1
|
Sinaei R, Eslami M, Dadfar M, Saberi A. Identification of a new mutation in the ACTL9 gene in men with unexplained infertility. Mol Genet Genomic Med 2024; 12:e2448. [PMID: 38769899 PMCID: PMC11106586 DOI: 10.1002/mgg3.2448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/04/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Infertility is defined as the failure to achieve pregnancy after one year of unprotected intercourse within a marital relationship. Approximately 10%-15% of couples worldwide experience infertility issues, with nearly half of these cases attributed to male factors. Among men with unexplained infertility, genetic mutations have been identified as a potential cause. Studies have indicated that mutations affecting the function of the protein encoded by the ACTL9 gene may play a role in male infertility. METHODS The purpose of this research was to identify mutations in the ACTL9 gene associated with male infertility in a sample of 40 infertile men with unknown causes. Genomic DNA extraction and PCR amplification were carried out on samples from each individual. The genetic material was then analyzed using Sanger sequencing, followed by bioinformatics and segregation analysis to determine the potential effects of the observed variations. RESULT A novel genetic variant, c.376G>A (p.Glu126Lys), was identified in an infertile male individual, representing a previously unreported finding that was validated through segregation analyses. This specific variant induces a change from glutamate to lysine at the amino acid level by replacing the nucleotide G with A in the genomic DNA sequence, consequently impacting the secondary structure and function of the protein. CONCLUSIONS The conclusive analysis of the procedure indicated that this alteration has the potential to interfere with the process of fertilization, ultimately resulting in the complete failure of fertilization (TFF) and causing male infertility.
Collapse
Affiliation(s)
- Roya Sinaei
- Department of Genetics, Faculty of Advanced Science and TechnologyTehran Medical Sciences, Islamic Azad UniversityTehranIran
| | - Maryam Eslami
- Department of Genetics, Faculty of Advanced Science and TechnologyTehran Medical Sciences, Islamic Azad UniversityTehranIran
- Applied Biotechnology Research Center, Tehran Medical SciencesIslamic Azad UniversityTehranIran
- International FacultyTehran Medical Sciences, Islamic Azad UniversityTehranIran
| | - Mohammadreza Dadfar
- Department of Urology, Imam Khomeini Hospital, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Alihossein Saberi
- Department of Medical Genetics, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
- Cellular and Molecular Research CenterAhvaz Jundishapur University of Medical SciencesAhvazIran
| |
Collapse
|
2
|
Ferrer P, Upadhyay S, Cai JJ, Clement TM. Novel Nuclear Roles for Testis-Specific ACTL7A and ACTL7B Supported by In Vivo Characterizations and AI Facilitated In Silico Mechanistic Modeling with Implications for Epigenetic Regulation in Spermiogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582797. [PMID: 38464253 PMCID: PMC10925299 DOI: 10.1101/2024.02.29.582797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
A mechanistic role for nuclear function of testis-specific actin related proteins (ARPs) is proposed here through contributions of ARP subunit swapping in canonical chromatin regulatory complexes. This is significant to our understanding of both mechanisms controlling regulation of spermiogenesis, and the expanding functional roles of the ARPs in cell biology. Among these roles, actins and ARPs are pivotal not only in cytoskeletal regulation, but also in intranuclear chromatin organization, influencing gene regulation and nucleosome remodeling. This study focuses on two testis-specific ARPs, ACTL7A and ACTL7B, exploring their intranuclear activities and broader implications utilizing combined in vivo, in vitro, and in silico approaches. ACTL7A and ACTL7B, previously associated with structural roles, are hypothesized here to serve in chromatin regulation during germline development. This study confirms the intranuclear presence of ACTL7B in spermatocytes and round spermatids, revealing a potential role in intranuclear processes, and identifies a putative nuclear localization sequence conserved across mammalian ACTL7B, indicating a potentially unique mode of nuclear transport which differs from conventional actin. Ablation of ACTL7B leads to varied transcriptional changes reported here. Additionally, in the absence of ACTL7A or ACTL7B there is a loss of intranuclear localization of HDAC1 and HDAC3, which are known regulators of epigenetic associated acetylation changes that in turn regulate gene expression. Thus, these HDACs are implicated as contributors to the aberrant gene expression observed in the KO mouse testis transcriptomic analysis. Furthermore, this study employed and confirmed the accuracy of in silico models to predict ARP interactions with Helicase-SANT-associated (HSA) domains, uncovering putative roles for testis-specific ARPs in nucleosome remodeling complexes. In these models, ACTL7A and ACTL7B were found capable of binding to INO80 and SWI/SNF nucleosome remodeler family members in a manner akin to nuclear actin and ACTL6A. These models thus implicate germline-specific ARP subunit swapping within chromatin regulatory complexes as a potential regulatory mechanism for chromatin and associated molecular machinery adaptations in nuclear reorganizations required during spermiogenesis. These results hold implications for male fertility and epigenetic programing in the male-germline that warrant significant future investigation. In summary, this study reveals that ACTL7A and ACTL7B play intranuclear gene regulation roles in male gametogenesis, adding to the multifaceted roles identified also spanning structural, acrosomal, and flagellar stability. ACTL7A and ACTL7B unique nuclear transport, impact on HDAC nuclear associations, impact on transcriptional processes, and proposed mechanism for involvement in nucleosome remodeling complexes supported by AI facilitated in silico modeling contribute to a more comprehensive understanding of the indispensable functions of ARPs broadly in cell biology, and specifically in male fertility.
Collapse
Affiliation(s)
- Pierre Ferrer
- Interdisciplinary Faculty of Toxicology Program, Texas A&M University, College Station, TX 77843
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843
| | - Srijana Upadhyay
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843
| | - James J Cai
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843
| | - Tracy M Clement
- Interdisciplinary Faculty of Toxicology Program, Texas A&M University, College Station, TX 77843
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843
| |
Collapse
|
3
|
Merges GE, Arévalo L, Kovacevic A, Lohanadan K, de Rooij DG, Simon C, Jokwitz M, Witke W, Schorle H. Actl7b deficiency leads to mislocalization of LC8 type dynein light chains and disruption of murine spermatogenesis. Development 2023; 150:dev201593. [PMID: 37800308 PMCID: PMC10652042 DOI: 10.1242/dev.201593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
Actin-related proteins (Arps) are classified according to their similarity to actin and are involved in diverse cellular processes. ACTL7B is a testis-specific Arp, and is highly conserved in rodents and primates. ACTL7B is specifically expressed in round and elongating spermatids during spermiogenesis. Here, we have generated an Actl7b-null allele in mice to unravel the role of ACTL7B in sperm formation. Male mice homozygous for the Actl7b-null allele (Actl7b-/-) were infertile, whereas heterozygous males (Actl7b+/-) were fertile. Severe spermatid defects, such as detached acrosomes, disrupted membranes and flagella malformations start to appear after spermiogenesis step 9 in Actl7b-/- mice, finally resulting in spermatogenic arrest. Abnormal spermatids were degraded and levels of autophagy markers were increased. Co-immunoprecipitation with mass spectrometry experiments identified an interaction between ACTL7B and the LC8 dynein light chains DYNLL1 and DYNLL2, which are first detected in step 9 spermatids and mislocalized when ACTL7B is absent. Our data unequivocally establish that mutations in ACTL7B are directly related to male infertility, pressing for additional research in humans.
