Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Int J Mol Sci ; 24(21)2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37958614

ABSTRACT

Calmodulin (CaM) is a small, multifunctional calcium (Ca2+)-binding sensor that binds and regulates the open probability of cardiac ryanodine receptor 2 (RyR2) at both low and high cytosolic Ca2+ concentrations. Recent isothermal titration calorimetry (ITC) studies of a number of peptides that correspond to different regions of human RyR2 showed that two regions of human RyR2 (3584-3602aa and 4255-4271aa) bind with high affinity to CaM, suggesting that these two regions might contribute to a putative RyR2 intra-subunit CaM-binding pocket. Moreover, a previously characterized de novo long QT syndrome (LQTS)-associated missense CaM mutation (E105A) which was identified in a 6-year-old boy, who experienced an aborted first episode of cardiac arrest revealed that this mutation dysregulates normal cardiac function in zebrafish by a complex mechanism that involves alterations in both CaM-Ca2+ and CaM-RyR2 interactions. Herein, to gain further insight into how the CaM E105A mutation leads to severe cardiac arrhythmia, we generated large quantities of recombinant CaMWT and CaME105A proteins. We then performed ITC experiments to investigate and compare the interactions of CaMWT and CaME105A mutant protein with two synthetic peptides that correspond to the two aforementioned human RyR2 regions, which we have proposed to contribute to the RyR2 CaM-binding pocket. Our data reveal that the E105A mutation has a significant negative effect on the interaction of CaM with both RyR2 regions in the presence and absence of Ca2+, highlighting the potential contribution of these two human RyR2 regions to an RyR2 CaM-binding pocket, which may be essential for physiological CaM/RyR2 association and thus channel regulation.


Subject(s)
Calmodulin , Ryanodine Receptor Calcium Release Channel , Male , Animals , Humans , Child , Calmodulin/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Arrhythmias, Cardiac/genetics , Mutation , Calcium/metabolism
2.
Pharmaceuticals (Basel) ; 16(2)2023 Jan 28.
Article in English | MEDLINE | ID: mdl-37259347

ABSTRACT

Mammalian oocyte activation is initiated by intracellular calcium (Ca2+) oscillations, driven by the testis-specific phospholipase C zeta (PLCζ). Sperm PLCζ analysis represents a diagnostic measure of sperm fertilisation capacity. The application of antigen unmasking/retrieval (AUM) generally enhanced the visualisation efficacy of PLCζ in mammalian sperm, but differentially affected the PLCζ profiles in sperm from different human males. It is unclear whether AUM affects the diagnosis of PLCζ in human sperm. Herein, we examined whether the application of AUM affected the correlation of PLCζ profiles with sperm parameters and fertilisation capacity. PLCζ fluorescence levels and localisation patterns were examined within the sperm of males undergoing fertility treatment (55 patients aged 29-53) using immunofluorescence in the absence/presence of AUM. The changes in PLCζ profiles following AUM were examined in relation to sperm health and fertilisation outcome. AUM enhanced the observable levels and specific localisation patterns of PLCζ in relation to both optimal sperm parameters and fertilisation outcome, without which significant differences were not observed. The extent of the change in levels and localisation ratios of PLCζ was also affected to a larger degree in terms of the optimal parameters of sperm fertility and fertilisation capacity by AUM. Collectively, AUM was essential to accurately assesses PLCζ in human sperm in both scientific and clinical contexts.

3.
Biochim Biophys Acta Gen Subj ; 1867(4): 130313, 2023 04.
Article in English | MEDLINE | ID: mdl-36693454

ABSTRACT

Calmodulin (CaM) modulates the activity of several proteins that play a key role in excitation-contraction coupling (ECC). In cardiac muscle, the major binding partner of CaM is the type-2 ryanodine receptor (RyR2) and altered CaM binding contributes to defects in sarcoplasmic reticulum (SR) calcium (Ca2+) release. Many genetic studies have reported a series of CaM missense mutations in patients with a history of severe arrhythmogenic cardiac disorders. In the present study, we generated four missense CaM mutants (CaMN98I, CaMD132E, CaMD134H and CaMQ136P) and we used a CaM-RyR2 co-immunoprecipitation and a [3H]ryanodine binding assay to directly compare the relative RyR2-binding of wild type and mutant CaM proteins and to investigate the functional effects of these CaM mutations on RyR2 activity. Furthermore, isothermal titration calorimetry (ITC) experiments were performed to investigate and compare the interactions of the wild-type and mutant CaM proteins with various synthetic peptides located in the well-established RyR2 CaM-binding region (3584-3602aa), as well as another CaM-binding region (4255-4271aa) of human RyR2. Our data revealed that all four CaM mutants displayed dramatically reduced RyR2 interaction and defective modulation of [3H]ryanodine binding to RyR2, regardless of LQTS or CPVT association. Moreover, our isothermal titration calorimetry ITC data suggest that RyR2 3584-3602aa and 4255-4271aa regions interact with significant affinity with wild-type CaM, in the presence and absence of Ca2+, two regions that might contribute to a putative intra-subunit CaM-binding pocket. In contrast, screening the interaction of the four arrhythmogenic CaM mutants with two synthetic peptides that correspond to these RyR2 regions, revealed disparate binding properties and signifying differential mechanisms that contribute to reduced RyR2 association.


