH.
Collapse
Affiliation(s)
- George A Stamatiades
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
30
|
Martínez-Moreno CG, Calderón-Vallejo D, Harvey S, Arámburo C, Quintanar JL. Growth Hormone (GH) and Gonadotropin-Releasing Hormone (GnRH) in the Central Nervous System: A Potential Neurological Combinatory Therapy? Int J Mol Sci 2018; 19:E375. [PMID: 29373545 PMCID: PMC5855597 DOI: 10.3390/ijms19020375] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 01/21/2018] [Accepted: 01/23/2018] [Indexed: 12/15/2022] Open
Abstract
This brief review of the neurological effects of growth hormone (GH) and gonadotropin-releasing hormone (GnRH) in the brain, particularly in the cerebral cortex, hypothalamus, hippocampus, cerebellum, spinal cord, neural retina, and brain tumors, summarizes recent information about their therapeutic potential as treatments for different neuropathologies and neurodegenerative processes. The effect of GH and GnRH (by independent administration) has been associated with beneficial impacts in patients with brain trauma and spinal cord injuries. Both GH and GnRH have demonstrated potent neurotrophic, neuroprotective, and neuroregenerative action. Positive behavioral and cognitive effects are also associated with GH and GnRH administration. Increasing evidence suggests the possibility of a multifactorial therapy that includes both GH and GnRH.
Collapse
Affiliation(s)
- Carlos G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro 76230, Mexico.
| | - Denisse Calderón-Vallejo
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Av. Universidad 940, Ciudad Universitaria, Aguascalientes 20131, Mexico.
| | - Steve Harvey
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro 76230, Mexico.
| | - José Luis Quintanar
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Av. Universidad 940, Ciudad Universitaria, Aguascalientes 20131, Mexico.
| |
Collapse
|
31
|
Tommiska J, Känsäkoski J, Skibsbye L, Vaaralahti K, Liu X, Lodge EJ, Tang C, Yuan L, Fagerholm R, Kanters JK, Lahermo P, Kaunisto M, Keski-Filppula R, Vuoristo S, Pulli K, Ebeling T, Valanne L, Sankila EM, Kivirikko S, Lääperi M, Casoni F, Giacobini P, Phan-Hug F, Buki T, Tena-Sempere M, Pitteloud N, Veijola R, Lipsanen-Nyman M, Kaunisto K, Mollard P, Andoniadou CL, Hirsch JA, Varjosalo M, Jespersen T, Raivio T. Two missense mutations in KCNQ1 cause pituitary hormone deficiency and maternally inherited gingival fibromatosis. Nat Commun 2017; 8:1289. [PMID: 29097701 PMCID: PMC5668380 DOI: 10.1038/s41467-017-01429-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 09/14/2017] [Indexed: 01/05/2023] Open
Abstract
Familial growth hormone deficiency provides an opportunity to identify new genetic causes of short stature. Here we combine linkage analysis with whole-genome resequencing in patients with growth hormone deficiency and maternally inherited gingival fibromatosis. We report that patients from three unrelated families harbor either of two missense mutations, c.347G>T p.(Arg116Leu) or c.1106C>T p.(Pro369Leu), in KCNQ1, a gene previously implicated in the long QT interval syndrome. Kcnq1 is expressed in hypothalamic GHRH neurons and pituitary somatotropes. Co-expressing KCNQ1 with the KCNE2 β-subunit shows that both KCNQ1 mutants increase current levels in patch clamp analyses and are associated with reduced pituitary hormone secretion from AtT-20 cells. In conclusion, our results reveal a role for the KCNQ1 potassium channel in the regulation of human growth, and show that growth hormone deficiency associated with maternally inherited gingival fibromatosis is an allelic disorder with cardiac arrhythmia syndromes caused by KCNQ1 mutations.
Collapse
Affiliation(s)
- Johanna Tommiska
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland.,Children's Hospital, Pediatric Research Center, Helsinki University Central Hospital (HUCH), 00029, Helsinki, Finland
| | - Johanna Känsäkoski
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Lasse Skibsbye
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Kirsi Vaaralahti
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Xiaonan Liu
- Institute of Biotechnology, Biocenter 3, University of Helsinki, 00014, Helsinki, Finland
| | - Emily J Lodge
- Centre for Craniofacial and Regenerative Biology, King's College London, Floor 27 Tower Wing, Guy's Campus, London, SE1 9RT, UK
| | - Chuyi Tang
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Lei Yuan
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Rainer Fagerholm
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland.,Department of Obstetrics and Gynecology, HUCH, 00029, Helsinki, Finland
| | - Jørgen K Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, 22000, Copenhagen, Denmark.,Department of Cardiology, Herlev & Gentofte University Hospitals, University of Copenhagen, 22000, Copenhagen, Denmark
| | - Päivi Lahermo
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Mari Kaunisto
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | | | - Sanna Vuoristo
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Kristiina Pulli
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Tapani Ebeling
- Department of Medicine, Oulu University Hospital, Finland and Research Unit of Internal Medicine, University of Oulu, 90014, Oulu, Finland
| | - Leena Valanne
- Helsinki Medical Imaging Center, HUCH, 00029, Helsinki, Finland
| | | | - Sirpa Kivirikko
- Department of Clinical Genetics, HUCH, 00029, Helsinki, Finland
| | - Mitja Lääperi
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Filippo Casoni
- Inserm U1172, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, 59045, Lille, France.,University of Lille, School of Medicine, 59045, Lille, France
| | - Paolo Giacobini
- Inserm U1172, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, 59045, Lille, France.,University of Lille, School of Medicine, 59045, Lille, France
| | - Franziska Phan-Hug
- Pediatrics, Division of Pediatric Endocrinology, Diabetology and Obesity, University Hospital Lausanne (CHUV), 1011, Lausanne, Switzerland
| | - Tal Buki
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Institute of Structural Biology, 69978, Ramat Aviv, Israel
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14071, Cordoba, Spain.,Instituto Maimonides de Investigacion Biomedica (IMIBIC/HURS), 14004, Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Nelly Pitteloud
