1
|
Machaty Z. The signal that stimulates mammalian embryo development. Front Cell Dev Biol 2024; 12:1474009. [PMID: 39355121 PMCID: PMC11442298 DOI: 10.3389/fcell.2024.1474009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Embryo development is stimulated by calcium (Ca2+) signals that are generated in the egg cytoplasm by the fertilizing sperm. Eggs are formed via oogenesis. They go through a cell division known as meiosis, during which their diploid chromosome number is halved and new genetic combinations are created by crossing over. During formation the eggs also acquire cellular components that are necessary to produce the Ca2+ signal and also, to support development of the newly formed embryo. Ionized calcium is a universal second messenger used by cells in a plethora of biological processes and the eggs develop a "toolkit", a set of molecules needed for signaling. Meiosis stops twice and these arrests are controlled by a complex interaction of regulatory proteins. The first meiotic arrest lasts until after puberty, when a luteinizing hormone surge stimulates meiotic resumption. The cell cycle proceeds to stop again in the middle of the second meiotic division, right before ovulation. The union of the female and male gametes takes place in the oviduct. Following gamete fusion, the sperm triggers the release of Ca2+ from the egg's intracellular stores which in mammals is followed by repetitive Ca2+ spikes known as Ca2+ oscillations in the cytosol that last for several hours. Downstream sensor proteins help decoding the signal and stimulate other molecules whose actions are required for proper development including those that help to prevent the fusion of additional sperm cells to the egg and those that assist in the release from the second meiotic arrest, completion of meiosis and entering the first mitotic cell division. Here I review the major steps of egg formation, discuss the signaling toolkit that is essential to generate the Ca2+ signal and describe the steps of the signal transduction mechanism that activates the egg's developmental program and turns it into an embryo.
Collapse
Affiliation(s)
- Zoltan Machaty
- Department of Animal Sciences Purdue University West Lafayette, West Lafayette, IN, United States
| |
Collapse
|
2
|
Chen C, Huang Z, Dong S, Ding M, Li J, Wang M, Zeng X, Zhang X, Sun X. Calcium signaling in oocyte quality and functionality and its application. Front Endocrinol (Lausanne) 2024; 15:1411000. [PMID: 39220364 PMCID: PMC11361953 DOI: 10.3389/fendo.2024.1411000] [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: 04/02/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Calcium (Ca2+) is a second messenger for many signal pathways, and changes in intracellular Ca2+ concentration ([Ca2+]i) are an important signaling mechanism in the oocyte maturation, activation, fertilization, function regulation of granulosa and cumulus cells and offspring development. Ca2+ oscillations occur during oocyte maturation and fertilization, which are maintained by Ca2+ stores and extracellular Ca2+ ([Ca2+]e). Abnormalities in Ca2+ signaling can affect the release of the first polar body, the first meiotic division, and chromosome and spindle morphology. Well-studied aspects of Ca2+ signaling in the oocyte are oocyte activation and fertilization. Oocyte activation, driven by sperm-specific phospholipase PLCζ, is initiated by concerted intracellular patterns of Ca2+ release, termed Ca2+ oscillations. Ca2+ oscillations persist for a long time during fertilization and are coordinately engaged by a variety of Ca2+ channels, pumps, regulatory proteins and their partners. Calcium signaling also regulates granulosa and cumulus cells' function, which further affects oocyte maturation and fertilization outcome. Clinically, there are several physical and chemical options for treating fertilization failure through oocyte activation. Additionally, various exogenous compounds or drugs can cause ovarian dysfunction and female infertility by inducing abnormal Ca2+ signaling or Ca2+ dyshomeostasis in oocytes and granulosa cells. Therefore, the reproductive health risks caused by adverse stresses should arouse our attention. This review will systematically summarize the latest research progress on the aforementioned aspects and propose further research directions on calcium signaling in female reproduction.
Collapse
Affiliation(s)
- Chen Chen
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Zefan Huang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Shijue Dong
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Mengqian Ding
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Jinran Li
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Miaomiao Wang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xuhui Zeng
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xiaoning Zhang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xiaoli Sun
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
3
|
Palay P, Fathi D, Saffari H, Hassani F, Hajiaghalou S, Fathi R. Simple bioelectrical microsensor: oocyte quality prediction via membrane electrophysiological characterization. LAB ON A CHIP 2024; 24:3909-3929. [PMID: 38985018 DOI: 10.1039/d3lc01120h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Oocyte selection is a crucial step of assisted reproductive treatment. The most common approach relies on the embryologist experience which is inevitably prone to human error. One potential approach could be the use of an electrical-based approach as an ameliorative alternative. Here, we developed a simple electrical microsensor to characterize mouse oocytes. The sensor is designed similarly to embryo culture dishes and is familiar to embryologists. Different microelectrode models were simulated for oocyte cells and a more sensitive model was determined. The final microsensor was fabricated. A differential measuring technique was proposed based on the cell presence/absence. We predicted oocyte quality by using three electrical characteristics, oocyte radii, and zona thicknesses, and also these predictions were compared with an embryologist evaluation. The evaluation of the oocyte membrane capacitance, as an electrophysiological characteristic, was found to be a more reliable method for predicting oocytes with fertilization and blastocyst formation success competence. It achieved 94% and 58% prediction accuracies, respectively, surpassing other methods and yielding lower errors. This groundbreaking research represents the first of its kind in this field and we hope that this will be a step towards improving the accuracy of the treatment.
Collapse
Affiliation(s)
- Peyman Palay
- Department of Electrical and Computer Engineering, Tarbiat Modares University (TMU), Tehran, Iran.
| | - Davood Fathi
- Department of Electrical and Computer Engineering, Tarbiat Modares University (TMU), Tehran, Iran.
| | - Hassan Saffari
- Department of Electrical and Computer Engineering, Tarbiat Modares University (TMU), Tehran, Iran.
| | - Fatemeh Hassani
- Department of Embryology, Reproductive Biomedicine Research Center, Academic Center for Education, Culture and Research (ACECR), Royan Institute for Reproductive Biomedicine, Tehran, Iran.
| | - Samira Hajiaghalou
- Department of Embryology, Reproductive Biomedicine Research Center, Academic Center for Education, Culture and Research (ACECR), Royan Institute for Reproductive Biomedicine, Tehran, Iran.
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Academic Center for Education, Culture and Research (ACECR), Royan Institute for Reproductive Biomedicine, Tehran, Iran.
| |
Collapse
|
4
|
Zeber-Lubecka N, Suchta K, Kulecka M, Kluska A, Piątkowska M, Dabrowski MJ, Jankowska K, Grymowicz M, Smolarczyk R, Hennig EE. Exome sequencing to explore the possibility of predicting genetic susceptibility to the joint occurrence of polycystic ovary syndrome and Hashimoto's thyroiditis. Front Immunol 2023; 14:1193293. [PMID: 37545519 PMCID: PMC10397507 DOI: 10.3389/fimmu.2023.1193293] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
A large body of evidence indicates that women with polycystic ovary syndrome (PCOS) have a higher risk of developing Hashimoto's thyroiditis (HT) than healthy individuals. Given the strong genetic impact on both diseases, common predisposing genetic factors are possibly involved but are not fully understood. Here, we performed whole-exome sequencing (WES) for 250 women with sporadic PCOS, HT, combined PCOS and HT (PCOS+HT), and healthy controls to explore the genetic background of the joint occurrence of PCOS and HT. Based on relevant comparative analyses, multivariate logistic regression prediction modeling, and the most informative feature selection using the Monte Carlo feature selection and interdependency discovery algorithm, 77 variants were selected for further validation by TaqMan genotyping in a group of 533 patients. In the allele frequency test, variants in RAB6A, GBP3, and FNDC7 genes were found to significantly (padjusted < 0.05) differentiated the PCOS+HT and PCOS groups, variant in HIF3A differentiated the PCOS+HT and HT groups, whereas variants in CDK20 and CCDC71 differentiated the PCOS+HT and both single disorder groups. TaqMan genotyping data were used to create final prediction models, which differentiated between PCOS+HT and PCOS or HT with a prediction accuracy of AUC = 0.78. Using a 70% cutoff of the prediction score improved the model parameters, increasing the AUC value to 0.87. In summary, we demonstrated the polygenic burden of both PCOS and HT, and many common and intersecting signaling pathways and biological processes whose disorders mutually predispose patients to the development of both diseases.