Subject(s)
Calmodulin , Ryanodine Receptor Calcium Release Channel , Humans , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Calcium Signaling , Calmodulin/chemistry , Mutation , Ryanodine , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism
4.
Front Physiol ; 13: 1032132, 2022.
Article in English | MEDLINE | ID: mdl-36311249

ABSTRACT

The ryanodine receptor (RyR) is a homotetrameric channel mediating sarcoplasmic reticulum Ca2+ release required for skeletal and cardiac muscle contraction. Mutations in RyR1 and RyR2 lead to life-threatening malignant hyperthermia episodes and ventricular tachycardia, respectively. In this brief report, we use chemical cross-linking to demonstrate that pathogenic RyR1 R163C and RyR2 R169Q mutations reduce N-terminus domain (NTD) tetramerization. Introduction of positively-charged residues (Q168R, M399R) in the NTD-NTD inter-subunit interface normalizes RyR2-R169Q NTD tetramerization. These results indicate that perturbation of NTD-NTD inter-subunit interactions is an underlying molecular mechanism in both RyR1 and RyR2 pathophysiology. Importantly, our data provide proof of concept that stabilization of this critical RyR1/2 structure-function parameter offers clear therapeutic potential.

6.
Reproduction ; 164(1): F1-F8, 2022 05 23.
Article in English | MEDLINE | ID: mdl-35521907

ABSTRACT

In 2002, sperm-specific phospholipase C zeta1 (PLCZ1) was discovered and through these 20 years, it has been established as the predominant sperm oocyte-activating factor. PLCZ1 cRNA expression or direct protein microinjection into mammalian oocytes triggers calcium (Ca2+) oscillations indistinguishable from those observed at fertilization. The imperative role of PLCZ1 in oocyte activation is revealed by the vast number of human mutations throughout the PLCZ1 gene that have been identified and directly linked with certain forms of male infertility due to oocyte activation deficiency. PLCZ1 is the smallest PLC in size, comprising four N-terminal EF-hand domains, followed by X and Y catalytic domains, which are separated by the XY-linker, and ending with a C-terminal C2 domain. The EF hands are responsible for the high Ca2+ sensitivity of PLCZ1. The X and Y catalytic domains are responsible for the catalysis of the phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2] substrate to produce the Ca2+-mobilising messenger, inositol 1,4,5-trisphosphate (IP3), while the XY-linker plays multiple roles in the unique mode of PLCZ1 action. Finally, the C2 domain has been proposed to facilitate the anchoring of PLCZ1 to intracellular vesicles through its direct interactions with specific phosphoinositides. This review discusses recent advances in the structure and function relationship of PLCZ1 and the potential binding partners of this important sperm-specific protein in the sperm and oocyte. The unravelling of all the remaining hidden secrets of sperm PLCZ1 should help us to understand the precise mechanism of fertilization, as well as enabling the diagnosis and treatment of currently unknown forms of PLCZ1 -linked human infertility.


Subject(s)
Calcium , Type C Phospholipases , Animals , Calcium/metabolism , Fertilization/physiology , Humans , Male , Mammals/metabolism , Oocytes , Phosphoinositide Phospholipase C/genetics , Phosphoinositide Phospholipase C/metabolism , Spermatozoa/metabolism , Type C Phospholipases/metabolism
8.
Asian J Androl ; 23(2): 178-187, 2021.
Article in English | MEDLINE | ID: mdl-33208563