Collapse
Affiliation(s)
- Gina E. Merges
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Lena Arévalo
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Andjela Kovacevic
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Keerthika Lohanadan
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Dirk G. de Rooij
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Carla Simon
- Cell Migration Unit, Institute of Genetics, University of Bonn, 53115 Bonn, Germany
| | - Melanie Jokwitz
- Cell Migration Unit, Institute of Genetics, University of Bonn, 53115 Bonn, Germany
| | - Walter Witke
- Cell Migration Unit, Institute of Genetics, University of Bonn, 53115 Bonn, Germany
| | - Hubert Schorle
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
4
|
Moreno RD. Human globozoospermia-related genes and their role in acrosome biogenesis. WIREs Mech Dis 2023; 15:e1589. [PMID: 36493758 DOI: 10.1002/wsbm.1589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 12/13/2022]
Abstract
The mammalian acrosome is a secretory vesicle attached to the sperm nucleus whose fusion with the overlying plasma membrane is required to achieve fertilization. Acrosome biogenesis starts during meiosis, but it lasts through the entire process of haploid cell differentiation (spermiogenesis). Acrosome biogenesis is a stepwise process that involves membrane traffic from the Golgi apparatus, but it also seems that the lysosome/endosome system participates in this process. Defective sperm head morphology is accompanied by defective acrosome shape and function, and patients with these characteristics are infertile or subfertile. The most extreme case of acrosome biogenesis failure is globozoospermia syndrome, which is primarily characterized by the presence of round-headed spermatozoa without acrosomes with cytoskeleton defects around the nucleus and infertility. Several genes participating in acrosome biogenesis have been uncovered using genetic deletions in mice, but only a few of them have been found to be deleted or modified in patients with globozoospermia. Understanding acrosome biogenesis is crucial to uncovering the molecular basis of male infertility and developing new diagnostic tools and assisted reproductive technologies that may help infertile patients through more effective treatment techniques. This article is categorized under: Reproductive System Diseases > Environmental Factors Infectious Diseases > Stem Cells and Development Reproductive System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Ricardo D Moreno
- Departmento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
5
|
Davalieva K, Rusevski A, Velkov M, Noveski P, Kubelka-Sabit K, Filipovski V, Plaseski T, Dimovski A, Plaseska-Karanfilska D. Comparative proteomics analysis of human FFPE testicular tissues reveals new candidate biomarkers for distinction among azoospermia types and subtypes. J Proteomics 2022; 267:104686. [PMID: 35914715 DOI: 10.1016/j.jprot.2022.104686] [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: 04/01/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 10/16/2022]
Abstract
Understanding molecular mechanisms that underpin azoospermia and discovery of biomarkers that could enable reliable, non-invasive diagnosis is highly needed. Using label-free data-independent LC-MS/MS acquisition coupled with ion mobility, we compared the FFPE testicular proteome of patients with obstructive (OA) and non-obstructive azoospermia (NOA) subtypes hypospermatogenesis (Hyp) and Sertoli cell-only syndrome (SCO). Out of 2044 proteins identified based on ≥2 peptides, 61 proteins had the power to quantitatively discriminate OA from NOA and 30 to quantitatively discriminate SCO from Hyp and OA. Among these, H1-6, RANBP1 and TKTL2 showed superior potential for quantitative discrimination among OA, Hyp and SCO. Integrin signaling pathway, adherens junction, planar cell polarity/convergent extension pathway and Dectin-1 mediated noncanonical NF-kB signaling were significantly associated with the proteins that could discriminate OA from NOA. Comparison with 2 transcriptome datasets revealed 278 and 55 co-differentially expressed proteins/genes with statistically significant positive correlation. Gene expression analysis by qPCR of 6 genes (H1-6, RANBP1, TKTL2, TKTL1, H2BC1, and ACTL7B) with the highest discriminatory power on protein level and the same regulation trend with transcriptomic datasets, confirmed proteomics results. In summary, our results suggest some underlying pathways in azoospermia and broaden the range of potential novel candidates for diagnosis. SIGNIFICANCE: Using a comparative proteomics approach on testicular tissue we have identified several pathways associated with azoospermia and a number of testis-specific and germ cell-specific proteins that have the potential to pinpoint the type of spermatogenesis failure. Furthermore, comparison with transcriptomics datasets based on genome-wide gene expression analyses of human testis specimens from azoospermia patients identified proteins that could discriminate between obstructive and non-obstructive azoospermia subtypes on both protein and mRNA levels. Up to our knowledge, this is the first integrated comparative analysis of proteomics and transcriptomics data from testicular tissues. We believe that the data from our study contributes significantly to increase the knowledge of molecular mechanisms of azoospermia and pave the way for new investigations in regards to non-invasive diagnosis.
Collapse
Affiliation(s)
- Katarina Davalieva
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia.
| | - Aleksandar Rusevski
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia
| | - Milan Velkov
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia
| | - Predrag Noveski
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia
| | - Katerina Kubelka-Sabit
- Laboratory for Histopathology, Clinical Hospital "Sistina", 1000 Skopje, North Macedonia, Macedonia
| | - Vanja Filipovski
- Laboratory for Histopathology, Clinical Hospital "Sistina", 1000 Skopje, North Macedonia, Macedonia
| | - Toso Plaseski
- Faculty of Medicine, Endocrinology and Metabolic Disorders Clinic, 1000 Skopje, North Macedonia, Macedonia
| | - Aleksandar Dimovski
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia; Faculty of Pharmacy, University "St. Cyril and Methodius", 1000 Skopje, North Macedonia, Macedonia
| | - Dijana Plaseska-Karanfilska
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia, Macedonia.
| |
Collapse
|
6
|
Zhang XZ, Wei LL, Zhang XH, Jin HJ, Chen SR. Loss of perinuclear theca ACTRT1 causes acrosome detachment and severe male subfertility in mice. Development 2022; 149:275523. [DOI: 10.1242/dev.200489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 05/12/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The perinuclear theca (PT) is a cytoskeletal element encapsulating the sperm nucleus; however, the physiological roles of the PT in sperm are largely uncertain. Here, we reveal that ACTRT1, ACTRT2, ACTL7A and ACTL9 proteins interact to form a multimeric complex and localize to the subacrosomal region of spermatids. Furthermore, we engineered Actrt1-knockout (KO) mice to define the functions of ACTRT1. Despite normal sperm count and motility, Actrt1-KO males were severely subfertile owing to a deficiency in fertilization. Loss of ACTRT1 caused a high incidence of malformed heads and detachment of acrosomes from sperm nuclei, caused by loosened acroplaxome structure during spermiogenesis. Furthermore, Actrt1-KO sperm showed reduced ACTL7A and PLCζ protein content as a potential cause of fertilization defects. Moreover, we reveal that ACTRT1 anchors developing acrosomes to the nucleus, likely by interacting with the inner acrosomal membrane protein SPACA1 and the nuclear envelope proteins PARP11 and SPATA46. Loss of ACTRT1 weakened the interaction between ACTL7A and SPACA1. Our study and recent findings of ACTL7A/ACTL9-deficient sperm together reveal that the sperm PT-specific ARP complex mediates the acrosome-nucleus connection.