Collapse
Affiliation(s)
- Natalia Zeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Katarzyna Suchta
- Department of Gynaecological Endocrinology, Medical University of Warsaw, Warsaw, Poland
| | - Maria Kulecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Anna Kluska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Magdalena Piątkowska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Katarzyna Jankowska
- Department of Endocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Monika Grymowicz
- Department of Gynaecological Endocrinology, Medical University of Warsaw, Warsaw, Poland
| | - Roman Smolarczyk
- Department of Gynaecological Endocrinology, Medical University of Warsaw, Warsaw, Poland
| | - Ewa E. Hennig
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
5
|
Savy V, Stein P, Shi M, Williams CJ. PMCA1 depletion in mouse eggs amplifies calcium signaling and impacts offspring growth†. Biol Reprod 2022; 107:1439-1451. [PMID: 36130203 PMCID: PMC10144700 DOI: 10.1093/biolre/ioac180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/19/2022] [Accepted: 09/19/2022] [Indexed: 11/12/2022] Open
Abstract
Egg activation in mammals is triggered by oscillations in egg intracellular calcium (Ca2+) level. Ca2+ oscillation patterns can be modified in vitro by changing the ionic composition of culture media or in vivo by conditions affecting mitochondrial function, such as obesity and inflammation. In mice, disruption of Ca2+ oscillations in vitro impacts embryo development and offspring growth. Here we tested the hypothesis that, even without in vitro manipulation, abnormal Ca2+ signaling following fertilization impacts offspring growth. Plasma membrane Ca2+ ATPases (PMCA) extrude cytosolic Ca2+ to restore Ca2+ homeostasis. To disrupt Ca2+ signaling in vivo, we conditionally deleted PMCA1 (cKO) in oocytes. As anticipated, in vitro fertilized cKO eggs had increased Ca2+ exposure relative to controls. To assess the impact on offspring growth, cKO females were mated to wild type males to generate pups that had high Ca2+ exposure at fertilization. Because these offspring would be heterozygous, we also tested the impact of global PMCA1 heterozygosity on offspring growth. Control heterozygous pups that had normal Ca2+ at fertilization were generated by mating wild type females to heterozygous males; these control offspring weighed significantly less than their wild type siblings. However, heterozygous offspring from cKO eggs (and high Ca2+ exposure) were larger than heterozygous controls at 12 week-of-age and males had altered body composition. Our results show that global PMCA1 haploinsufficiency impacts growth and support that abnormal Ca2+ signaling after fertilization in vivo has a long-term impact on offspring weight. These findings are relevant for environmental and medical conditions affecting Ca2+ handling and for design of culture conditions and procedures for domestic animal and human assisted reproduction.
Collapse
Affiliation(s)
- Virginia Savy
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Paula Stein
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Min Shi
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Carmen J Williams
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| |
Collapse
|
6
|
吴 江, 吴 永, 杨 韵, 余 靖, 傅 饶, 孙 悦, 肖 谦. [Mibefradil improves skeletal muscle mass, function and structure in obese mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1032-1037. [PMID: 35869766 PMCID: PMC9308873 DOI: 10.12122/j.issn.1673-4254.2022.07.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To observe the effect of mibefradil on skeletal muscle mass, function and structure in obese mice. METHODS Fifteen 6-week-old C57BL/6 mice were randomized equally into normal diet group (control group), high-fat diet (HFD) group and high-fat diet +mibefradil intervention group (HFD +Mibe group). The grip strength of the mice was measured using an electronic grip strength meter, and the muscle content of the hindlimb was analyzed by X-ray absorptiometry (DXA). Triglyceride (TG) and total cholesterol (TC) levels of the mice were measured with GPO-PAP method. The cross-sectional area of the muscle fibers was observed with HE staining. The changes in the level of autophagy in the muscles were detected by Western blotting and immunofluorescence assay, and the activation of the Akt/mTOR signaling pathway was detected with Western blotting. RESULTS Compared with those in the control group, the mice in HFD group had a significantly greater body weight, lower relative grip strength, smaller average cross sectional area of the muscle fibers, and a lower hindlimb muscle ratio (P < 0.05). Immunofluorescence assay revealed a homogenous distribution of LC3 emitting light red fluorescence in the cytoplasm in the muscle cells in HFD group and HFD+Mibe group, while bright spots of red fluorescence were detected in HFD group. In HFD group, the muscular tissues of the mice showed an increased expression level of LC3 II protein with lowered expressions of p62 protein and phosphorylated AKT and mTOR (P < 0.05). Mibefradil treatment significantly reduced body weight of the mice, lowered the expression level of p62 protein, and increased forelimb grip strength, hindlimb muscle ratio, cross-sectional area of the muscle fibers, and the expression levels of LC3 II protein and phosphorylated AKT and mTOR (P < 0.05). CONCLUSION Mibefradil treatment can moderate high-fat diet-induced weight gain and improve muscle mass and function in obese mice possibly by activating AKT/mTOR signal pathway to improve lipid metabolism and inhibit obesityinduced autophagy.
Collapse
Affiliation(s)
- 江豪 吴
- />重庆医科大学附属第一医院老年病科,重庆 400016Department of Geriatrics, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - 永鑫 吴
- />重庆医科大学附属第一医院老年病科,重庆 400016Department of Geriatrics, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - 韵菲 杨
- />重庆医科大学附属第一医院老年病科,重庆 400016Department of Geriatrics, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - 靖 余
- />重庆医科大学附属第一医院老年病科,重庆 400016Department of Geriatrics, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - 饶 傅
- />重庆医科大学附属第一医院老年病科,重庆 400016Department of Geriatrics, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - 悦 孙
- />重庆医科大学附属第一医院老年病科,重庆 400016Department of Geriatrics, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - 谦 肖
- />重庆医科大学附属第一医院老年病科,重庆 400016Department of Geriatrics, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
7
|
Shafqat A, Kashir J, Alsalameh S, Alkattan K, Yaqinuddin A. Fertilization, Oocyte Activation, Calcium Release and Epigenetic Remodelling: Lessons From Cancer Models. Front Cell Dev Biol 2022; 10:781953. [PMID: 35309905 PMCID: PMC8931327 DOI: 10.3389/fcell.2022.781953] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/14/2022] [Indexed: 12/11/2022] Open
Abstract
Oocyte activation deficiency (OAD) is the basis of Total Fertilisation Failure (TFF) and is attributed to mutations in the PLCζ gene—termed male factor infertility. This derives abnormal Ca2+ oscillations and could be the main cause of primary disruptions in the gene expression of Ca2+-related proteins. Epigenetic mechanisms are universally accepted as key regulators of gene expression. However, epigenetic dysregulations have not been considered as potential mechanisms of oocyte-borne OAD. Herein, we discuss changes in the DNA methylome during oogenesis and embryogenesis. We further highlight key pathways comprising the oocyte Ca2+ toolkit, which could be targets of epigenetic alterations, especially aberrations in DNA methylation. Considering that the vast majority of epigenetic modifications examined during fertilization revolve around alterations in DNA methylation, we aim in this article to associate Ca2+-specific mechanisms with these alterations. To strengthen this perspective, we bring evidence from cancer research on the intricate link between DNA methylation and Ca2+ signaling as cancer research has examined such questions in a lot more detail. From a therapeutic standpoint, if our hypothesis is proven to be correct, this will explain the cause of TFF in idiopathic cases and will open doors for novel therapeutic targets.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- *Correspondence: Ahmed Yaqinuddin,
| |
Collapse
|
8
|
Rojas J, Hinostroza F, Vergara S, Pinto-Borguero I, Aguilera F, Fuentes R, Carvacho I. Knockin' on Egg's Door: Maternal Control of Egg Activation That Influences Cortical Granule Exocytosis in Animal Species. Front Cell Dev Biol 2021; 9:704867. [PMID: 34540828 PMCID: PMC8446563 DOI: 10.3389/fcell.2021.704867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/16/2021] [Indexed: 12/23/2022] Open
Abstract
Fertilization by multiple sperm leads to lethal chromosomal number abnormalities, failed embryo development, and miscarriage. In some vertebrate and invertebrate eggs, the so-called cortical reaction contributes to their activation and prevents polyspermy during fertilization. This process involves biogenesis, redistribution, and subsequent accumulation of cortical granules (CGs) at the female gamete cortex during oogenesis. CGs are oocyte- and egg-specific secretory vesicles whose content is discharged during fertilization to block polyspermy. Here, we summarize the molecular mechanisms controlling critical aspects of CG biology prior to and after the gametes interaction. This allows to block polyspermy and provide protection to the developing embryo. We also examine how CGs form and are spatially redistributed during oogenesis. During egg activation, CG exocytosis (CGE) and content release are triggered by increases in intracellular calcium and relies on the function of maternally-loaded proteins. We also discuss how mutations in these factors impact CG dynamics, providing unprecedented models to investigate the genetic program executing fertilization. We further explore the phylogenetic distribution of maternal proteins and signaling pathways contributing to CGE and egg activation. We conclude that many important biological questions and genotype–phenotype relationships during fertilization remain unresolved, and therefore, novel molecular players of CG biology need to be discovered. Future functional and image-based studies are expected to elucidate the identity of genetic candidates and components of the molecular machinery involved in the egg activation. This, will open new therapeutic avenues for treating infertility in humans.