ABSTRACT

Sperm-specific phospholipase C zeta (PLCζ) initiates intracellular calcium (Ca2+) transients which drive a series of concurrent events collectively termed oocyte activation. Numerous investigations have linked abrogation and absence/reduction of PLCζ with forms of male infertility in humans where oocyte activation fails. However, very few studies have examined potential relationships between PLCζ and advancing male age, both of which are increasingly considered to be major effectors of male fertility. Initial efforts in humans may be hindered by inherent PLCζ variability within the human population, alongside a lack of sufficient controllable repeats. Herein, utilizing immunoblotting, immunofluorescence, and quantitative reverse transcription PCR (qRT-PCR) we examined for the first time PLCζ protein levels and localization patterns in sperm, and PLCζ mRNA levels within testes, from mice at 8 weeks, 12 weeks, 24 weeks, and 36 weeks of age, from two separate strains of mice, C57BL/6 (B6; inbred) and CD1 (outbred). Collectively, advancing male age generally diminished levels and variability of PLCζ protein and mRNA in sperm and testes, respectively, when both strains were examined. Furthermore, advancing male age altered the predominant pattern of PLCζ localization in mouse sperm, with younger mice exhibiting predominantly post-acrosomal, and older mice exhibiting both post-acrosomal and acrosomal populations of PLCζ. However, the specific pattern of such decline in levels of protein and mRNA was strain-specific. Collectively, our results demonstrate a negative relationship between advancing male age and PLCζ levels and localization patterns, indicating that aging male mice from different strains may serve as useful models to investigate PLCζ in cases of male infertility and subfertility in humans.


Subject(s)
Aging/genetics , Phosphoinositide Phospholipase C/genetics , Spermatozoa/metabolism , Testis/metabolism , Aging/metabolism , Animals , Fluorescent Antibody Technique , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Phosphoinositide Phospholipase C/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
Andrology ; 8(5): 1143-1159, 2020 09.
Article in English | MEDLINE | ID: mdl-32298520

ABSTRACT

BACKGROUND: Oocyte activation is driven by intracellular calcium (Ca2+ ) oscillations induced by sperm-specific PLCζ, abrogation of which causes oocyte activation deficiency in humans. Clinical PLCζ investigations have been limited to severe male infertility conditions, while PLCζ levels and localisation patterns have yet to be associated with general sperm viability. MATERIALS AND METHODS: PLCζ profiles were examined within a general population of males attending a fertility clinic (65 patients; aged 29-53), examining PLCζ throughout various fractions of sperm viability. Male recruitment criteria required a minimum sperm count of 5 × 106 spermatozoa/mL, while all female patients included in this study yielded at least five oocytes for treatment. Sperm count, motility and semen volume were recorded according to standard WHO reference guidelines and correlated with PLCζ profiles examined via immunoblotting and immunofluorescence. Appropriate fertility treatments were performed following routine clinical standard operating protocols, and fertilisation success determined by successful observation of second polar body extrusion. RESULTS AND DISCUSSION: Four distinct PLCζ patterns were observed at the equatorial, acrosomal + equatorial regions of the sperm head, alongside a dispersed pattern, and a population of spermatozoa without any PLCζ. Acrosomal + equatorial PLCζ correlated most to sperm health, while dispersed PLCζ correlated to decreased sperm viability. Total levels of PLCζ exhibited significant correlations with sperm parameters. PLCζ variance corresponded to reduced sperm health, potentially underlying cases of male sub-fertility and increasing male age. Finally, significantly higher levels of PLCζ were exhibited by cases of fertilisation success, alongside higher proportions of Ac + Eq, and lower levels of dispersed PLCζ. CONCLUSIONS: PLCζ potentially represents a biomarker of sperm health, and fertilisation capacity in general cases of patients seeking fertility treatment, and not just cases of repeated fertilisation. Further focused investigations are required with larger cohorts to examine the full clinical potential of PLCζ.


Subject(s)
Fertilization , Infertility, Male/enzymology , Phosphoinositide Phospholipase C/metabolism , Spermatozoa/enzymology , Acrosome/enzymology , Adult , Cell Survival , Humans , Immunoblotting , Infertility, Male/diagnosis , Infertility, Male/therapy , Male , Middle Aged , Reproductive Techniques, Assisted
10.
Front Cell Dev Biol ; 8: 28, 2020.
Article in English | MEDLINE | ID: mdl-32064262