Collapse
Affiliation(s)
- Xiao-Zhen Zhang
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| | - Lin-Lin Wei
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| | - Xiao-Hui Zhang
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| | - Hui-Juan Jin
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| | - Su-Ren Chen
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| |
Collapse
|
7
|
Dai J, Zhang T, Guo J, Zhou Q, Gu Y, Zhang J, Hu L, Zong Y, Song J, Zhang S, Dai C, Gong F, Lu G, Zheng W, Lin G. Homozygous pathogenic variants in ACTL9 cause fertilization failure and male infertility in humans and mice. Am J Hum Genet 2021; 108:469-481. [PMID: 33626338 DOI: 10.1016/j.ajhg.2021.02.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Total fertilization failure (TFF) can occur during in vitro fertilization (IVF) treatments, even following intracytoplasmic sperm injection (ICSI). Various male or female factors could contribute to TFF. Increasing evidence suggested that genetic variations in PLCZ1, which encodes 1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase zeta-1 (PLCζ), is involved in oocyte activation and is a key male factor in TFF. In the present study, we explored the genetic variants in male individuals that led to TFF. A total of 54 couples with TFF or poor fertilization (fertilization rate < 20%) were screened, and 21 couples were determined to have a male infertility factor by the mouse oocyte activation test. Whole-exome sequencing of these 21 male individuals identified three homozygous pathogenic variants in ACTL9 (actin like 9) in three individuals. ACTL9 variations led to abnormal ultrastructure of the perinuclear theca (PT), and PLCζ was absent in the head and present in the neck of the mutant sperm, which contributed to failed normal calcium oscillations in oocytes and subsequent TFF. The key roles of ACTL9 in the PT structure and TFF after ICSI were further confirmed in an Actl9-mutated mouse model. Furthermore, assisted oocyte activation by calcium ionophore exposure successfully overcame TFF and achieved live births in a couple with an ACTL9 variant. These findings identified the role of ACTL9 in the PT structure and the correct localization of PLCζ. The results also provide a genetic marker and a therapeutic option for individuals who have undergone ICSI without successful fertilization.
Collapse
|
8
|
Tanaka H, Miyagawa Y, Tsujimura A, Wada M. Genetic Polymorphisms within The Intronless ACTL7A and ACTL7B Genes Encoding Spermatogenesis-Specific Actin-Like Proteins in Japanese Males. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2019; 13:245-249. [PMID: 31310081 PMCID: PMC6642428 DOI: 10.22074/ijfs.2019.5702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 02/16/2019] [Indexed: 11/18/2022]
Abstract
Actins play essential roles in cellular morphogenesis. In mice, the T-actin1 and 2 genes, which encode actin-like
proteins, are specifically expressed in haploid germ cells. Both T-ACTIN1/ACTLB and T-ACTIN2/ACTL7A have also
been cloned and studied. The orthologous genes in humans are present on chromosome 9q31.3 as intronless genes.
Defects of germ cell-specific genes can introduce infertility without somatic function impairment. We determined T-
ACTIN1 and 2, specifically expressed in the testis using reverse-transcription polymerase chain reaction (RT-PCR).
To examine whether genetic polymorphisms of the T-ACTIN1 and 2 genes are associated with male infertility, we
screened for T-ACTIN1 and 2 polymorphisms by direct sequencing of DNA from 282 sterile and 89 fertile Japanese
men. We identified five and six single nucleotide polymorphisms (SNPs) in the T-ACTIN1 and 2 regions of the sterile
and fertile subjects respectively. Among these genetic polymorphisms was a novel SNP that was not in the National
Center for Biotechnology Information SNP database. Although we could not determine whether these SNPs cause
infertility, the prevalence of these genetic polymorphisms may be useful for analyzing polymorphisms in future large-
scale genetic analyses.
Collapse
Affiliation(s)
- Hiromitsu Tanaka
- Faculty of Pharmaceutical Sciences, Nagasaki International University, Huis Ten Bosch, Sasebo, Nagasaki, Japan.Electronic Address:
| | - Yasushi Miyagawa
- Department of Urology, Graduate School of Medicine, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | - Akira Tsujimura
- Department of Urology, Juntendo University Hospital, Hongo, bunnkyouku, Tokyo, Japan
| | - Morimasa Wada
- Faculty of Pharmaceutical Sciences, Nagasaki International University, Huis Ten Bosch, Sasebo, Nagasaki, Japan
| |
Collapse
|
9
|
Ahn J, Park YJ, Chen P, Lee TJ, Jeon YJ, Croce CM, Suh Y, Hwang S, Kwon WS, Pang MG, Kim CH, Lee SS, Lee K. Comparative expression profiling of testis-enriched genes regulated during the development of spermatogonial cells. PLoS One 2017; 12:e0175787. [PMID: 28414809 PMCID: PMC5393594 DOI: 10.1371/journal.pone.0175787] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/31/2017] [Indexed: 12/12/2022] Open
Abstract
The testis has been identified as the organ in which a large number of tissue-enriched genes are present. However, a large portion of transcripts related to each stage or cell type in the testis still remains unknown. In this study, databases combined with confirmatory measurements were used to investigate testis-enriched genes, localization in the testis, developmental regulation, gene expression profiles of testicular disease, and signaling pathways. Our comparative analysis of GEO DataSets showed that 24 genes are predominantly expressed in testis. Cellular locations of 15 testis-enriched proteins in human testis have been identified and most of them were located in spermatocytes and round spermatids. Real-time PCR revealed that expressions of these 15 genes are significantly increased during testis development. Also, an analysis of GEO DataSets indicated that expressions of these 15 genes were significantly decreased in teratozoospermic patients and polyubiquitin knockout mice, suggesting their involvement in normal testis development. Pathway analysis revealed that most of those 15 genes are implicated in various sperm-related cell processes and disease conditions. This approach provides effective strategies for discovering novel testis-enriched genes and their expression patterns, paving the way for future characterization of their functions regarding infertility and providing new biomarkers for specific stages of spematogenesis.
Collapse
Affiliation(s)
- Jinsoo Ahn
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Yoo-Jin Park
- Center for Systems Biology, Program in Membrane Biology/Nephrology Division, Massachusetts General Hospital, Boston, MA and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Paula Chen
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Tae Jin Lee
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, United States of America
| | - Young-Jun Jeon
- Stanford Cancer Institute, Stanford University, Stanford, California, United States of America
| | - Carlo M. Croce
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, United States of America
| | - Yeunsu Suh
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Seongsoo Hwang
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Wanju-gun, Jeonbuk, Republic of Korea
| | - Woo-Sung Kwon
- Department of Animal Biotechnology, Kyungpook National University, Sangju, Republic of Korea
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea
| | - Myung-Geol Pang
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea
| | - Cheorl-Ho Kim
- Department of Biological Sciences, SungKyunKwan University, Chunchun-Dong, Jangan-Gu, Suwon City, Kyunggi-Do, Republic of Korea
| | - Sang Suk Lee
- Department of Animal Science and Technology, Sunchon National University, Suncheon, Republic of Korea
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
10
|
Caso E, Maestro A, Sabiers CC, Godino M, Caracuel Z, Pons J, Gonzalez FJ, Bautista R, Claros MG, Caso-Onzain J, Viejo-Allende E, Giannoudis PV, Alvarez S, Maietta P, Guerado E. Whole-exome sequencing analysis in twin sibling males with an anterior cruciate ligament rupture. Injury 2016; 47 Suppl 3:S41-S50. [PMID: 27692106 DOI: 10.1016/s0020-1383(16)30605-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Familial predisposition is among the major genetic risk factors for non-contact musculoskeletal tissue injuries. Personal genome sequence shows that different polymorphism profiles may account for the number and the degree of injuries and the recovery time. Genotyping studies allow investigation into genome factors with potential impact on pathogenesis of non-contact ligament injuries. We have studied a family with twin sibling males surgically diagnosed of an anterior cruciate ligament non-contact rupture and non-affected progenitors (father and mother) were subjected to whole exome sequencing (WES) analysis. WES analysis previously carried out on 16 individuals, without ACL injury medical records, were also included in this study for single nucleotide variants (SNVs) and small insertions and deletions detection (indels), variant filtering and to prioritize variants relative to the disease. WES analysis to identify SNVs and indels was performed using open web-based bioinformatics tools. A set of 11 new variants shared by family members can be associated to ACL non-contact injury, including SerpinA11, ARSI, NOCHT4, EPB41, FDFT1, POMC, KIF26A, OLFML2B, ATG7, FAH and WDR6. All of them, except ATG7 and WDR6, have shown a damaging predictive pattern by combinatorial standard predictive scores. In combination to the identified SNVs of EPB41 and SerpinA11 genes, ACTL7A gene showed a predicted deleterious variant reinforcing the idea these variants impact on of fibroblast-like cells deformability and ECM misbalance, Differential gene expression and RNA sequencing analysis will help to understand the combined participation of these protein coding genes in ACL non-contact injuries.