Collapse
Affiliation(s)
- Japhet Rojas
- Laboratorio Fisiología de la Reproducción, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile.,Escuela de Ingeniería en Biotecnología, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Talca, Chile
| | - Fernando Hinostroza
- Laboratorio Fisiología de la Reproducción, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile.,Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile.,Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Facultad de Ciencias de la Salud, Universidad Católica del Maule, Talca, Chile
| | - Sebastián Vergara
- Laboratorio Fisiología de la Reproducción, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile.,Escuela de Ingeniería en Biotecnología, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Talca, Chile
| | - Ingrid Pinto-Borguero
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Felipe Aguilera
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ricardo Fuentes
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ingrid Carvacho
- Laboratorio Fisiología de la Reproducción, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile
| |
Collapse
|
9
|
Papazoglou A, Henseler C, Broich K, Daubner J, Weiergräber M. Breeding of Ca v2.3 deficient mice reveals Mendelian inheritance in contrast to complex inheritance in Ca v3.2 null mutant breeding. Sci Rep 2021; 11:13972. [PMID: 34234221 PMCID: PMC8263769 DOI: 10.1038/s41598-021-93391-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/23/2021] [Indexed: 11/10/2022] Open
Abstract
High voltage-activated Cav2.3 R-type Ca2+ channels and low voltage-activated Cav3.2 T-type Ca2+ channels were reported to be involved in numerous physiological and pathophysiological processes. Many of these findings are based on studies in Cav2.3 and Cav3.2 deficient mice. Recently, it has been proposed that inbreeding of Cav2.3 and Cav3.2 deficient mice exhibits significant deviation from Mendelian inheritance and might be an indication for potential prenatal lethality in these lines. In our study, we analyzed 926 offspring from Cav3.2 breedings and 1142 offspring from Cav2.3 breedings. Our results demonstrate that breeding of Cav2.3 deficient mice shows typical Mendelian inheritance and that there is no indication of prenatal lethality. In contrast, Cav3.2 breeding exhibits a complex inheritance pattern. It might be speculated that the differences in inheritance, particularly for Cav2.3 breeding, are related to other factors, such as genetic specificities of the mutant lines, compensatory mechanisms and altered sperm activity.
Collapse
Affiliation(s)
- Anna Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Christina Henseler
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Johanna Daubner
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Marco Weiergräber
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany.
| |
Collapse
|
10
|
Mehregan A, Ardestani G, Akizawa H, Carvacho I, Fissore R. Deletion of TRPV3 and CaV3.2 T-type channels in mice undermines fertility and Ca2+ homeostasis in oocytes and eggs. J Cell Sci 2021; 134:jcs257956. [PMID: 34313315 PMCID: PMC8313860 DOI: 10.1242/jcs.257956] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 05/17/2021] [Indexed: 12/19/2022] Open
Abstract
Ca2+ influx during oocyte maturation and after sperm entry is necessary to fill the internal Ca2+ stores and for complete egg activation. We knocked out the transient receptor potential vanilloid member 3 (TRPV3) and the T-type channel, CaV3.2, to determine their necessity for maintaining these functions in mammalian oocytes/eggs. Double-knockout (dKO) females were subfertile, their oocytes and eggs showed reduced internal Ca2+ stores, and, following sperm entry or Plcz (also known as Plcz1) cRNA injection, fewer dKO eggs displayed Ca2+ responses compared to wild-type eggs, which were also of lower frequency. These parameters were rescued and/or enhanced by removing extracellular Mg2+, suggesting that the residual Ca2+ influx could be mediated by the TRPM7 channel, consistent with the termination of divalent-cation oscillations in dKO eggs by a TRPM7 inhibitor. In total, we demonstrated that TRPV3 and CaV3.2 mediate the complete filling of the Ca2+ stores in mouse oocytes and eggs. We also showed that they are required for initiating and maintaining regularly spaced-out oscillations, suggesting that Ca2+ influx through PM ion channels dictates the periodicity and persistence of Ca2+ oscillations during mammalian fertilization.
Collapse
Affiliation(s)
- Aujan Mehregan
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA 01003, USA
| | - Goli Ardestani
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA 01003, USA
| | - Hiroki Akizawa
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA 01003, USA
| | - Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, 3480112 Talca, Chile
| | - Rafael Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|
11
|
Tarsani E, Kranis A, Maniatis G, Hager-Theodorides AL, Kominakis A. Detection of loci exhibiting pleiotropic effects on body weight and egg number in female broilers. Sci Rep 2021; 11:7441. [PMID: 33811218 PMCID: PMC8018976 DOI: 10.1038/s41598-021-86817-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
The objective of the present study was to discover the genetic variants, functional candidate genes, biological processes and molecular functions underlying the negative genetic correlation observed between body weight (BW) and egg number (EN) traits in female broilers. To this end, first a bivariate genome-wide association and second stepwise conditional-joint analyses were performed using 2586 female broilers and 240 k autosomal SNPs. The aforementioned analyses resulted in a total number of 49 independent cross-phenotype (CP) significant SNPs with 35 independent markers showing antagonistic action i.e., positive effects on one trait and negative effects on the other trait. A number of 33 independent CP SNPs were located within 26 and 14 protein coding and long non-coding RNA genes, respectively. Furthermore, 26 independent markers were situated within 44 reported QTLs, most of them related to growth traits. Investigation of the functional role of protein coding genes via pathway and gene ontology analyses highlighted four candidates (CPEB3, ACVR1, MAST2 and CACNA1H) as most plausible pleiotropic genes for the traits under study. Three candidates (CPEB3, MAST2 and CACNA1H) were associated with antagonistic pleiotropy, while ACVR1 with synergistic pleiotropic action. Current results provide a novel insight into the biological mechanism of the genetic trade-off between growth and reproduction, in broilers.
Collapse
Affiliation(s)
- Eirini Tarsani
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece.
| | - Andreas Kranis
- Aviagen, Newbridge, EH28 8SZ, Midlothian, UK
- The Roslin Institute, University of Edinburgh, Midlothian, EH25 9RG, UK
| | | | - Ariadne L Hager-Theodorides
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| | - Antonios Kominakis
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| |
Collapse
|
12
|
Chen M, Li S, Hao M, Chen J, Zhao Z, Hong S, Min J, Tang J, Hu M, Hong L. T-type calcium channel blockade induces apoptosis in C2C12 myotubes and skeletal muscle via endoplasmic reticulum stress activation. FEBS Open Bio 2020; 10:2122-2136. [PMID: 32865339 PMCID: PMC7530395 DOI: 10.1002/2211-5463.12965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/11/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022] Open
Abstract
Loss of T‐type calcium channel (TCC) function has been reported to result in decreased cell viability and impaired muscle regeneration, but the underlying mechanisms remain largely unknown. We previously found that expression of TCC is reduced in aged pelvic floor muscle of multiple vaginal delivery mice, and this is related to endoplasmic reticulum stress (ERS) activation and autophagy flux blockade. In the present work, we further investigated the effects of TCC function loss on C2C12 myotubes and skeletal muscle, which is mediated by promotion of ERS and ultimately contributes to mitochondrial‐related apoptotic cell death. We found that application of a TCC inhibitor induced mitochondria‐related apoptosis in a dose‐dependent manner and also reduced mitochondrial transmembrane potential (MMP), induced mito‐ROS generation, and enhanced expression of mitochondrial apoptosis proteins. Functional inhibition of TCC induced ERS, resulting in disorder of Ca2+ homeostasis in endoplasmic reticulum, and ultimately leading to cell apoptosis in C2C12 myotubes. Tibialis anterior muscles of T‐type α1H channel knockout mice displayed a smaller skeletal muscle fiber size and elevated ERS‐mediated apoptosis signaling. Our data point to a novel mechanism whereby TCC blockade leads to ERS activation and terminal mitochondrial‐related apoptotic events in C2C12 myotubes and skeletal muscles.