ABSTRACT

Sperm-specific phospholipase C zeta (PLCζ) is widely considered to be the physiological stimulus responsible for generating calcium (Ca2+) oscillations that induce egg activation and early embryonic development during mammalian fertilization. In the mammalian testis, PLCζ expression is detected at spermiogenesis following elongated spermatid differentiation. Sperm-delivered PLCζ induces Ca2+ release via the inositol 1,4,5-trisphosphate (InsP3) signaling pathway. PLCζ is the smallest known mammalian PLC isoform identified to date, with the simplest domain organization. However, the distinctive biochemical properties of PLCζ compared with other PLC isoforms contribute to its unique potency in stimulating cytosolic Ca2+ oscillations within mammalian eggs. Moreover, studies describing PLCζ "knockout" mouse phenotypes confirm the supreme importance of PLCζ at egg activation and monospermic fertilization in mice. Importantly, a number of clinical reports have highlighted the crucial importance of PLCζ in human fertilization by associating PLCζ deficiencies with certain forms of male factor infertility. Herein, we give an update on recent advances that have refined our understanding of how sperm PLCζ triggers Ca2 + oscillations and egg activation in mammals, while also discussing the nature of a potential "alternative" sperm factor. We summarise PLCζ localization in mammalian sperm, and the direct links observed between defective PLCζ protein in sperm and documented cases of male infertility. Finally, we postulate how this sperm protein can be used as a potential diagnostic marker, and also as a powerful therapeutic agent for treatment of certain types of male infertility due to egg activation failure or even in more general cases of male subfertility.

11.
Ann N Y Acad Sci ; 1448(1): 19-29, 2019 07.
Article in English | MEDLINE | ID: mdl-30937913

ABSTRACT

Calmodulin (CaM) is a universal calcium (Ca2+ )-binding messenger that regulates many vital cellular events. In cardiac muscle, CaM associates with ryanodine receptor 2 (RyR2) and regulates excitation-contraction coupling. Mutations in human genes CALM1, CALM2, and CALM3 have been associated with life-threatening heart disorders, such as long QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia. A novel de novo LQTS-associated missense CaM mutation (E105A) was recently identified in a 6-year-old boy, who experienced an aborted first episode of cardiac arrest. Herein, we report the first molecular characterization of the CaM E105A mutation. Expression of the CaM E105A mutant in zebrafish embryos resulted in cardiac arrhythmia and increased heart rate, suggestive of ventricular tachycardia. In vitro biophysical and biochemical analysis revealed that E105A confers a deleterious effect on protein stability and a reduced Ca2+ -binding affinity due to loss of cooperativity. Finally, the CaM E105A mutation resulted in reduced CaM-RyR2 interaction and defective modulation of ryanodine binding. Our findings suggest that the CaM E105A mutation dysregulates normal cardiac function by a complex mechanism involving alterations in both CaM-Ca2+ and CaM-RyR2 interactions.


Subject(s)
Arrhythmias, Cardiac/genetics , Calmodulin/genetics , Calmodulin/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/genetics , Animals , Arrhythmias, Cardiac/pathology , Calcium/metabolism , Calcium Signaling/physiology , Child , Excitation Contraction Coupling/physiology , Heart Rate/genetics , Heart Rate/physiology , Humans , Male , Myocytes, Cardiac/metabolism , Tachycardia, Ventricular/physiopathology , Zebrafish
12.
Biochem J ; 475(24): 3933-3948, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30446606

ABSTRACT

The most common inherited cardiac disorder, hypertrophic cardiomyopathy (HCM), is characterized by thickening of heart muscle, for which genetic mutations in cardiac myosin-binding protein C3 (c-MYBPC3) gene, is the leading cause. Notably, patients with HCM display a heterogeneous clinical presentation, onset and prognosis. Thus, delineating the molecular mechanisms that explain how disparate c-MYBPC3 variants lead to HCM is essential for correlating the impact of specific genotypes on clinical severity. Herein, five c-MYBPC3 missense variants clinically associated with HCM were investigated; namely V1 (R177H), V2 (A216T), V3 (E258K), V4 (E441K) and double mutation V5 (V3 + V4), all located within the C1 and C2 domains of MyBP-C, a region known to interact with sarcomeric protein, actin. Injection of the variant complementary RNAs in zebrafish embryos was observed to recapitulate phenotypic aspects of HCM in patients. Interestingly, V3- and V5-cRNA injection produced the most severe zebrafish cardiac phenotype, exhibiting increased diastolic/systolic myocardial thickness and significantly reduced heart rate compared with control zebrafish. Molecular analysis of recombinant C0-C2 protein fragments revealed that c-MYBPC3 variants alter the C0-C2 domain secondary structure, thermodynamic stability and importantly, result in a reduced binding affinity to cardiac actin. V5 (double mutant), displayed the greatest protein instability with concomitant loss of actin-binding function. Our study provides specific mechanistic insight into how c-MYBPC3 pathogenic variants alter both functional and structural characteristics of C0-C2 domains leading to impaired actin interaction and reduced contractility, which may provide a basis for elucidating the disease mechanism in HCM patients with c-MYBPC3 mutations.