Collapse
Affiliation(s)
- Enrique Caso
- Research & Development Unit, Hospital Universitario Costa del Sol, University of Malaga, Marbella, Malaga, Spain.
| | - Antonio Maestro
- Department of Orthopaedic Surgery, FREMAP, Gijon, Asturias, Spain
| | - Cristina C Sabiers
- Research & Development Unit, Hospital Universitario Costa del Sol, University of Malaga, Marbella, Malaga, Spain
| | - Manuel Godino
- Department of Orthopaedic Surgery and Traumatology, Hospital Universitario Costa del Sol, University of Malaga, Marbella, Malaga, Spain
| | - Zaira Caracuel
- Research & Development Unit, Hospital Universitario Costa del Sol, University of Malaga, Marbella, Malaga, Spain
| | - Joana Pons
- Research & Development Unit, Hospital Universitario Costa del Sol, University of Malaga, Marbella, Malaga, Spain
| | - F Jesus Gonzalez
- Research & Development Unit, Hospital Universitario Costa del Sol, University of Malaga, Marbella, Malaga, Spain
| | - Rocio Bautista
- Supercomputing and Bioinnovating Centre (SCBI), University of Malaga, Malaga, Spain
| | - M Gonzalo Claros
- Supercomputing and Bioinnovating Centre (SCBI), University of Malaga, Malaga, Spain
| | - Jaime Caso-Onzain
- Research & Development Unit, Hospital Universitario Costa del Sol, University of Malaga, Marbella, Malaga, Spain
| | - Elena Viejo-Allende
- Research & Development Unit, Hospital Universitario Costa del Sol, University of Malaga, Marbella, Malaga, Spain
| | - Peter V Giannoudis
- Academic Department of Trauma & Orthopaedic Surgery, University of Leeds, NIHR Leeds Biomedical Research Unit, Chapel Allerton Hospital, Leeds, UK
| | | | | | - Enrique Guerado
- Department of Orthopaedic Surgery and Traumatology, Hospital Universitario Costa del Sol, University of Malaga, Marbella, Malaga, Spain
| |
Collapse
|
11
|
Norcliffe-Kaufmann L, Slaugenhaupt SA, Kaufmann H. Familial dysautonomia: History, genotype, phenotype and translational research. Prog Neurobiol 2016; 152:131-148. [PMID: 27317387 DOI: 10.1016/j.pneurobio.2016.06.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/10/2016] [Accepted: 06/11/2016] [Indexed: 01/30/2023]
Abstract
Familial dysautonomia (FD) is a rare neurological disorder caused by a splice mutation in the IKBKAP gene. The mutation arose in the 1500s within the small Jewish founder population in Eastern Europe and became prevalent during the period of rapid population expansion within the Pale of Settlement. The carrier rate is 1:32 in Jews descending from this region. The mutation results in a tissue-specific deficiency in IKAP, a protein involved in the development and survival of neurons. Patients homozygous for the mutations are born with multiple lesions affecting mostly sensory (afferent) fibers, which leads to widespread organ dysfunction and increased mortality. Neurodegenerative features of the disease include progressive optic atrophy and worsening gait ataxia. Here we review the progress made in the last decade to better understand the genotype and phenotype. We also discuss the challenges of conducting controlled clinical trials in this rare medically fragile population. Meanwhile, the search for better treatments as well as a neuroprotective agent is ongoing.
Collapse
Affiliation(s)
| | - Susan A Slaugenhaupt
- Center for Human Genetic Research, Massachusetts General Hospital Research Institute and Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Horacio Kaufmann
- Department of Neurology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Dkhil MA, Al-Quraishy S, Abdel-Baki AA, Ghanjati F, Arauzo-Bravo MJ, Delic D, Wunderlich F. Epigenetic modifications of gene promoter DNA in the liver of adult female mice masculinized by testosterone. J Steroid Biochem Mol Biol 2015; 145:121-30. [PMID: 25448745 DOI: 10.1016/j.jsbmb.2014.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 11/02/2014] [Accepted: 11/04/2014] [Indexed: 12/17/2022]
Abstract
Testosterone (T) is known to masculinize the female phenotype of the liver, evidenced as up- and down-regulated expressions of male- and female-predominant genes, respectively, involved in hepatic metabolism. This study is aimed at identifying epigenetic modifications of promoters of these differently expressed genes in the liver after masculinization by T of adult female C57BL/6 mice using methylated DNA immunoprecipitation and NimbleGen microarrays. Among the 17,354 promoters examined, 82 promoters in the liver have been identified to be significantly changed by T (p<0.05), with 47 and 35 promoters exhibiting increased and decreased DNA methylation, respectively. Most of these promoters display the changes of DNA methylation in their Ups-regions, which are between +500 and +2000 bp upstream from the transcription start site (TSS) of the genes. Less T-induced modifications have been detected in the Cor-regions of the promoters, i.e., +500 to -500 bp around the TSS. Only 13 and 7 Cor-promoters are hyper- and hypo-methylated, respectively, among which are 10 hyper- and 5 hypo-methylated promoters of genes with annotated functions. Surprisingly, the promoters are largely unmethylated in those genes whose expression has been previously found to be permanently deregulated by T in the liver, as e.g. the T-upregulated male-predominant genes Cyp7b1, Cyp2d9, Cyp4a10, Ugt2b1, Ugt2b38, Hsd3b5, Slco1a1 as well as the T-downregulated female-predominant genes Cyp2b9, Cyp2b13, Cyp3a41, Cyp3a44, Fmo3, Sult2a2, respectively. Though methylatable, the promoter DNA of Ar, Esr1, and Esr2 remained unaffected by T. However, T decreases DNA-methylation of the Cor-promoter region of Ddc encoding the AR-coactivator dopa decarboxylase. Among the identified 15 Cor-promoters of genes with annotated functions are also those of Defb43, Cst11, and Sele involved in innate immunity. Our data support the view that T may exert long-lasting epigenetic effects on functions of the liver-inherent immune system.