Collapse
Affiliation(s)
- Mao Chen
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Suting Li
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Menglei Hao
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Jue Chen
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Zhihan Zhao
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Shasha Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Jie Min
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Jianming Tang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Ming Hu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| |
Collapse
|
13
|
Handayani N, Wiweko B, Zakirah SC, Boediono A. In vitro Activation of Mouse Oocytes through Intracellular Ca2+ Regulation. J Hum Reprod Sci 2020; 13:138-144. [PMID: 32792763 PMCID: PMC7394099 DOI: 10.4103/jhrs.jhrs_122_19] [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: 10/25/2019] [Revised: 12/26/2019] [Accepted: 02/28/2020] [Indexed: 11/30/2022] Open
Abstract
Background: Ca2+ signaling pathway is suggested to play an essential role in mediating oocyte maturation. Aims: The aim of this study was to evaluate intracellular Ca2+ of resistant immature oocytes that failed to resume meiosis following subsequent in vitro culture reach metaphase II after calcium ionophore A23187 activation. Settings and Design: This in vitro analytical experimental study was conducted at Animal Science Laboratory of Indonesian Medical Education and Research Institute (IMERI), Human Reproductive Infertility and Family Planning of IMERI, and Electrophysiology Imaging of Terpadu Laboratory, Faculty of Medicine, University of Indonesia. Methods: A total of 308 oocytes classed as resistant immature following in vitro culture were randomly allocated to control (n = 113) and treatment groups (n = 195). The oocyte activation group was exposed to A23187 solution for 15 min and then washed extensively. Maturation was evaluated by observing the first polar body extrusion 20‒24 h after A23187 exposure. Ca2+ imaging was conducted using a confocal laser scanning microscope to identify the dynamic of Ca2+ response. Statistical Analysis: SPSS 20, Chi-square, and Mann–Whitney U-test were used in this study. Results: Activation of resistant immature oocytes with A23187 significantly increased the number of oocyte maturation compared with the control group (P < 0.001). Furthermore, fluorescent intensity measurements exhibited a significant increase in the germinal vesicle stage when activated (P = 0.005), as well as the metaphase I stage, even though differences were not significant (P = 0.146). Conclusion: Artificial activation of resistant immature oocyte using chemical A23187/calcimycin was adequate to initiate meiosis progress.
Collapse
Affiliation(s)
- Nining Handayani
- Reproductive Science Master Program of Biomedical Science, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Budi Wiweko
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia.,Yasmin IVF Clinic, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia.,Human Reproductive, Infertility, and Family Planning Research Center, Indonesian Medical Education and Research Institutes, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Sarah Chairani Zakirah
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia.,Human Reproductive, Infertility, and Family Planning Research Center, Indonesian Medical Education and Research Institutes, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Arief Boediono
- Department of Anatomy, Physiology and Pharmacology, IPB University, Bogor, Indonesia
| |
Collapse
|
14
|
Stein P, Savy V, Williams AM, Williams CJ. Modulators of calcium signalling at fertilization. Open Biol 2020; 10:200118. [PMID: 32673518 PMCID: PMC7574550 DOI: 10.1098/rsob.200118] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
Calcium (Ca2+) signals initiate egg activation across the animal kingdom and in at least some plants. These signals are crucial for the success of development and, in the case of mammals, health of the offspring. The mechanisms associated with fertilization that trigger these signals and the molecules that regulate their characteristic patterns vary widely. With few exceptions, a major contributor to fertilization-induced elevation in cytoplasmic Ca2+ is release from endoplasmic reticulum stores through the IP3 receptor. In some cases, Ca2+ influx from the extracellular space and/or release from alternative intracellular stores contribute to the rise in cytoplasmic Ca2+. Following the Ca2+ rise, the reuptake of Ca2+ into intracellular stores or efflux of Ca2+ out of the egg drive the return of cytoplasmic Ca2+ back to baseline levels. The molecular mediators of these Ca2+ fluxes in different organisms include Ca2+ release channels, uptake channels, exchangers and pumps. The functions of these mediators are regulated by their particular activating mechanisms but also by alterations in their expression and spatial organization. We discuss here the molecular basis for modulation of Ca2+ signalling at fertilization, highlighting differences across several animal phyla, and we mention key areas where questions remain.
Collapse
Affiliation(s)
- Paula Stein
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Virginia Savy
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Audrey M. Williams
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Carmen J. Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
15
|
Li S, Hao M, Li B, Chen M, Chen J, Tang J, Hong S, Min J, Hu M, Hong L. CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade. Cell Death Dis 2020; 11:279. [PMID: 32332705 PMCID: PMC7181873 DOI: 10.1038/s41419-020-2484-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023]
Abstract
Multiple vaginal delivery (MVD) is an important factor for pelvic floor muscle (PFM) function decline and pelvic floor dysfunction (PFD). PFD is common in middle-aged and elderly women, but its pathogenesis is not clear. In this study, we found that the expression of CACNA1H was lower in the PFM of old mice after MVD compared with old or adult mice. In in-vitro studies, we found that treatment with the T-type Ca2+ channel (T-channel) inhibitor NNC-55 or downregulation of the CACNA1H gene by siRNA intervention promoted myotube atrophy and apoptosis. Mechanistically, we revealed that NNC-55 increased the expression of GRP78 and DDIT3 in myotubes, indicating endoplasmic reticulum stress (ERS) activation, and that the IRE1 and PERK pathways might be involved in this effect. NNC-55 induced the formation of autophagosomes but inhibited autophagy flux. Moreover, rapamycin, an autophagy activator, did not rescue myotube atrophy or apoptosis induced by NNC-55, and the autophagy inhibitors 3-MA and HCQ accelerated this damage. Further studies showed that the ERS inhibitors 4-PBA and TUDAC relieved NNC-55-induced damage and autophagy flux blockade. Finally, we found multisite muscle atrophy and decreased muscle function in Cacna1h−/− (TH-null) mice, as well as increased autophagy inhibition and apoptotic signals in the PFM of old WT mice after MVD and TH-null mice. Taken together, our results suggest that MVD-associated PFD is partially attributed to CACNA1H downregulation-induced PFM atrophy and that ERS is a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Suting Li
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Menglei Hao
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Bingshu Li
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Mao Chen
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Jue Chen
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Jianming Tang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Shasha Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Jie Min
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Ming Hu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China.
| |
Collapse
|
16
|
Ardestani G, Mehregan A, Fleig A, Horgen FD, Carvacho I, Fissore RA. Divalent cation influx and calcium homeostasis in germinal vesicle mouse oocytes. Cell Calcium 2020; 87:102181. [PMID: 32097818 DOI: 10.1016/j.ceca.2020.102181] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 11/30/2022]
Abstract
Prior to maturation, mouse oocytes are arrested at the germinal vesicle (GV) stage during which they experience constitutive calcium (Ca2+) influx and spontaneous Ca2+ oscillations. The oscillations cease during maturation but Ca2+ influx continues, as the oocytes' internal stores attain maximal content at the culmination of maturation, the metaphase II stage. The identity of the channel(s) that underlie this Ca2+ influx has not been completely determined. GV and matured oocytes are known to express three Ca2+ channels, CaV3.2, TRPV3 and TRPM7, but females null for each of these channels are fertile and their oocytes display minor modifications in Ca2+ homeostasis, suggesting a complex regulation of Ca2+ influx. To define the contribution of these channels at the GV stage, we used different divalent cations, pharmacological inhibitors and genetic models. We found that the three channels are active at this stage. CaV3.2 and TRPM7 channels contributed the majority of Ca2+ influx, as inhibitors and oocytes from homologous knockout (KO) lines showed severely reduced Ca2+ entry. Sr2+ influx was promoted by CaV3.2 channels, as Sr2+ oscillations were negligible in CaV3.2-KO oocytes but robust in control and Trpv3-KO GV oocytes. Mn2+ entry relied on expression of CaV3.2 and TRPM7 channels, but Ni2+ entry depended on the latter. CaV3.2 and TRPV3 channels combined to fill the Ca2+ stores, although CaV3.2 was the most impactful. Studies with pharmacological inhibitors effectively blocked the influx of divalent cations, but displayed off-target effects, and occasionally agonist-like properties. In conclusion, GV oocytes express channels mediating Ca2+ and other divalent cation influx that are pivotal for fertilization and early development. These channels may serve as targets for intervention to improve the success of assisted reproductive technologies.
Collapse
Affiliation(s)
- Goli Ardestani
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA, 01003, USA
| | - Aujan Mehregan
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA, 01003, USA
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and University of Hawaii Cancer Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, HI, 96813, USA
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI, 96744, USA
| | - Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, 3480112, Talca, Chile
| | - Rafael A Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA, 01003, USA.
| |
Collapse
|
17
|
McDonough CE, Bernhardt ML, Williams CJ. Mouse strain-dependent egg factors regulate calcium signals at fertilization. Mol Reprod Dev 2020; 87:284-292. [PMID: 31944466 DOI: 10.1002/mrd.23316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/12/2019] [Indexed: 01/29/2023]
Abstract
Calcium (Ca2+ ) signals triggered at fertilization initiate resumption of the cell cycle and initial steps of embryonic development. In mammals, the sperm factor phospholipase Cζ triggers the release of Ca2+ from the endoplasmic reticulum (ER), initiating an oscillatory pattern of Ca2+ transients that is modulated by egg factors including Ca2+ influx channels, Ca2+ transporters, and phosphoinositide-regulating enzymes. Here we compared characteristics of Ca2+ oscillations following in vitro fertilization (IVF) and ER Ca2+ stores among nine common laboratory mouse strains: CF1, C57BL6, SJL, CD1, DBA, FVB, 129X1, BALBc, 129S1, and the F1 hybrid B6129SF1. Sperm from B6SJLF1/J males was used for all IVF experiments. There were significant differences among the strains with respect to duration and maximum amplitude of the first Ca2+ transient, frequency of oscillations, and ER Ca2+ stores. With male strain held constant, the differences in Ca2+ oscillation patterns observed result from variation in egg factors across different mouse strains. Our results support the importance of egg-intrinsic properties in determining Ca2+ oscillation patterns and have important implications for the interpretation and comparison of studies on Ca2+ dynamics at fertilization.