Subject(s)
Actins/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Carrier Proteins/metabolism , Genetic Variation/physiology , Mutation, Missense/physiology , Actins/genetics , Adult , Animals , Cardiomyopathy, Hypertrophic/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Humans , Protein Binding/physiology , Protein Structure, Secondary , Zebrafish
13.
J Cell Sci ; 131(15)2018 08 03.
Article in English | MEDLINE | ID: mdl-29930088

ABSTRACT

The cardiac muscle ryanodine receptor-Ca2+ release channel (RyR2) constitutes the sarcoplasmic reticulum (SR) Ca2+ efflux mechanism that initiates myocyte contraction, while cardiac myosin-binding protein-C (cMyBP-C; also known as MYBPC3) mediates regulation of acto-myosin cross-bridge cycling. In this paper, we provide the first evidence for the presence of direct interaction between these two proteins, forming a RyR2-cMyBP-C complex. The C-terminus of cMyBP-C binds with the RyR2 N-terminus in mammalian cells and the interaction is not mediated by a fibronectin-like domain. Notably, we detected complex formation between both recombinant cMyBP-C and RyR2, as well as between the native proteins in cardiac tissue. Cellular Ca2+ dynamics in HEK293 cells is altered upon co-expression of cMyBP-C and RyR2, with lowered frequency of RyR2-mediated spontaneous Ca2+ oscillations, suggesting that cMyBP-C exerts a potential inhibitory effect on RyR2-dependent Ca2+ release. Discovery of a functional RyR2 association with cMyBP-C provides direct evidence for a putative mechanistic link between cytosolic soluble cMyBP-C and SR-mediated Ca2+ release, via RyR2. Importantly, this interaction may have clinical relevance to the observed cMyBP-C and RyR2 dysfunction in cardiac pathologies, such as hypertrophic cardiomyopathy.


Subject(s)
Carrier Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Cytosol/metabolism , HEK293 Cells , Humans , Protein Binding , Sarcoplasmic Reticulum/metabolism
14.
Reprod Biomed Online ; 36(3): 348-355, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29339016

ABSTRACT

Globozoospermia is characterized by the presence of 100% acrosomeless round-headed spermatozoa in an ejaculate. Failed fertilization after intracytoplasmic sperm injection (ICSI) is commonly reported for globozoospermic couples and can be overcome by artificial oocyte activation (AOA). Phospholipase C zeta (PLCζ) is one of the main sperm factors involved in oocyte activation and its low expression levels mainly account for fertilization failure. Deletion of the DPY19L2 gene is reported as a main genetic cause in over 70% of infertile men with globozoospermia. The current study assesses the expression profile of sperm PLCζ at RNA and protein levels in 32 DPY19L2 deletion-mediated globozoospermic men and reports corresponding clinical outcomes following ICSI with AOA. The expression of PLCζ relative to GAPDH at RNA (0.78 ± 0.16 versus 1.65 ± 0.24; P = 0.02) and protein (0.39 ± 0.12 versus 0.83 ± 0.13; P = 0.01) levels in globozoospermic men with DPY19L2 deletion was significantly lower compared with fertile men (n = 32). Fertilization rate in globozoospermic couples following ICSI-AOA was significantly lower compared with fertile men (53.14 ± 5.13% versus 87.64 ± 2.38%, P < 0.001). However, implantation (26.2%) and pregnancy (53.8%) rates were not jeopardized by DPY19L2 deletion in these couples.


Subject(s)
Gene Deletion , Membrane Proteins/genetics , Ovulation Induction/methods , Phosphoinositide Phospholipase C/metabolism , Sperm Injections, Intracytoplasmic/methods , Spermatozoa/metabolism , Teratozoospermia/pathology , Adult , Case-Control Studies , Female , Fertilization , Gene Expression Regulation , Humans , Male , Oocytes , Phosphoinositide Phospholipase C/genetics , Pregnancy , Sperm Motility , Teratozoospermia/genetics , Teratozoospermia/metabolism
15.
Adv Biol Regul ; 67: 148-162, 2018 01.
Article in English | MEDLINE | ID: mdl-29108881