- Pediatrics, Division of Pediatric Endocrinology, Diabetology and Obesity, University Hospital Lausanne (CHUV), 1011, Lausanne, Switzerland
| | - Riitta Veijola
- Department of Children and Adolescents, Oulu University Hospital, 90029, Oulu, Finland.,Department of Pediatrics, PEDEGO Research Center, Medical Research Center, University of Oulu, 90014, Oulu, Finland
| | - Marita Lipsanen-Nyman
- Children's Hospital, Pediatric Research Center, Helsinki University Central Hospital (HUCH), 00029, Helsinki, Finland
| | - Kari Kaunisto
- Department of Children and Adolescents, Oulu University Hospital, 90029, Oulu, Finland
| | - Patrice Mollard
- IGF, CNRS, INSERM, Univ. Montpellier, F-34094, Montpellier, France
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, King's College London, Floor 27 Tower Wing, Guy's Campus, London, SE1 9RT, UK.,Department of Internal Medicine III, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Joel A Hirsch
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Institute of Structural Biology, 69978, Ramat Aviv, Israel
| | - Markku Varjosalo
- Institute of Biotechnology, Biocenter 3, University of Helsinki, 00014, Helsinki, Finland
| | - Thomas Jespersen
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Taneli Raivio
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland. .,Children's Hospital, Pediatric Research Center, Helsinki University Central Hospital (HUCH), 00029, Helsinki, Finland.
| |
Collapse
|
32
|
Sergi C, Shen F, Lim DW, Liu W, Zhang M, Chiu B, Anand V, Sun Z. Cardiovascular dysfunction in sepsis at the dawn of emerging mediators. Biomed Pharmacother 2017; 95:153-160. [PMID: 28841455 DOI: 10.1016/j.biopha.2017.08.066] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/03/2017] [Accepted: 08/13/2017] [Indexed: 12/14/2022] Open
Abstract
Subcellular dysfunction and impaired metabolism derived from the complex interaction of cytokines and mediators with cellular involvement are on the basis of the cardiovascular response to sepsis. The lethal consequences of an infection are intimately related to its ability to spread to other organ sites and the immune system of the host. About one century ago, William Osler (1849-1919), a Canadian physician, remarkably defined the sequelae of the host response in sepsis: "except on few occasions, the patient appears to die from the body's response to infection rather than from it." Cardiac dysfunction has received considerable attention to explain the heart failure in patients progressing from infection to sepsis, but our understanding of the processes remains limited. In fact, most concepts are linked to a mechanical concept of the sarcomeric structure, and physiological data seems to be often disconnected. Cytokines, prostanoids, and nitric oxide release are high direct impact factors, but coronary circulation and cardiomyocyte physiology also play a prominent role in modulating the effects of monocyte adhesion and infiltration. Damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) are involved in the host response. The identification of microRNAs, as well as the cyclic activation of the inflammatory cascade, has further added complexity to the scene. In this review, we delineate the current concepts of cellular dysfunction of the cardiomyocyte in the setting of sepsis and consider potential therapeutic strategies.
Collapse
Affiliation(s)
- Consolato Sergi
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430068, PR China; Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, PR China; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Stollery Children's Hospital, University Alberta Hospital, Edmonton, AB, Canada.
| | - Fan Shen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - David W Lim
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Weiyong Liu
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Mingyong Zhang
- Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, PR China
| | - Brian Chiu
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Vijay Anand
- Department of Critical Care Medicine, University of Alberta, Edmonton, AB, Canada
| | - Ziyong Sun
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| |
Collapse
|
33
|
Domínguez-Mancera B, Barrientos-Morales M, Cervantes-Acosta P, Hernández-Beltrán A, Rodríguez-Andrade A, González-Ramírez R, Monjaraz E, Felix R. Leptin regulation of inward membrane currents, electrical activity and LH release in isolated bovine gonadotropes. Biochem Biophys Res Commun 2017; 491:53-58. [PMID: 28705737 DOI: 10.1016/j.bbrc.2017.07.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/07/2017] [Indexed: 01/16/2023]
Abstract
Leptin, a peptide hormone produced by adipocytes, is recognized as one of the signals involved in the onset of reproductive activity. The leptin receptor has been found in hypothalamic neurons and pituitary gonadotropes, suggesting that the hormone may act at both sites to stimulate the secretion of GnRH and consequently, FSH and LH. In response to a stimulus such as a hypothalamic secretagogue, gonadotropes respond with changes in electrical activity, intracellular Ca2+ and hormone release. The main aim of this report was to investigate whether leptin promotes a change in the electrical and secretory activities of bovine gonadotropes. After 48 h of treatment with leptin (10 nM) significant changes in the action potential properties were observed in gonadotropes, which included an increase in amplitude, time-to-pike and post-hyperpolarization, as well as a decrease in firing threshold. Likewise, leptin induced a significant (∼1.3-fold) up-regulation of voltage-gated Na+ channel current density, and a selective increase (∼2.1-fold) in Ca2+ current density through high voltage-activated channels. Consistent with this, leptin enhanced GnRH-induced secretion of LH measured by ELISA. We suggest that leptin enhances membrane expression of voltage-gated Na+ and Ca2+ channels, which results in a modulation of the action potential properties and an increase in hormone release from gonadotropes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eduardo Monjaraz
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ricardo Felix
- Centre for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico.
| |
Collapse
|