Collapse
Affiliation(s)
- Caitlin E McDonough
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Miranda L Bernhardt
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Carmen J Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
18
|
Wan L, Wu W, Jiang S, Wan S, Meng D, Wang Z, Zhang J, Wei L, Yu P. Mibefradil and Flunarizine, Two T-Type Calcium Channel Inhibitors, Protect Mice against Lipopolysaccharide-Induced Acute Lung Injury. Mediators Inflamm 2020; 2020:3691701. [PMID: 33223955 PMCID: PMC7671802 DOI: 10.1155/2020/3691701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/04/2020] [Accepted: 10/21/2020] [Indexed: 12/28/2022] Open
Abstract
Recent studies have illuminated that blocking Ca2+ influx into effector cells is an attractive therapeutic strategy for lung injury. We hypothesize that T-type calcium channel may be a potential therapeutic target for acute lung injury (ALI). In this study, the pharmacological activity of mibefradil (a classical T-type calcium channel inhibitor) was assessed in a mouse model of lipopolysaccharide- (LPS-) induced ALI. In LPS challenged mice, mibefradil (20 and 40 mg/kg) dramatically decreased the total cell number, as well as the productions of TNF-α and IL-6 in bronchoalveolar lavage fluid (BALF). Mibefradil also suppressed total protein concentration in BALF, attenuated Evans blue extravasation, MPO activity, and NF-κB activation in lung tissue. Furthermore, flunarizine, a widely prescripted antimigraine agent with potent inhibition on T-type channel, was also found to protect mice against lung injury. These data demonstrated that T-type calcium channel inhibitors may be beneficial for treating acute lung injury. The important role of T-type calcium channel in the acute lung injury is encouraged to be further investigated.
Collapse
Affiliation(s)
- Limei Wan
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Weibin Wu
- Department of Basic Medicine, Zhaoqing Medical College, Zhaoqing 526020, China
| | - Shunjun Jiang
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Shanhe Wan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, China
| | - Dongmei Meng
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Zhipeng Wang
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jiajie Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, China
| | - Li Wei
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Pengjiu Yu
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
19
|
Wakai T, Mehregan A, Fissore RA. Ca 2+ Signaling and Homeostasis in Mammalian Oocytes and Eggs. Cold Spring Harb Perspect Biol 2019; 11:a035162. [PMID: 31427376 PMCID: PMC6886447 DOI: 10.1101/cshperspect.a035162] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes in the intracellular concentration of calcium ([Ca2+]i) represent a vital signaling mechanism enabling communication between and among cells as well as with the environment. Cells have developed a sophisticated set of molecules, "the Ca2+ toolkit," to adapt [Ca2+]i changes to specific cellular functions. Mammalian oocytes and eggs, the subject of this review, are not an exception, and in fact the initiation of embryo devolvement in all species is entirely dependent on distinct [Ca2+]i responses. Here, we review the components of the Ca2+ toolkit present in mammalian oocytes and eggs, the regulatory mechanisms that allow these cells to accumulate Ca2+ in the endoplasmic reticulum, release it, and maintain basal and stable cytoplasmic concentrations. We also discuss electrophysiological and genetic studies that have uncovered Ca2+ influx channels in oocytes and eggs, and we analyze evidence supporting the role of a sperm-specific phospholipase C isoform as the trigger of Ca2+ oscillations during mammalian fertilization including its implication in fertility.
Collapse
Affiliation(s)
- Takuya Wakai
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| | - Aujan Mehregan
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| | - Rafael A Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
20
|
Yang R, Sun HH, Ji CL, Zhang J, Yuan HJ, Luo MJ, Liu XY, Tan JH. Role of calcium-sensing receptor in regulating spontaneous activation of postovulatory aging rat oocytes. Biol Reprod 2019; 98:218-226. [PMID: 29267849 DOI: 10.1093/biolre/iox178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/17/2017] [Indexed: 11/14/2022] Open
Abstract
Mechanisms for postovulatory aging (POA) of oocytes and for spontaneous activation (SA) of rat oocytes are largely unknown. Expression of calcium-sensing receptor (CaSR) in rat oocytes and its role in POA remain unexplored. In this study, expression of CaSR in rat oocytes aging for different times was detected by immunofluorescence microscopy, and western blotting and the role of CaSR in POA was determined by observing the effects of regulating its activity on SA susceptibility and cytoplasmic calcium levels. The results showed that CaSR was expressed in rat oocytes. Oocytes recovered 19 h post human chorionic gonadotropin (hCG) injection were more susceptible to SA and expressed more functional CaSR than oocytes recovered 13 h after hCG injection, although both expressed the same level of total CaSR protein. Treatment with CaSR antagonist significantly suppressed cytoplasmic calcium elevation and SA of oocytes. Activation of Na-Ca2+ exchanger with NaCl inhibited SA to a greater extent than suppression of CaSR with NPS-2143, suggesting that calcium sources other than CaSR-controlled channels contributed to the elevation of cytoplasmic calcium. Treatment with T- or L-type calcium channel blockers significantly reduced SA. Suppression of all calcium channels tested reduced SA to minimum. It is concluded that the level of CaSR functional dimer protein, but not that of the total CaSR protein, was positively correlated with the SA susceptibility during POA of rat oocytes confirming that CaSR is involved in POA regulation. Blocking multiple calcium channels might be a better choice for efficient control of SA in rat oocytes.
Collapse
Affiliation(s)
- Rui Yang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, P. R. China
| | - Huan-Huan Sun
- College of Life Science, Northeast Agricultural University, Harbin, P. R. China
| | - Chang-Li Ji
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, P. R. China
| | - Jie Zhang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, P. R. China
| | - Hong-Jie Yuan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, P. R. China
| | - Ming-Jiu Luo
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, P. R. China
| | - Xin-Yong Liu
- Center for Reproductive Medicine, Tai'an City Central Hospital, Tai'an City, P. R. China
| | - Jing-He Tan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, P. R. China
| |
Collapse
|
21
|
Wakai T, Fissore RA. Constitutive IP 3R1-mediated Ca 2+ release reduces Ca 2+ store content and stimulates mitochondrial metabolism in mouse GV oocytes. J Cell Sci 2019; 132:jcs.225441. [PMID: 30659110 DOI: 10.1242/jcs.225441] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/02/2019] [Indexed: 12/23/2022] Open
Abstract
In mammals, fertilization initiates Ca2+ oscillations in metaphase II oocytes, which are required for the activation of embryo development. Germinal vesicle (GV) oocytes also display Ca2+ oscillations, although these unfold spontaneously in the absence of any known agonist(s) and their function remains unclear. We found that the main intracellular store of Ca2+ in GV oocytes, the endoplasmic reticulum ([Ca2+]ER), constitutively 'leaks' Ca2+ through the type 1 inositol 1,4,5-trisphosphate receptor. The [Ca2+]ER leak ceases around the resumption of meiosis, the GV breakdown (GVBD) stage, which coincides with the first noticeable accumulation of Ca2+ in the stores. It also concurs with downregulation of the Ca2+ influx and termination of the oscillations, which seemed underpinned by the inactivation of the putative plasma membrane Ca2+ channels. Lastly, we demonstrate that mitochondria take up Ca2+ during the Ca2+ oscillations, mounting their own oscillations that stimulate the mitochondrial redox state and increase the ATP levels of GV oocytes. These distinct features of Ca2+ homeostasis in GV oocytes are likely to underpin the acquisition of both maturation and developmental competence, as well as fulfill stage-specific cellular functions during oocyte maturation.
Collapse
Affiliation(s)
- Takuya Wakai
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA 01003, USA
| | - Rafael A Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|
22
|
Paudel S, Sindelar R, Saha M. Calcium Signaling in Vertebrate Development and Its Role in Disease. Int J Mol Sci 2018; 19:E3390. [PMID: 30380695 PMCID: PMC6274931 DOI: 10.3390/ijms19113390] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/18/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence over the past three decades suggests that altered calcium signaling during development may be a major driving force for adult pathophysiological events. Well over a hundred human genes encode proteins that are specifically dedicated to calcium homeostasis and calcium signaling, and the majority of these are expressed during embryonic development. Recent advances in molecular techniques have identified impaired calcium signaling during development due to either mutations or dysregulation of these proteins. This impaired signaling has been implicated in various human diseases ranging from cardiac malformations to epilepsy. Although the molecular basis of these and other diseases have been well studied in adult systems, the potential developmental origins of such diseases are less well characterized. In this review, we will discuss the recent evidence that examines different patterns of calcium activity during early development, as well as potential medical conditions associated with its dysregulation. Studies performed using various model organisms, including zebrafish, Xenopus, and mouse, have underscored the critical role of calcium activity in infertility, abortive pregnancy, developmental defects, and a range of diseases which manifest later in life. Understanding the underlying mechanisms by which calcium regulates these diverse developmental processes remains a challenge; however, this knowledge will potentially enable calcium signaling to be used as a therapeutic target in regenerative and personalized medicine.