ABSTRACT

Oocyte activation is a fundamental event at mammalian fertilisation, initiated by a series of characteristic calcium (Ca2+) oscillations in mammals. This characteristic pattern of Ca2+ release is induced in a species-specific manner by a sperm-specific enzyme termed phospholipase C zeta (PLCζ). Reduction or absence of functional PLCζ within sperm underlies male factor infertility in humans, due to mutational inactivation or abrogation of PLCζ protein expression. Underlying such clinical implications, a significant body of evidence has now been accumulated that has characterised the unique biochemical and biophysical properties of this enzyme, further aiding the unique clinical opportunities presented. Herein, we present and discuss evidence accrued over the past decade and a half that serves to support the identity of PLCζ as the mammalian sperm factor. Furthermore, we also discuss the potential novel avenues that have yet to be examined regarding PLCζ mechanism of action in both the oocyte, and the sperm. Finally, we discuss the advances that have been made regarding the clinical therapeutic and diagnostic applications of PLCζ in potentially treating male infertility as a result of oocyte activation deficiency (OAD), and also possibly more general cases of male subfertility.


Subject(s)
Calcium Signaling , Calcium/metabolism , Fertilization , Oocytes/enzymology , Phosphoinositide Phospholipase C/metabolism , Spermatozoa/enzymology , Animals , Female , Humans , Infertility, Male/enzymology , Infertility, Male/genetics , Male , Phosphoinositide Phospholipase C/genetics
16.
Biochem J ; 474(21): 3659-3673, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29061915

ABSTRACT

At mammalian fertilisation, the fundamental stimulus that triggers oocyte (egg) activation and initiation of early embryonic development is an acute rise of the intracellular-free calcium (Ca2+) concentration inside the egg cytoplasm. This essential Ca2+ increase comprises a characteristic series of repetitive Ca2+ oscillations, starting soon after sperm-egg fusion. Over the last 15 years, accumulating scientific and clinical evidence supports the notion that the physiological stimulus that precedes the cytosolic Ca2+ oscillations is a novel, testis-specific phospholipase C (PLC) isoform, known as PLC-zeta (PLCζ). Sperm PLCζ catalyses the hydrolysis of phosphatidylinositol 4,5-bisphosphate triggering cytosolic Ca2+ oscillations through the inositol 1,4,5-trisphosphate signalling pathway. PLCζ is the smallest known mammalian PLC isoform with the most elementary domain organisation. However, relative to somatic PLCs, the PLCζ isoform possesses a unique potency in stimulating Ca2+ oscillations in eggs that is attributed to its novel biochemical characteristics. In this review, we discuss the latest developments that have begun to unravel the vital role of PLCζ at mammalian fertilisation and decipher its unique mechanism of action within the fertilising egg. We also postulate the significant potential diagnostic and therapeutic capacity of PLCζ in alleviating certain types of male infertility.


Subject(s)
Calcium Signaling/physiology , Phosphoinositide Phospholipase C/metabolism , Sperm Capacitation/physiology , Sperm-Ovum Interactions/physiology , Spermatozoa/enzymology , Animals , Female , Humans , Infertility, Male/enzymology , Infertility, Male/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Phosphoinositide Phospholipase C/genetics
17.
Sci Rep ; 7(1): 6312, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740084

ABSTRACT

The Cardiomyopathy-associated gene 5 (Cmya5) encodes myospryn, a large tripartite motif (TRIM)-related protein found predominantly in cardiac and skeletal muscle. Cmya5 is an expression biomarker for a number of diseases affecting striated muscle and may also be a schizophrenia risk gene. To further understand the function of myospryn in striated muscle, we searched for additional myospryn paralogs. Here we identify a novel muscle-expressed TRIM-related protein minispryn, encoded by Fsd2, that has extensive sequence similarity with the C-terminus of myospryn. Cmya5 and Fsd2 appear to have originated by a chromosomal duplication and are found within evolutionarily-conserved gene clusters on different chromosomes. Using immunoaffinity purification and mass spectrometry we show that minispryn co-purifies with myospryn and the major cardiac ryanodine receptor (RyR2) from heart. Accordingly, myospryn, minispryn and RyR2 co-localise at the junctional sarcoplasmic reticulum of isolated cardiomyocytes. Myospryn redistributes RyR2 into clusters when co-expressed in heterologous cells whereas minispryn lacks this activity. Together these data suggest a novel role for the myospryn complex in the assembly of ryanodine receptor clusters in striated muscle.