Collapse
Affiliation(s)
- Mohamed A Dkhil
- Department of Zoology, College of Science, King Saud University, 11451 Riyadh, Saudi Arabia; Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt.
| | - Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Abdel-Azeem Abdel-Baki
- Department of Zoology, College of Science, King Saud University, 11451 Riyadh, Saudi Arabia; Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Foued Ghanjati
- Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University, Duesseldorf, Germany
| | - Marcos J Arauzo-Bravo
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, San Sebastion, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Denis Delic
- Department of Biology, Heinrich-Heine-University, Duesseldorf, Germany; Boehringer-Ingelheim, Biberach, Germany
| | - Frank Wunderlich
- Department of Biology, Heinrich-Heine-University, Duesseldorf, Germany
| |
Collapse
|
13
|
Vered K, Durrant C, Mott R, Iraqi FA. Susceptibility to Klebsiella pneumonaie infection in collaborative cross mice is a complex trait controlled by at least three loci acting at different time points. BMC Genomics 2014; 15:865. [PMID: 25283706 PMCID: PMC4201739 DOI: 10.1186/1471-2164-15-865] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 09/24/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Klebsiella pneumoniae (Kp) is a bacterium causing severe pneumonia in immunocompromised hosts and is often associated with sepsis. With the rise of antibiotic resistant bacteria, there is a need for new effective and affordable control methods; understanding the genetic architecture of susceptibility to Kp will help in their development. We performed the first quantitative trait locus (QTL) mapping study of host susceptibility to Kp infection in immunocompetent Collaborative Cross mice (CC). We challenged 328 mice from 73 CC lines intraperitoneally with 104 colony forming units of Kp strain K2. Survival and body weight were monitored for 15 days post challenge. 48 of the CC lines were genotyped with 170,000 SNPs, with which we mapped QTLs. RESULTS CC lines differed significantly (P < 0.05) in mean survival time, between 1 to 15 days post infection, and broad sense heritability was 0.45. Distinct QTL were mapped at specific time points during the challenge. A QTL on chromosome 4 was found only on day 2 post infection, and QTL on chromosomes 8 and 18, only on day 8. By using the sequence variations of the eight inbred strain founders of the CC to refine QTL localization we identify several candidate genes. CONCLUSION Host susceptibility to Kp is a complex trait, controlled by multiple genetic factors that act sequentially during the course of infection.
Collapse
Affiliation(s)
| | | | | | - Fuad A Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
14
|
Djureinovic D, Fagerberg L, Hallström B, Danielsson A, Lindskog C, Uhlén M, Pontén F. The human testis-specific proteome defined by transcriptomics and antibody-based profiling. Mol Hum Reprod 2014; 20:476-88. [PMID: 24598113 DOI: 10.1093/molehr/gau018] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The testis' function is to produce haploid germ cells necessary for reproduction. Here we have combined a genome-wide transcriptomics analysis with immunohistochemistry-based protein profiling to characterize the molecular components of the testis. Deep sequencing (RNA-Seq) of normal human testicular tissue from seven individuals was performed and compared with 26 other normal human tissue types. All 20 050 putative human genes were classified into categories based on expression patterns. The analysis shows that testis is the tissue with the most tissue-specific genes by far. More than 1000 genes show a testis-enriched expression pattern in testis when compared with all other analyzed tissues. Highly testis enriched genes were further characterized with respect to protein localization within the testis, such as spermatogonia, spermatocytes, spermatids, sperm, Sertoli cells and Leydig cells. Here we present an immunohistochemistry-based analysis, showing the localization of corresponding proteins in different cell types and various stages of spermatogenesis, for 62 genes expressed at >50-fold higher levels in testis when compared with other tissues. A large fraction of these genes were unexpectedly expressed in early stages of spermatogenesis. In conclusion, we have applied a genome-wide analysis to identify the human testis-specific proteome using transcriptomics and antibody-based protein profiling, providing lists of genes expressed in a tissue-enriched manner in the testis. The majority of these genes and proteins were previously poorly characterised in terms of localization and function, and our list provides an important starting point to increase our molecular understanding of human reproductive biology and disease.
Collapse
Affiliation(s)
- D Djureinovic
- Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala SE-751 85, Sweden Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-751 85, Sweden
| | - L Fagerberg
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-171 21, Sweden
| | - B Hallström
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-171 21, Sweden
| | - A Danielsson
- Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala SE-751 85, Sweden Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-751 85, Sweden
| | - C Lindskog
- Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala SE-751 85, Sweden Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-751 85, Sweden
| | - M Uhlén
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-171 21, Sweden
| | - F Pontén
- Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala SE-751 85, Sweden Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-751 85, Sweden
| |
Collapse
|
15
|
Oh SD, Park SY, Park JI, Chun SY, Ryu TH, Soh J. The novel, actin-like protein Tact3 is expressed in rodent testicular haploid germ cells. Mol Reprod Dev 2013; 80:988-99. [PMID: 24038581 DOI: 10.1002/mrd.22262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 08/16/2013] [Accepted: 08/28/2013] [Indexed: 11/06/2022]
Abstract
Mouse testis actin-like proteins 1 and 2 (mTact1 and mTact2), which are expressed in murine haploid germ cells, have been described previously. Here, we report the cloning and characterization of a third actin-like protein from rat, rat testis actin-like protein 3 (rTact3). The complete cDNA of the rTact3 gene was approximately 3.7 kb in length, and its corresponding amino acid sequence consisted of 1219 amino acids. The rTact3 gene lacks introns, similar to mTact1 and mTact2. The 356 C-terminal amino acids of rTact3 showed 43% homology with mTact1, whereas the 863 N-terminal amino acids did not show any significant homology. Northern blot analysis revealed that rTact3 mRNA was expressed only in adult rat testes and not during the prepubescent stage. In situ hybridization revealed that rTact3 was expressed exclusively during round and elongated spermatids maturation stages in rat testes. Immunohistochemical experiments using antibodies raised against a synthetic peptide showed that the expression of the rTact3 protein was also restricted in round and elongated spermatids, specifically in the head and acrosome of mature rat sperm. The 5′-flanking region of the mTact3 gene was found to contain a TATA-box motif as well as two putative CREB/c-Jun and five C/EBP motifs. mTact3 promoter activity was enhanced in a dose-dependent manner by the transfection of CREB, c-Jun, or C/EBP in NIH3T3 cells. These results suggest that Tact3 proteins might play an important role in rodent germ-cell development.
Collapse
|
16
|
Kierszenbaum AL, Rivkin E, Tres LL. Cytoskeletal track selection during cargo transport in spermatids is relevant to male fertility. SPERMATOGENESIS 2011; 1:221-230. [PMID: 22319670 DOI: 10.4161/spmg.1.3.18018] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 09/06/2011] [Indexed: 11/19/2022]
Abstract
Spermatids generate diverse and unusual actin and microtubule populations during spermiogenesis to fulfill mechanical and cargo transport functions assisted by motor and non-motor proteins. Disruption of cargo transport may lead to teratozoospermia and consequent male infertility. How motor and non-motor proteins utilize the cytoskeleton to transport cargos during sperm development is not clear. Filamentous actin (F-actin) and the associated motor protein myosin Va participate in the transport of Golgi-derived proacrosomal vesicles to the acrosome and along the manchette. The acrosome is stabilized by the acroplaxome, a cytoskeletal plate anchored to the nuclear envelope. The acroplaxome plate harbors F-actin and actin-like proteins as well as several other proteins, including keratin 5/Sak57, Ran GTPase, Hook1, dynactin p150Glued, cenexin-derived ODF2, testis-expressed profilin-3 and profilin-4, testis-expressed Fer tyrosine kinase (FerT), members of the ubiquitin-proteasome system and cortactin. Spermatids express transcripts encoding the non-spliced form of cortactin, a F-actin-regulatory protein. Tyrosine phosphorylated cortactin and FerT coexist in the acrosome-acroplaxome complex. Hook1 and p150Glued, known to participate in vesicle cargo transport, are sequentially seen from the acroplaxome to the manchette to the head-tail coupling apparatus (HTCA). The golgin Golgi-microtubule associated protein GMAP210 resides in the cis-Golgi whereas the intraflagellar protein IFT88 localizes in the trans-Golgi network. Like Hook1 and p150Glued, GMAP210 and IFT88 colocalize at the cytosolic side of proacrosomal vesicles and, following vesicle fusion, become part of the outer and inner acrosomal membranes before relocating to the acroplaxome, manchette and HTCA. A hallmark of the manchette and axoneme is microtubule heterogeneity, determined by the abundance of acetylated, tysosinated and glutamylated tubulin isoforms produced by post-translational modifications. We postulate that the construction of the male gamete requires microtubule and F-actin tracks and specific molecular motors and associated non-motor proteins for the directional positioning of vesicular and non-vesicular cargos at specific intracellular sites.