Collapse
Affiliation(s)
- Sudip Paudel
- College of William and Mary, Williamsburg, VA 23187, USA.
| | - Regan Sindelar
- College of William and Mary, Williamsburg, VA 23187, USA.
| | - Margaret Saha
- College of William and Mary, Williamsburg, VA 23187, USA.
| |
Collapse
|
23
|
TRPM7 and Ca V3.2 channels mediate Ca 2+ influx required for egg activation at fertilization. Proc Natl Acad Sci U S A 2018; 115:E10370-E10378. [PMID: 30322909 DOI: 10.1073/pnas.1810422115] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The success of mammalian development following fertilization depends on a series of transient increases in egg cytoplasmic Ca2+, referred to as Ca2+ oscillations. Maintenance of these oscillations requires Ca2+ influx across the plasma membrane, which is mediated in part by T-type, CaV3.2 channels. Here we show using genetic mouse models that TRPM7 channels are required to support this Ca2+ influx. Eggs lacking both TRPM7 and CaV3.2 stop oscillating prematurely, indicating that together they are responsible for the majority of Ca2+ influx immediately following fertilization. Fertilized eggs lacking both channels also frequently display delayed resumption of Ca2+ oscillations, which appears to require sperm-egg fusion. TRPM7 and CaV3.2 channels almost completely account for Ca2+ influx observed following store depletion, a process previously attributed to canonical store-operated Ca2+ entry mediated by STIM/ORAI interactions. TRPM7 serves as a membrane sensor of extracellular Mg2+ and Ca2+ concentrations and mediates the effects of these ions on Ca2+ oscillation frequency. When bred to wild-type males, female mice carrying eggs lacking TRPM7 and CaV3.2 are subfertile, and their offspring have increased variance in postnatal weight. These in vivo findings confirm previous observations linking in vitro experimental alterations in Ca2+ oscillatory patterns with developmental potential and offspring growth. The identification of TRPM7 and CaV3.2 as key mediators of Ca2+ influx following fertilization provides a mechanistic basis for the rational design of culture media that optimize developmental potential in research animals, domestic animals, and humans.
Collapse
|
24
|
Carvacho I, Piesche M, Maier TJ, Machaca K. Ion Channel Function During Oocyte Maturation and Fertilization. Front Cell Dev Biol 2018; 6:63. [PMID: 29998105 PMCID: PMC6028574 DOI: 10.3389/fcell.2018.00063] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/04/2018] [Indexed: 12/20/2022] Open
Abstract
The proper maturation of both male and female gametes is essential for supporting fertilization and the early embryonic divisions. In the ovary, immature fully-grown oocytes that are arrested in prophase I of meiosis I are not able to support fertilization. Acquiring fertilization competence requires resumption of meiosis which encompasses the remodeling of multiple signaling pathways and the reorganization of cellular organelles. Collectively, this differentiation endows the egg with the ability to activate at fertilization and to promote the egg-to-embryo transition. Oocyte maturation is associated with changes in the electrical properties of the plasma membrane and alterations in the function and distribution of ion channels. Therefore, variations on the pattern of expression, distribution, and function of ion channels and transporters during oocyte maturation are fundamental to reproductive success. Ion channels and transporters are important in regulating membrane potential, but also in the case of calcium (Ca2+), they play a critical role in modulating intracellular signaling pathways. In the context of fertilization, Ca2+ has been shown to be the universal activator of development at fertilization, playing a central role in early events associated with egg activation and the egg-to-embryo transition. These early events include the block of polyspermy, the completion of meiosis and the transition to the embryonic mitotic divisions. In this review, we discuss the role of ion channels during oocyte maturation, fertilization and early embryonic development. We will describe how ion channel studies in Xenopus oocytes, an extensively studied model of oocyte maturation, translate into a greater understanding of the role of ion channels in mammalian oocyte physiology.
Collapse
Affiliation(s)
- Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile
| | - Matthias Piesche
- Biomedical Research Laboratories, Medicine Faculty, Universidad Católica del Maule, Talca, Chile
| | - Thorsten J. Maier
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt, Germany
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell-Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| |
Collapse
|
25
|
Zhang L, Chao CH, Jaeger LA, Papp AB, Machaty Z. Calcium oscillations in fertilized pig oocytes are associated with repetitive interactions between STIM1 and ORAI1. Biol Reprod 2018; 98:510-519. [PMID: 29365044 PMCID: PMC5905661 DOI: 10.1093/biolre/ioy016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/01/2017] [Accepted: 01/19/2018] [Indexed: 11/15/2022] Open
Abstract
The Ca2+ entry mechanism that sustains the Ca2+ oscillations in fertilized pig oocytes was investigated. Stromal interaction molecule 1 (STIM1) and ORAI1 proteins tagged with various fluorophores were expressed in the oocytes. In some cells, the Ca2+ stores were depleted using cyclopiazonic acid (CPA); others were inseminated. Changes in the oocytes' cytosolic free Ca2+ concentration were monitored, while interaction between the expressed fusion proteins was investigated using fluorescence resonance energy transfer (FRET). Store depletion led to an increase of the FRET signal in oocytes co-expressing mVenus-STIM1 and mTurquoise2-ORAI1, indicating that Ca2+ release was followed by an interaction between these proteins. A similar FRET increase in response to CPA was also detected in oocytes co-expressing mVenus-STIM1 and mTurquoise2-STIM1, which is consistent with STIM1 forming punctae after store depletion. ML-9, an inhibitor that can interfere with STIM1 puncta formation, blocked store-operated Ca2+ entry (SOCE) induced by Ca2+ add-back after a CPA treatment; it also disrupted the Ca2+ oscillations in fertilized oocytes. In addition, oocytes overexpressing mVenus-STIM1 showed high-frequency Ca2+ oscillations when fertilized, arguing for an active role of the protein. High-frequency Ca2+ oscillations were also detected in fertilized oocytes co-expressing mVenus-STIM1 and mTurquoise2-ORAI1, and both of these high-frequency Ca2+ oscillations could be stopped by inhibitors of SOCE. Importantly, in oocytes co-expressing mVenus-STIM1 and mTurquoise2-ORAI1, we were also able to detect cyclic increases of the FRET signal indicating repetitive interactions between STIM1 and ORAI1. The results confirm the notion that in pig oocytes, SOCE is involved in the maintenance of the repetitive Ca2+ transients at fertilization.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | | | - Laurie A Jaeger
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Agnes Bali Papp
- Department of Animal Sciences, Széchenyi István University, Győr, Hungary
| | - Zoltan Machaty
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
26
|
Xu YR, Yang WX. Calcium influx and sperm-evoked calcium responses during oocyte maturation and egg activation. Oncotarget 2017; 8:89375-89390. [PMID: 29179526 PMCID: PMC5687696 DOI: 10.18632/oncotarget.19679] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/19/2017] [Indexed: 12/18/2022] Open
Abstract
Under the guidance and regulation of hormone signaling, large majority of mammalian oocytes go through twice cell cycle arrest-resumption prior to the fertilized egg splits: oocyte maturation and egg activation. Cytosolic free calcium elevations and endoplasmic reticulum calcium store alternations are actively involved in triggering the complex machineries and events during oogenesis. Among these, calcium influx had been implicated in the replenishment of endoplasmic reticulum store during oocyte maturation and calcium oscillation during egg activation. This process also drove successful fertilization and early embryo development. Store-operated Ca2+ entry, acts as the principal force of calcium influx, is composed of STIM1 and Orai1 on the plasma membrane. Besides, transient receptor potential channels also participate in the process of calcium inwards. In this review, we summarize the recent researches on the spatial-temporal distribution of store-operated calcium entry components and transient receptor potential channels. Questions about how these channels play function for calcium influx and what impacts these channels have on oocytes are discussed. At the time of sperm-egg fusion, sperm-specific factor(s) diffuse and enable eggs to mount intracellular calcium oscillations. In this review, we also focus on the basic knowledge and the modes of action of the potential sperm factor phospholipase C zeta, as well as the downstream receptor, type 1 inositol 1,4,5-trisphosphate receptor. From the achievement in the previous several decades, it is easy to find that there are too many doubtful points in the field that need researchers take into consideration and take action in the future.