Subject(s)
Carrier Proteins/genetics , Cloning, Molecular/methods , Muscle Proteins/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , COS Cells , Carrier Proteins/metabolism , Chlorocebus aethiops , Chromatography, Affinity , Chromosome Duplication , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Mass Spectrometry , Mice , Muscle Proteins/metabolism , Sarcoplasmic Reticulum/metabolism
18.
Biochem J ; 474(6): 1003-1016, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28270562

ABSTRACT

Sperm-specific phospholipase C zeta (PLCζ) is widely considered to be the physiological stimulus that evokes intracellular calcium (Ca2+) oscillations that are essential for the initiation of egg activation during mammalian fertilisation. A recent genetic study reported a male infertility case that was directly associated with a point mutation in the PLCζ C2 domain, where an isoleucine residue had been substituted with a phenylalanine (I489F). Here, we have analysed the effect of this mutation on the in vivo Ca2+ oscillation-inducing activity and the in vitro biochemical properties of human PLCζ. Microinjection of cRNA or recombinant protein corresponding to PLCζI489F mutant at physiological concentrations completely failed to cause Ca2+ oscillations and trigger development. However, this infertile phenotype could be effectively rescued by microinjection of relatively high (non-physiological) amounts of recombinant mutant PLCζI489F protein, leading to Ca2+ oscillations and egg activation. Our in vitro biochemical analysis suggested that the PLCζI489F mutant displayed similar enzymatic properties, but dramatically reduced binding to PI(3)P and PI(5)P-containing liposomes compared with wild-type PLCζ. Our findings highlight the importance of PLCζ at fertilisation and the vital role of the C2 domain in PLCζ function, possibly due to its novel binding characteristics.


Subject(s)
C2 Domains , Calcium/metabolism , Infertility, Male/genetics , Phosphoinositide Phospholipase C/chemistry , Point Mutation , Amino Acid Substitution , Animals , Calcium Signaling , Cattle , Female , Fertilization , Gene Expression , Humans , Isoleucine/chemistry , Isoleucine/metabolism , Liposomes/chemistry , Liposomes/metabolism , Male , Mice , Microinjections , Oocytes/cytology , Oocytes/metabolism , Phenylalanine/chemistry , Phenylalanine/metabolism , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphoinositide Phospholipase C/genetics , Phosphoinositide Phospholipase C/metabolism , Protein Binding , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , RNA, Complementary/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spermatozoa/metabolism , Spermatozoa/pathology
19.
Mol Hum Reprod ; 23(1): 54-67, 2017 01.
Article in English | MEDLINE | ID: mdl-27932551

ABSTRACT

STUDY QUESTION: Is it possible to improve clinical visualization of phospholipase C zeta (PLCζ) as a diagnostic marker of sperm oocyte activation capacity and male fertility? SUMMARY ANSWER: Poor PLCζ visualization efficacy using current protocols may be due to steric or conformational occlusion of native PLCζ, hindering antibody access, and is significantly enhanced using antigen unmasking/retrieval (AUM) protocols. WHAT IS KNOWN ALREADY: Mammalian oocyte activation is mediated via a series of intracellular calcium (Ca2+) oscillations induced by sperm-specific PLCζ. PLCζ represents not only a potential clinical therapeutic in cases of oocyte activation deficiency but also a diagnostic marker of sperm fertility. However, there are significant concerns surrounding PLCζ antibody specificity and detection protocols. STUDY DESIGN, SIZE DURATION: Two PLCζ polyclonal antibodies, with confirmed PLCζ specificity, were employed in mouse, porcine and human sperm. Experiments evaluated PLCζ visualization efficacy, and whether AUM improved this. Antibodies against two sperm-specific proteins [post-acrosomal WW-binding protein (PAWP) and acrosin] were used as controls. PARTICIPANTS/MATERIALS, SETTING, METHODS: Aldehyde- and methanol-fixed sperm were subject to immunofluorescence analysis following HCl exposure (pH = 0.1-0.5), acid Tyrode's solution exposure (pH = 2.5) or heating in 10 mM sodium citrate solution (pH = 6.0). Fluorescence intensity of at least 300 cells was recorded for each treatment, with three independent repeats. MAIN RESULTS AND THE ROLE OF CHANCE: Despite high specificity for native PLCζ following immunoblotting using epitope-specific polyclonal PLCζ antibodies in mouse, porcine and human sperm, immunofluorescent visualization efficacy was poor. In contrast, sperm markers PAWP and acrosin exhibited relatively impressive results. All methods of AUM on aldehyde-fixed sperm enhanced visualization efficacy for PLCζ compared to visualization efficacy before AUM (P < 0.05 for all AUM interventions), but exerted no significant change upon PAWP or acrosin immunofluorescence following AUM. All methods of AUM enhanced PLCζ visualization efficacy in mouse and human methanol-fixed sperm compared to without AUM (P < 0.05 for all AUM interventions), while no significant change was observed in methanol-fixed porcine sperm before and after. In the absence of aldehyde-induced cross-linkages, such results suggest that poor PLCζ visualization efficacy may be due to steric or conformational occlusion of native PLCζ, hindering antibody access. Importantly, examination of sperm from individual donors revealed that AUM differentially affects observable PLCζ fluorescence, and the proportion of sperm exhibiting detectable PLCζ fluorescence in sperm from different males. LIMITATIONS, REASONS FOR CAUTION: Direct correlation of fertility outcomes with the level of PLCζ in the sperm samples studied was not available. Such analyses would be required in future to determine whether the improved methodology for PLCζ visualization we propose would indeed reflect fertility status. WIDER IMPLICATIONS OF THE FINDINGS: We propose that AUM alters conformational interactions to enhance PLCζ epitope availability and visualization efficacy, supporting prospective application of AUM to reduce misinterpretation in clinical diagnosis of PLCζ-linked male infertility. Our current results suggest that it is perhaps prudent that previous studies investigating links between PLCζ and fertility parameters are re-examined in the context of AUM, and may pave the way for future work to answer significant questions such as how PLCζ appears to be kept in an inactive form in the sperm. LARGE SCALE DATA: Not applicable. STUDY FUNDING/COMPETING INTERESTS: J.K. is supported by a Health Fellowship award from the National Institute for Social Care and Health Research (NISCHR). M.N. is supported by a Marie Curie Intra-European Research Fellowship award. This work was also partly funded by a research grant from Cook Medical Technologies LLC. There are no competing financial interests to declare.