Collapse
Affiliation(s)
- Abraham L Kierszenbaum
- Department of Cell Biology and Anatomy; The Sophie Davis School of Biomedical Education; The City University of New York; New York, NY USA
| | | | | |
Collapse
|
17
|
Boëda B, Knowles PP, Briggs DC, Murray-Rust J, Soriano E, Garvalov BK, McDonald NQ, Way M. Molecular recognition of the Tes LIM2-3 domains by the actin-related protein Arp7A. J Biol Chem 2011; 286:11543-54. [PMID: 21278383 PMCID: PMC3064208 DOI: 10.1074/jbc.m110.171264] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 12/15/2010] [Indexed: 11/06/2022] Open
Abstract
Actin-related proteins (Arps) are a highly conserved family of proteins that have extensive sequence and structural similarity to actin. All characterized Arps are components of large multimeric complexes associated with chromatin or the cytoskeleton. In addition, the human genome encodes five conserved but largely uncharacterized "orphan" Arps, which appear to be mostly testis-specific. Here we show that Arp7A, which has 43% sequence identity with β-actin, forms a complex with the cytoskeletal proteins Tes and Mena in the subacrosomal layer of round spermatids. The N-terminal 65-residue extension to the actin-like fold of Arp7A interacts directly with Tes. The crystal structure of the 1-65(Arp7A)·LIM2-3(Tes)·EVH1(Mena) complex reveals that residues 28-49 of Arp7A contact the LIM2-3 domains of Tes. Two alanine residues from Arp7A that occupy equivalent apolar pockets in both LIM domains as well as an intervening GPAK linker that binds the LIM2-3 junction are critical for the Arp7A-Tes interaction. Equivalent occupied apolar pockets are also seen in the tandem LIM domain structures of LMO4 and Lhx3 bound to unrelated ligands. Our results indicate that apolar pocket interactions are a common feature of tandem LIM domain interactions, but ligand specificity is principally determined by the linker sequence.
Collapse
Affiliation(s)
- Batiste Boëda
- From the Cell Motility and
- the Cell Polarity and Migration Group, CNRS 2582, Institut Pasteur, 75724 Paris, France, and
| | - Phillip P. Knowles
- Structural Biology Laboratories, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, United Kingdom
| | - David C. Briggs
- Structural Biology Laboratories, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, United Kingdom
- the Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Judith Murray-Rust
- Structural Biology Laboratories, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, United Kingdom
| | - Erika Soriano
- Structural Biology Laboratories, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, United Kingdom
| | - Boyan K. Garvalov
- the Institute of Neuropathology, Justus Liebig University, Aulweg 123, 35392 Giessen, Germany
| | - Neil Q. McDonald
- Structural Biology Laboratories, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, United Kingdom
- the Institute of Structural and Molecular Biology, University College London and Birkbeck College, Malet Street, London WC1E 7HX, United Kingdom
| | | |
Collapse
|
18
|
Kierszenbaum AL, Rivkin E, Tres LL. Expression of Fer testis (FerT) tyrosine kinase transcript variants and distribution sites of FerT during the development of the acrosome-acroplaxome-manchette complex in rat spermatids. Dev Dyn 2009; 237:3882-91. [PMID: 18985748 DOI: 10.1002/dvdy.21789] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
We report the association of testicular Fer, a non-receptor tyrosine kinase, with acrosome development and remodeling of the acrosome-associated acroplaxome plate during spermatid head shaping. A single gene expresses two forms of Fer tyrosine kinases in testis: a somatic form (FerS) and a truncated testis-type form (FerT). FerT transcript variants are seen in spermatocytes and spermatids. FerS transcripts are not detected in round spermatids but are moderately transcribed in spermatocytes. FerT protein is associated with the spermatid medial/trans-Golgi region, proacrosomal vesicles, the cytosolic side of the outer acrosome membrane and adjacent to the inner acrosome membrane facing the acroplaxome. FerT coexist in the acroplaxome with phosphorylated cortactin, a regulator of F-actin dynamics. We propose that FerT participates in acrosome development and that phosphorylated cortactin may contribute to structural changes in F-actin in the acroplaxome during spermatid head shaping.
Collapse
Affiliation(s)
- Abraham L Kierszenbaum
- Department of Cell Biology and Anatomy, CUNY Medical School, Harris Hall Suite 306, 160 Convent Avenue, New York, NY 10031, USA.
| | | | | |
Collapse
|
19
|
Abstract
The spectrum of hereditary neuropathies has evolved recently as a result of the exponential growth of genetic research. For the purpose of this review, we will use Charcot-Marie-Tooth (CMT), hereditary liability to pressure palsy (HNPP) and hereditary sensory and autonomic neuropathies (HSAN) to illustrate the current clinical and genetic approach to such neuropathies.
Collapse
Affiliation(s)
- Safwan S Jaradeh
- From the Department of Neurology, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
20
|
Tanaka H, Iguchi N, Egydio de Carvalho C, Tadokoro Y, Yomogida K, Nishimune Y. Novel actin-like proteins T-ACTIN 1 and T-ACTIN 2 are differentially expressed in the cytoplasm and nucleus of mouse haploid germ cells. Biol Reprod 2003; 69:475-82. [PMID: 12672658 DOI: 10.1095/biolreprod.103.015867] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
We isolated cDNA clones for the novel actin-like proteins T-ACTIN 1 and T-ACTIN 2, which are expressed specifically in the mouse testis. These clones were from a subtracted cDNA library that was enriched for haploid germ cell-specific cDNAs. The mRNA sizes and deduced molecular masses of t-actin 1/mACTl7b and t-actin 2/mACTl7a were 2.2 kilobases (kb) and 1.8 kb, and Mr 43.1 x 10(3) and Mr 47.2 x 10(3), respectively. The two deduced amino acid sequences had 60% homology, and they had approximately 40% homology with other actins. The T-ACTINs contained some of the conserved regions seen in other actins. Although the cellular locations of these two proteins are quite different (T-ACTIN-1 was found in the cytoplasm and T-ACTIN-2 was located in the nucleus), the expression of their proteins and mRNAs is controlled during development and limited during spermiogenesis. In contrast, only T-ACTIN-2 was present in sperm heads and tails. These results suggest that T-ACTINs play important roles in sperm function and in the specific morphogenesis of spermatozoa during spermiogenesis.