Collapse
Affiliation(s)
- Ya-Ru Xu
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
Bernhardt ML, Padilla-Banks E, Stein P, Zhang Y, Williams CJ. Store-operated Ca 2+ entry is not required for fertilization-induced Ca 2+ signaling in mouse eggs. Cell Calcium 2017; 65:63-72. [PMID: 28222911 DOI: 10.1016/j.ceca.2017.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 01/01/2023]
Abstract
Repetitive oscillations in cytoplasmic Ca2+ due to periodic Ca2+ release from the endoplasmic reticulum (ER) drive mammalian embryo development following fertilization. Influx of extracellular Ca2+ to support the refilling of ER stores is required for sustained Ca2+ oscillations, but the mechanisms underlying this Ca2+ influx are controversial. Although store-operated Ca2+ entry (SOCE) is an appealing candidate mechanism, several groups have arrived at contradictory conclusions regarding the importance of SOCE in oocytes and eggs. To definitively address this question, Ca2+ influx was assessed in oocytes and eggs lacking the major components of SOCE, the ER Ca2+ sensor STIM proteins, and the plasma membrane Ca2+ channel ORAI1. We generated oocyte-specific conditional knockout (cKO) mice for Stim1 and Stim2, and also generated Stim1/2 double cKO mice. Females lacking one or both STIM proteins were fertile and their ovulated eggs displayed normal patterns of Ca2+ oscillations following fertilization. In addition, no impairment was observed in ER Ca2+ stores or Ca2+ influx following store depletion. Similar studies were performed on eggs from mice globally lacking ORAI1; no abnormalities were observed. Furthermore, spontaneous Ca2+ influx was normal in oocytes from Stim1/2 cKO and ORAI1-null mice. Finally, we tested if TRPM7-like channels could support spontaneous Ca2+ influx, and found that it was largely prevented by NS8593, a TRPM7-specific inhibitor. Fertilization-induced Ca2+ oscillations were also impaired by NS8593. Combined, these data robustly show that SOCE is not required to support appropriate Ca2+ signaling in mouse oocytes and eggs, and that TRPM7-like channels may contribute to Ca2+ influx that was previously attributed to SOCE.
Collapse
Affiliation(s)
- Miranda L Bernhardt
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Elizabeth Padilla-Banks
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Paula Stein
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Yingpei Zhang
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Carmen J Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
28
|
Fertility: Store-Operated Ca 2+ Entry in Germ Cells: Role in Egg Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:577-593. [PMID: 28900934 DOI: 10.1007/978-3-319-57732-6_29] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
At the time of fertilization, the sperm activates the egg and induces embryonic development by triggering an elevation in the egg's intracellular free Ca2+ concentration. In mammals the initial Ca2+ rise is followed by a series of repetitive Ca2+ transients (known as oscillations) that last for several hours. Although the source of Ca2+ during the signaling process is primarily the egg's smooth endoplasmic reticulum, the oscillations stop in the absence of extracellular Ca2+ indicating that a Ca2+ influx across the plasma membrane is essential to sustain them. Depletion of the intracellular stores using specific inhibitors generates a Ca2+ entry across the plasma membrane of eggs of various species, and a continuous influx of Ca2+ has been linked to the sperm-induced Ca2+ oscillations in the mouse; these data indicate that store-operated Ca2+ entry (SOCE) operates in eggs and may be the mechanism that maintains the long-lasting Ca2+ signal at fertilization. Recent findings suggest that the signaling proteins STIM1 and Orai1 are present in eggs; they are responsible for mediating SOCE, and their functions are essential for proper Ca2+ signaling at fertilization to support normal embryo development.
Collapse
|
29
|
Carvacho I, Ardestani G, Lee HC, McGarvey K, Fissore RA, Lykke-Hartmann K. TRPM7-like channels are functionally expressed in oocytes and modulate post-fertilization embryo development in mouse. Sci Rep 2016; 6:34236. [PMID: 27681336 PMCID: PMC5041074 DOI: 10.1038/srep34236] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/05/2016] [Indexed: 01/16/2023] Open
Abstract
The Transient Receptor Potential (TRP) channels are a family of cationic ion channels widely distributed in mammalian tissues. In general, the global genetic disruption of individual TRP channels result in phenotypes associated with impairment of a particular tissue and/or organ function. An exception is the genetic ablation of the TRP channel TRPM7, which results in early embryonic lethality. Nevertheless, the function of TRPM7 in oocytes, eggs and pre-implantation embryos remains unknown. Here, we described an outward rectifying non-selective current mediated by a TRP ion channel in immature oocytes (germinal vesicle stage), matured oocytes (metaphase II eggs) and 2-cell stage embryos. The current is activated by specific agonists and inhibited by distinct blockers consistent with the functional expression of TRPM7 channels. We demonstrated that the TRPM7-like channels are homo-tetramers and their activation mediates calcium influx in oocytes and eggs, which is fundamental to support fertilization and egg activation. Lastly, we showed that pharmacological inhibition of the channel function delays pre-implantation embryo development and reduces progression to the blastocyst stage. Our data demonstrate functional expression of TRPM7-like channels in mouse oocytes, eggs and embryos that may play an essential role in the initiation of embryo development.
Collapse
Affiliation(s)
- Ingrid Carvacho
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark.,Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Danish National Research Foundation, Aarhus University, Department of Molecular Biology and Genetics, DK-8000 Aarhus C, Denmark.,Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, 3480112 Talca, Chile
| | - Goli Ardestani
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Hoi Chang Lee
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Kaitlyn McGarvey
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Rafael A Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark.,Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Danish National Research Foundation, Aarhus University, Department of Molecular Biology and Genetics, DK-8000 Aarhus C, Denmark.,Aarhus Institute of Advanced Studies (AIAS), Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
30
|
Tosti E, Ménézo Y. Gamete activation: basic knowledge and clinical applications. Hum Reprod Update 2016; 22:420-39. [PMID: 27278231 PMCID: PMC4917743 DOI: 10.1093/humupd/dmw014] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 04/01/2016] [Indexed: 01/07/2023] Open
Abstract
Background The first clues to the process of gamete activation date back to nearly 60 years ago. The mutual activation of gametes is a crucial event during fertilization. In the testis and ovaries, spermatozoa and oocytes are in a state of meiotic and metabolic quiescence and require reciprocal signals in order to undergo functional changes that lead to competence for fertilization. First, the oocyte activates sperm by triggering motility, chemoattraction, binding and the acrosome reaction, culminating with the fusion of the two plasma membranes. At the end of this cascade of events, collectively known as sperm capacitation, sperm-induced oocyte activation occurs, generating electrical, morphological and metabolic modifications in the oocyte. Objective and rationale The aim of this review is to provide the current state of knowledge regarding the entire process of gamete activation in selected specific animal models that have contributed to our understanding of fertilization in mammals, including humans. Here we describe in detail the reciprocal induction of the two activation processes, the molecules involved and the mechanisms of cell interaction and signal transduction that ultimately result in successful embryo development and creation of a new individual. Search methods We carried out a literature survey with no restrictions on publication date (from the early 1950s to March 2016) using PubMed/Medline, Google Scholar and Web of Knowledge by utilizing common keywords applied in the field of fertilization and embryo development. We also screened the complete list of references published in the most recent research articles and relevant reviews published in English (both animal and human studies) on the topics investigated. Outcomes Literature on the principal animal models demonstrates that gamete activation is a pre-requisite for successful fertilization, and is a process common to all species studied to date. We provide a detailed description of the dramatic changes in gamete morphology and behavior, the regulatory molecules triggering gamete activation and the intracellular ions and second messengers involved in active metabolic pathways in different species. Recent scientific advances suggest that artificial gamete activation may represent a novel technique to improve human IVF outcomes, but this approach requires caution. Wider implications Although controversial, manipulation of gamete activation represents a promising tool for ameliorating the fertilization rate in assisted reproductive technologies. A better knowledge of mechanisms that transform the quiescent oocyte into a pluripotent cell may also provide new insights for the clinical use of stem cells.