Subject(s)
Fluorescent Antibody Technique/standards , Infertility, Male/enzymology , Phosphoinositide Phospholipase C/analysis , Sperm-Ovum Interactions/physiology , Spermatozoa/enzymology , Acrosin/genetics , Acrosin/immunology , Animals , Antibodies/chemistry , Antibody Specificity , Antigen-Antibody Complex/chemistry , Biomarkers/metabolism , Carrier Proteins/genetics , Carrier Proteins/immunology , Gene Expression , Humans , Infertility, Male/genetics , Male , Mice , Oocytes/cytology , Oocytes/physiology , Phosphoinositide Phospholipase C/chemistry , Phosphoinositide Phospholipase C/genetics , Phosphoinositide Phospholipase C/immunology , Protein Binding , Protein Conformation , Seminal Plasma Proteins/genetics , Seminal Plasma Proteins/immunology , Spermatozoa/pathology , Swine , Tissue Fixation/methods
20.
FEBS J ; 283(24): 4502-4514, 2016 12.
Article in English | MEDLINE | ID: mdl-27783455

ABSTRACT

Hereditary leukonychia is a rare genetic nail disorder characterized by distinctive whitening of the nail plate of all 20 nails. Hereditary leukonychia may exist as an isolated feature, or in simultaneous occurrence with other cutaneous or systemic pathologies. Associations between hereditary leukonychia and mutations in the gene encoding phospholipase C delta-1 (PLCδ1) have previously been identified. However, the molecular mechanisms underlying PLCδ1 mutations and hereditary leukonychia remain uncharacterized. In the present study, we introduced hereditary leukonychia-linked human PLCδ1 mutations (C209R, A574T and S740R) into equivalent residues of rat PLCδ1 (C188R, A553T and S719R), and investigated their effect on the biophysical and biochemical properties of the PLCδ1 protein. Our data suggest that these PLCδ1 mutations associated with hereditary leukonychia do not uniformly alter the enzymatic ability of this protein leading to loss/gain of function, but result in significantly divergent enzymatic properties. We demonstrate here for the first time the importance of PLC-mediated calcium (Ca2+ ) signalling within the manifestation of hereditary leukonychia. PLCδ1 is almost ubiquitous in mammalian cells, which may explain why hereditary leukonychia manifests in association with other systemic pathologies relating to keratin expression.


Subject(s)
Mutation , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C delta/genetics , Phospholipase C delta/metabolism , Animals , Binding Sites/genetics , Biocatalysis , Blotting, Western , Calcium/metabolism , Circular Dichroism , Enzyme Stability , Humans , Hydrolysis , Hypopigmentation/genetics , Kinetics , Models, Molecular , Nail Diseases/congenital , Nail Diseases/genetics , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phospholipase C delta/chemistry , Protein Binding , Protein Domains , Rats , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...