Collapse
Affiliation(s)
- Hiromitsu Tanaka
- Department of Science for Laboratory Animal Experimentation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Chung KW, Ferrell RE, Ellis D, Barmada M, Moritz M, Finegold DN, Jaffe R, Vats A. African American hypertensive nephropathy maps to a new locus on chromosome 9q31-q32. Am J Hum Genet 2003; 73:420-9. [PMID: 12840782 PMCID: PMC1180380 DOI: 10.1086/377184] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2003] [Accepted: 05/28/2003] [Indexed: 12/17/2022] Open
Abstract
Hypertensive nephropathy (HN) and focal segmental glomerulosclerosis (FSGS) are significant causes of end-stage renal disease (ESRD), but no genes or loci have been associated with this phenotype among African Americans, a group at high risk. We performed a genomewide linkage scan with approximately 400 microsatellite markers on 23 individuals of a large four-generation African American family with 18 affected individuals (7 with ESRD), in which the 13-year-old proband (also with ESRD) presented with hypertension and proteinuria (2-4 g/day) and underwent a kidney biopsy that revealed FSGS-like lesions with arteriolar thickening. A genomewide scan revealed LOD scores of >2.5 for markers on chromosomes 3 and 9, and fine mapping was performed on 5 additional members (total 28 members) that showed a maximum multipoint LOD score of 5.4 in the 9q31-q32 region, under an autosomal dominant model with 99% penetrance. This 8-cM (6-Mb) region is flanked by markers D9S172 and D9S105, and further candidate gene sequencing studies excluded the coding regions of three genes (ACTL7A, ACTL7B, and CTNNAL1). To our knowledge, this is the first report of a locus, denoted as "HNP1," for the HN/FSGS phenotype in a large African American family with dominantly inherited nephropathy characterized by ESRD, hypertension, and some features of FSGS.
Collapse
Affiliation(s)
- Ki Wha Chung
- Departments of Pediatrics and Pathology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, and Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh
| | - Robert E. Ferrell
- Departments of Pediatrics and Pathology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, and Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh
| | - Demetrius Ellis
- Departments of Pediatrics and Pathology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, and Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh
| | - Michael Barmada
- Departments of Pediatrics and Pathology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, and Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh
| | - Michael Moritz
- Departments of Pediatrics and Pathology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, and Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh
| | - David N. Finegold
- Departments of Pediatrics and Pathology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, and Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh
| | - Ronald Jaffe
- Departments of Pediatrics and Pathology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, and Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh
| | - Abhay Vats
- Departments of Pediatrics and Pathology, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, and Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh
| |
Collapse
|
22
|
Hisano M, Yamada S, Tanaka H, Nishimune Y, Nozaki M. Genomic structure and promoter activity of the testis haploid germ cell-specific intronless genes, Tact1 and Tact2. Mol Reprod Dev 2003; 65:148-56. [PMID: 12704725 DOI: 10.1002/mrd.10276] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Tact1 and Tact2 genes, each of which encodes an actin-like protein, are exclusively expressed and translated in haploid germ cells in testis. To characterize the haploid germ cell-specific gene structure, a mouse genomic library was screened with a Tact1 cDNA as a probe, and four independent phage clones containing the Tact1 gene were isolated. Southern hybridization and sequencing analyses revealed that Tact1 and Tact2 were single copy genes contained on a common fragment in a head-to-head orientation, and that the distance between these genes was less than 2 kb. Comparison of the nucleotide sequences of genomic DNA and cDNA demonstrated that Tact1 and Tact2 lack introns, although all known actin or actin-related genes in mammals contain introns. Human Tact orthologues also lack introns and are located within 6.4 kb in a head-to-head orientation. These findings indicate that Tact1 and Tact2 or one of these genes arose by retroposition of a spliced mRNA transcribed from an actin progenitor gene prior to the divergence of rodents and primates. The Tact1 and Tact2 genes are unusual retroposons in that they have retained an open reading frame and are expressed in testicular germ cells, because almost all retroposons become pseudogenes. It was revealed that a 2kb sequence between the two genes bidirectionally controls haploid germ-cell specific expression by analyzing transgenic mice. Comparison of the murine Tact genes with their human orthologues showed a high level of identity between the two species in the 5'-upstream and non-coding sequences as well as in the coding region, indicating that conserved elements in these regions may be involved in the regulation of haploid germ cell-specific expression. The promoter region contains no TATA-, CCAAT- or GC-boxes, although there are potential cAMP response element (CRE)-like motifs in the 5'-upstream region and the 5'-untranslated region in Tact1 and Tact2, respectively. Transient promoter analyses indicate that CREMtau may activate Tact1 and Tact2 expression in germ cells.
Collapse
Affiliation(s)
- Mizue Hisano
- Department of Science for Laboratory Animal Experimentation, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | |
Collapse
|
23
|
Coutts AS, MacKenzie E, Griffith E, Black DM. TES is a novel focal adhesion protein with a role in cell spreading. J Cell Sci 2003; 116:897-906. [PMID: 12571287 DOI: 10.1242/jcs.00278] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previously, we identified TES as a novel candidate tumour suppressor gene that mapped to human chromosome 7q31.1. In this report we demonstrate that the TES protein is localised at focal adhesions, actin stress fibres and areas of cell-cell contact. TES has three C-terminal LIM domains that appear to be important for focal adhesion targeting. Additionally, the N-terminal region is important for targeting TES to actin stress fibres. Yeast two-hybrid and biochemical analyses yielded interactions with several focal adhesion and/or cytoskeletal proteins including mena, zyxin and talin. The fact that TES localises to regions of cell adhesion suggests that it functions in events related to cell motility and adhesion. In support of this, we demonstrate that fibroblasts stably overexpressing TES have an increased ability to spread on fibronectin.
Collapse
Affiliation(s)
- Amanda S Coutts
- Cancer Research UK Laboratories, Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | | | | | | |
Collapse
|
24
|
Osada N, Hida M, Kusuda J, Tanuma R, Hirata M, Suto Y, Hirai M, Terao K, Sugano S, Hashimoto K. Cynomolgus monkey testicular cDNAs for discovery of novel human genes in the human genome sequence. BMC Genomics 2002; 3:36. [PMID: 12498619 PMCID: PMC140308 DOI: 10.1186/1471-2164-3-36] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2002] [Accepted: 12/23/2002] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In order to contribute to the establishment of a complete map of transcribed regions of the human genome, we constructed a testicular cDNA library for the cynomolgus monkey, and attempted to find novel transcripts for identification of their human homologues. RESULT The full-insert sequences of 512 cDNA clones were determined. Ultimately we found 302 non-redundant cDNAs carrying open reading frames of 300 bp-length or longer. Among them, 89 cDNAs were found not to be annotated previously in the Ensembl human database. After searching against the Ensembl mouse database, we also found 69 putative coding sequences have no homologous cDNAs in the annotated human and mouse genome sequences in Ensembl. We subsequently designed a DNA microarray including 396 non-redundant cDNAs (with and without open reading frames) to examine the expression of the full-sequenced genes. With the testicular probe and a mixture of probes of 10 other tissues, 316 of 332 effective spots showed intense hybridized signals and 75 cDNAs were shown to be expressed very highly in the cynomolgus monkey testis, but not ubiquitously. CONCLUSIONS In this report, we determined 302 full-insert sequences of cynomolgus monkey cDNAs with enough length of open reading frames to discover novel transcripts as human homologues. Among 302 cDNA sequences, human homologues of 89 cDNAs have not been predicted in the annotated human genome sequence in the Ensembl. Additionally, we identified 75 dominantly expressed genes in testis among the full-sequenced clones by using a DNA microarray. Our cDNA clones and analytical results will be valuable resources for future functional genomic studies.