Collapse
Affiliation(s)
- Elisabetta Tosti
- Stazione Zoologica Anton Dohrn, Villa Comunale, Naples 80121, Italy
| | - Yves Ménézo
- London Fertility Associates, 104 Harley Street, London WIG7JD, UK
| |
Collapse
|
31
|
Mackenzie ACL, Kyle DD, McGinnis LA, Lee HJ, Aldana N, Robinson DN, Evans JP. Cortical mechanics and myosin-II abnormalities associated with post-ovulatory aging: implications for functional defects in aged eggs. Mol Hum Reprod 2016; 22:397-409. [PMID: 26921397 PMCID: PMC4884917 DOI: 10.1093/molehr/gaw019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 02/12/2016] [Accepted: 02/24/2016] [Indexed: 12/20/2022] Open
Abstract
STUDY HYPOTHESIS Cellular aging of the egg following ovulation, also known as post-ovulatory aging, is associated with aberrant cortical mechanics and actomyosin cytoskeleton functions. STUDY FINDING Post-ovulatory aging is associated with dysfunction of non-muscle myosin-II, and pharmacologically induced myosin-II dysfunction produces some of the same deficiencies observed in aged eggs. WHAT IS KNOWN ALREADY Reproductive success is reduced with delayed fertilization and when copulation or insemination occurs at increased times after ovulation. Post-ovulatory aged eggs have several abnormalities in the plasma membrane and cortex, including reduced egg membrane receptivity to sperm, aberrant sperm-induced cortical remodeling and formation of fertilization cones at the site of sperm entry, and reduced ability to establish a membrane block to prevent polyspermic fertilization. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Ovulated mouse eggs were collected at 21-22 h post-human chorionic gonadotrophin (hCG) (aged eggs) or at 13-14 h post-hCG (young eggs), or young eggs were treated with the myosin light chain kinase (MLCK) inhibitor ML-7, to test the hypothesis that disruption of myosin-II function could mimic some of the effects of post-ovulatory aging. Eggs were subjected to various analyses. Cytoskeletal proteins in eggs and parthenogenesis were assessed using fluorescence microscopy, with further analysis of cytoskeletal proteins in immunoblotting experiments. Cortical tension was measured through micropipette aspiration assays. Egg membrane receptivity to sperm was assessed in in vitro fertilization (IVF) assays. Membrane topography was examined by low-vacuum scanning electron microscopy (SEM). MAIN RESULTS AND THE ROLE OF CHANCE Aged eggs have decreased levels and abnormal localizations of phosphorylated myosin-II regulatory light chain (pMRLC; P = 0.0062). Cortical tension, which is mediated in part by myosin-II, is reduced in aged mouse eggs when compared with young eggs, by ∼40% in the cortical region where the metaphase II spindle is sequestered and by ∼50% in the domain to which sperm bind and fuse (P < 0.0001). Aging-associated parthenogenesis is partly rescued by treating eggs with a zinc ionophore (P = 0.003), as is parthenogenesis induced by inhibition of mitogen-activated kinase (MAPK) 3/1 [also known as extracellular signal-regulated kinase (ERK)1/2] or MLCK. Inhibition of MLCK with ML-7 also results in effects that mimic those of post-ovulatory aging: fertilized ML-7-treated eggs show both impaired fertilization and increased extents of polyspermy, and ML-7-treated young eggs have several membrane abnormalities that are shared by post-ovulatory aged eggs. LIMITATIONS, REASONS FOR CAUTION These studies were done with mouse oocytes, and it remains to be fully determined how these findings from mouse oocytes would compare with other species. For studies using methods not amenable to analysis of large sample sizes and data are limited to what images one can capture (e.g. SEM), data should be interpreted conservatively. WIDER IMPLICATIONS OF THE FINDINGS These data provide insights into causes of reproductive failures at later post-copulatory times. LARGE SCALE DATA Not applicable. STUDY FUNDING AND COMPETING INTERESTS This project was supported by R01 HD037696 and R01 HD045671 from the NIH to J.P.E. Cortical tension studies were supported by R01 GM66817 to D.N.R. The authors declare there are no financial conflicts of interest.
Collapse
Affiliation(s)
- Amelia C L Mackenzie
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Diane D Kyle
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Lauren A McGinnis
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Hyo J Lee
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Nathalia Aldana
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Douglas N Robinson
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Janice P Evans
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| |
Collapse
|
32
|
Sanders JR, Swann K. Molecular triggers of egg activation at fertilization in mammals. Reproduction 2016; 152:R41-50. [PMID: 27165049 DOI: 10.1530/rep-16-0123] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/09/2016] [Indexed: 01/15/2023]
Abstract
In mammals, the sperm activates the development of the egg by triggering a series of oscillations in the cytosolic-free Ca(2+) concentration (Ca(2+) i). The sperm triggers these cytosolic Ca(2+i) oscillations after sperm-egg membrane fusion, as well as after intracytoplasmic sperm injection (ICSI). These Ca(2+) i oscillations are triggered by a protein located inside the sperm. The identity of the sperm protein has been debated over many years, but all the repeatable data now suggest that it is phospholipase Czeta (PLCζ). The main downstream target of Ca(2+) i oscillations is calmodulin-dependent protein kinase II (CAMKII (CAMK2A)), which phosphorylates EMI2 and WEE1B to inactivate the M-phase promoting factor protein kinase activity (MPF) and this ultimately triggers meiotic resumption. A later decline in the activity of mitogen-activated protein kinase (MAPK) then leads to the completion of activation which is marked by the formation of pronuclei and entry into interphase of the first cell cycle. The early cytosolic Ca(2+) increases also trigger exocytosis via a mechanism that does not involve CAMKII. We discuss some recent developments in our understanding of these triggers for egg activation within the framework of cytosolic Ca(2+) signaling.
Collapse
Affiliation(s)
| | - Karl Swann
- School of BiosciencesCardiff University, Cardiff, UK
| |
Collapse
|
33
|
Voisin T, Bourinet E, Lory P. Genetic alteration of the metal/redox modulation of Cav3.2 T-type calcium channel reveals its role in neuronal excitability. J Physiol 2016; 594:3561-74. [PMID: 26931411 DOI: 10.1113/jp271925] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/29/2016] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS In this study, we describe a new knock-in (KI) mouse model that allows the study of the H191-dependent regulation of T-type Cav3.2 channels. Sensitivity to zinc, nickel and ascorbate of native Cav3.2 channels is significantly impeded in the dorsal root ganglion (DRG) neurons of this KI mouse. Importantly, we describe that this H191-dependent regulation has discrete but significant effects on the excitability properties of D-hair (down-hair) cells, a sub-population of DRG neurons in which Cav3.2 currents prominently regulate excitability. Overall, this study reveals that the native H191-dependent regulation of Cav3.2 channels plays a role in the excitability of Cav3.2-expressing neurons. This animal model will be valuable in addressing the potential in vivo roles of the trace metal and redox modulation of Cav3.2 T-type channels in a wide range of physiological and pathological conditions. ABSTRACT Cav3.2 channels are T-type voltage-gated calcium channels that play important roles in controlling neuronal excitability, particularly in dorsal root ganglion (DRG) neurons where they are involved in touch and pain signalling. Cav3.2 channels are modulated by low concentrations of metal ions (nickel, zinc) and redox agents, which involves the histidine 191 (H191) in the channel's extracellular IS3-IS4 loop. It is hypothesized that this metal/redox modulation would contribute to the tuning of the excitability properties of DRG neurons. However, the precise role of this H191-dependent modulation of Cav3.2 channel remains unresolved. Towards this goal, we have generated a knock-in (KI) mouse carrying the mutation H191Q in the Cav3.2 protein. Electrophysiological studies were performed on a subpopulation of DRG neurons, the D-hair cells, which express large Cav3.2 currents. We describe an impaired sensitivity to zinc, nickel and ascorbate of the T-type current in D-hair neurons from KI mice. Analysis of the action potential and low-threshold calcium spike (LTCS) properties revealed that, contrary to that observed in WT D-hair neurons, a low concentration of zinc and nickel is unable to modulate (1) the rheobase threshold current, (2) the afterdepolarization amplitude, (3) the threshold potential necessary to trigger an LTCS or (4) the LTCS amplitude in D-hair neurons from KI mice. Together, our data demonstrate that this H191-dependent metal/redox regulation of Cav3.2 channels can tune neuronal excitability. This study validates the use of this Cav3.2-H191Q mouse model for further investigations of the physiological roles thought to rely on this Cav3.2 modulation.
Collapse
Affiliation(s)
- Tiphaine Voisin
- Centre National pour la Recherche Scientifique UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, F-34094, France.,Institut National de la Santé et de la Recherche Médicale, U 1191, Montpellier, F-34094, France.,LabEx 'Ion Channel Science and Therapeutics', Montpellier, F-34094, France
| | - Emmanuel Bourinet
- Centre National pour la Recherche Scientifique UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, F-34094, France.,Institut National de la Santé et de la Recherche Médicale, U 1191, Montpellier, F-34094, France.,LabEx 'Ion Channel Science and Therapeutics', Montpellier, F-34094, France
| | - Philippe Lory
- Centre National pour la Recherche Scientifique UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, F-34094, France.,Institut National de la Santé et de la Recherche Médicale, U 1191, Montpellier, F-34094, France.,LabEx 'Ion Channel Science and Therapeutics', Montpellier, F-34094, France
| |
Collapse
|
34
|
Bernhardt ML, Zhang Y, Erxleben CF, Padilla-Banks E, McDonough CE, Miao YL, Armstrong DL, Williams CJ. CaV3.2 T-type channels mediate Ca2+ entry during oocyte maturation and following fertilization. Development 2015. [DOI: 10.1242/dev.133629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|