Collapse
Affiliation(s)
- Naoki Osada
- Division of Genetic Resources, National Institute of Infectious Diseases, 1-23-1 Toyama-cho, Shinjuku-ku, 162-8640, Japan
- Laboratory of human evolution, Depertment of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba, 277-8562, Japan
| | - Munetomo Hida
- Department of Genome Structure Analysis, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Jun Kusuda
- Division of Genetic Resources, National Institute of Infectious Diseases, 1-23-1 Toyama-cho, Shinjuku-ku, 162-8640, Japan
| | - Reiko Tanuma
- Division of Genetic Resources, National Institute of Infectious Diseases, 1-23-1 Toyama-cho, Shinjuku-ku, 162-8640, Japan
| | - Makoto Hirata
- Division of Genetic Resources, National Institute of Infectious Diseases, 1-23-1 Toyama-cho, Shinjuku-ku, 162-8640, Japan
| | - Yumiko Suto
- Laboratory of human evolution, Depertment of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba, 277-8562, Japan
| | - Momoki Hirai
- Laboratory of human evolution, Depertment of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba, 277-8562, Japan
| | - Keiji Terao
- Tsukuba Primate Center For Medical Science, National Institute of Infectious Diseases, Hachimandai-1, Tsukuba-shi, Ibaraki 305-0843, Japan
| | - Sumio Sugano
- Department of Genome Structure Analysis, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Katsuyuki Hashimoto
- Division of Genetic Resources, National Institute of Infectious Diseases, 1-23-1 Toyama-cho, Shinjuku-ku, 162-8640, Japan
| |
Collapse
|
25
|
Fujii T, Tamura K, Masai K, Tanaka H, Nishimune Y, Nojima H. Use of stepwise subtraction to comprehensively isolate mouse genes whose transcription is up-regulated during spermiogenesis. EMBO Rep 2002; 3:367-72. [PMID: 11943763 PMCID: PMC1084061 DOI: 10.1093/embo-reports/kvf073] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2001] [Revised: 02/21/2002] [Accepted: 02/21/2002] [Indexed: 11/14/2022] Open
Abstract
We report the isolation of 153 mouse genes whose expression is dramatically up-regulated during spermiogenesis. We used a novel variation of the subtractive hybridization technique called stepwise subtraction, wherein the subtraction process is systematically repeated in a stepwise manner. We named the genes thus identified as TISP genes (transcript induced in spermiogenesis). The transcription of 80 of these TISP genes is almost completely specific to the testis. This transcription is abruptly turned on after 17 days of age, when the mice enter puberty and spermiogenesis is initiated. Considering that the most advanced cells present at these stages of spermatogenesis are the spermatids, it is likely that we could isolate most of the spermatid-specific genes. DNA sequencing revealed that about half the TISP genes are novel and uncharacterized genes, confirming the utility of the stepwise subtraction approach for gene discovery.
Collapse
Affiliation(s)
- Takayuki Fujii
- Departments of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Cuajungco MP, Leyne M, Mull J, Gill SP, Gusella JF, Slaugenhaupt SA. Cloning, characterization, and genomic structure of the mouse Ikbkap gene. DNA Cell Biol 2001; 20:579-86. [PMID: 11747609 DOI: 10.1089/104454901317094990] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
Abstract
Our laboratory recently reported that mutations in the human I-kappaB kinase-associated protein (IKBKAP) gene are responsible for familial dysautonomia (FD). Interestingly, amino acid substitutions in the IKAP correlate with increased risk for childhood bronchial asthma. Here, we report the cloning and genomic characterization of the mouse Ikbkap gene, the homolog of human IKBKAP. Like its human counterpart, Ikbkap encodes a protein of 1332 amino acids with a molecular weight of approximately 150 kDa. The Ikbkap gene product, Ikap, contains 37 exons that span approximately 51 kb. The protein shows 80% amino acid identity with human IKAP. It shows very high conservation across species and is homologous to the yeast Elp1/Iki3p protein, which is a member of the Elongator complex. The Ikbkap gene maps to chromosome 4 in a region that is syntenic to human chromosome 9q31.3. Because no animal model of FD currently exists, cloning of the mouse Ikbkap gene is an important first step toward creating a mouse model for FD. In addition, cloning of Ikbkap is crucial to the characterization of the putative mammalian Elongator complex.
Collapse
Affiliation(s)
- M P Cuajungco
- Harvard Institute of Human Genetics and Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
27
|
Slaugenhaupt SA, Blumenfeld A, Gill SP, Leyne M, Mull J, Cuajungco MP, Liebert CB, Chadwick B, Idelson M, Reznik L, Robbins CM, Makalowska I, Brownstein MJ, Krappmann D, Scheidereit C, Maayan C, Axelrod FB, Gusella JF. Tissue-specific expression of a splicing mutation in the IKBKAP gene causes familial dysautonomia. Am J Hum Genet 2001; 68:598-605. [PMID: 11179008 PMCID: PMC1274473 DOI: 10.1086/318810] [Citation(s) in RCA: 423] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2000] [Accepted: 01/10/2001] [Indexed: 11/04/2022] Open
Abstract
Familial dysautonomia (FD; also known as "Riley-Day syndrome"), an Ashkenazi Jewish disorder, is the best known and most frequent of a group of congenital sensory neuropathies and is characterized by widespread sensory and variable autonomic dysfunction. Previously, we had mapped the FD gene, DYS, to a 0.5-cM region on chromosome 9q31 and had shown that the ethnic bias is due to a founder effect, with >99.5% of disease alleles sharing a common ancestral haplotype. To investigate the molecular basis of FD, we sequenced the minimal candidate region and cloned and characterized its five genes. One of these, IKBKAP, harbors two mutations that can cause FD. The major haplotype mutation is located in the donor splice site of intron 20. This mutation can result in skipping of exon 20 in the mRNA of patients with FD, although they continue to express varying levels of wild-type message in a tissue-specific manner. RNA isolated from lymphoblasts of patients is primarily wild-type, whereas only the deleted message is seen in RNA isolated from brain. The mutation associated with the minor haplotype in four patients is a missense (R696P) mutation in exon 19, which is predicted to disrupt a potential phosphorylation site. Our findings indicate that almost all cases of FD are caused by an unusual splice defect that displays tissue-specific expression; and they also provide the basis for rapid carrier screening in the Ashkenazi Jewish population.
Collapse
Affiliation(s)
- Susan A. Slaugenhaupt
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Anat Blumenfeld
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Sandra P. Gill
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Maire Leyne
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - James Mull
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Math P. Cuajungco
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Christopher B. Liebert
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Brian Chadwick
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Maria Idelson
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Luba Reznik
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Christiane M. Robbins
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Izabela Makalowska
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Michael J. Brownstein
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Daniel Krappmann
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Claus Scheidereit
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Channa Maayan
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - Felicia B. Axelrod
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| | - James F. Gusella
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown; Harvard Institute of Human Genetics, Harvard Medical School, Boston; Departments of Clinical Biochemistry and Pediatrics, Hadassah University Hospital, Jerusalem; Laboratory of Genetics, National Institute of Mental Health and National Human Genome Research Institute, and Genome Technology Branch, National Human Genome Research Institute, and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda; Max-Delbrück-Centrum for Molecular Medicine, Berlin; and Department of Pediatrics, New York University Medical Center, New York
| |
Collapse
|