1
|
Ma K, Yang X, Mao Y. Advancing evolutionary medicine with complete primate genomes and advanced biotechnologies. Trends Genet 2025; 41:201-217. [PMID: 39627062 DOI: 10.1016/j.tig.2024.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/03/2024] [Accepted: 11/06/2024] [Indexed: 03/06/2025]
Abstract
Evolutionary medicine, which integrates evolutionary biology and medicine, significantly enhances our understanding of human traits and disease susceptibility. However, previous studies in this field have often focused on single-nucleotide variants due to technological limitations in characterizing complex genomic regions, hindering the comprehensive analyses of their evolutionary origins and clinical significance. In this review, we summarize recent advancements in complete telomere-to-telomere (T2T), primate genomes and other primate resources, and illustrate how these resources facilitate the research of complex regions. We focus on several biomedically relevant regions to examine the relationship between primate genome evolution and human diseases. We also highlight the potentials of high-throughput functional genomic technologies for assessing candidate loci. Finally, we discuss future directions for primate research within the context of evolutionary medicine.
Collapse
Affiliation(s)
- Kaiyue Ma
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangyu Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Yafei Mao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Center for Genomic Research, International Institutes of Medicine, Fourth Affiliated Hospital, Zhejiang University, Yiwu, Zhejiang, China.
| |
Collapse
|
2
|
Lea G, Doria-Borrell P, Ferrero-Micó A, Varma A, Simon C, Anderson H, Biggins L, De Clercq K, Andrews S, Niakan KK, Gahurova L, McGovern N, Pérez-García V, Hanna CW. Ectopic expression of DNMT3L in human trophoblast stem cells restores features of the placental methylome. Cell Stem Cell 2025; 32:276-292.e9. [PMID: 39788122 DOI: 10.1016/j.stem.2024.12.007] [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: 05/03/2024] [Revised: 10/07/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025]
Abstract
The placental DNA methylation landscape is unique, with widespread partially methylated domains (PMDs). The placental "methylome" is conserved across mammals, a shared feature of many cancers, and extensively studied for links with pregnancy complications. Human trophoblast stem cells (hTSCs) offer exciting potential for functional studies to better understand this epigenetic feature; however, whether the hTSC epigenome recapitulates primary trophoblast remains unclear. We find that hTSCs exhibit an atypical methylome compared with trophectoderm and 1st trimester cytotrophoblast. Regardless of cell origin, oxygen levels, or culture conditions, hTSCs show localized DNA methylation within transcribed gene bodies and a complete loss of PMDs. Unlike early human trophoblasts, hTSCs display a notable absence of DNMT3L expression, which is necessary for PMD establishment in mouse trophoblasts. Remarkably, we demonstrate that ectopic expression of DNMT3L in hTSCs restores placental PMDs, supporting a conserved role for DNMT3L in de novo methylation in trophoblast development in human embryogenesis.
Collapse
Affiliation(s)
- Georgia Lea
- Loke Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | | | | | - Anakha Varma
- Loke Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Claire Simon
- Loke Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Holly Anderson
- Loke Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Laura Biggins
- Babraham Bioinformatics, Babraham Institute, Cambridge, UK
| | | | - Simon Andrews
- Babraham Bioinformatics, Babraham Institute, Cambridge, UK
| | - Kathy K Niakan
- Loke Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Lenka Gahurova
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Naomi McGovern
- Loke Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Vicente Pérez-García
- Centro de Investigación Príncipe Felipe, Valencia, Spain; Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.
| | - Courtney W Hanna
- Loke Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
3
|
Zhao Y, Liang Y, Cai L, Cai L, Huang B, Han P, Zhang X, Zhang H, Chen Z, Yin X, Duan P, Shou H, Zhu X, Wang Z, Wan Q, Huang J, Qian J. Comprehensive Proteomic Analysis Reveals Distinct Features and a Diagnostic Biomarker Panel for Early Pregnancy Loss in Histological Subtypes. Mol Cell Proteomics 2024; 23:100848. [PMID: 39321873 PMCID: PMC11541848 DOI: 10.1016/j.mcpro.2024.100848] [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: 04/10/2024] [Revised: 07/21/2024] [Accepted: 08/16/2024] [Indexed: 09/27/2024] Open
Abstract
Early pregnancy loss (EPL) is a common event in human reproduction and is classified into histological subtypes such as hydropic abortion (HA) and hydatidiform moles, including complete hydatidiform moles (CHMs) and partial hydatidiform moles (PHMs). However, accurate diagnosis and improved patient management remain challenging due to high rates of misdiagnosis and diverse prognostic risks. Therefore, diagnostic biomarkers for EPL are urgently needed. Our study aimed to identify biomarkers for EPL through comprehensive proteomic analysis. Ten CHMs, six PHMs, ten HAs, and 10 normal control products of conception were used to obtain a proteomic portrait. Parallel reaction monitoring-targeted proteomic and regression analyses were used to verify and select the diagnostic signatures. Finally, 14 proteins were selected and a panel of diagnostic classifiers (DLK1, SPTB/COL21A1, and SAR1A) was built to represent the CHM, PHM, and normal control groups (area under the receiver operating characteristic curve = 0.900, 0.804/0.885, and 0.991, respectively). This high diagnostic power was further validated in another independent cohort (n = 148) by immunohistochemistry (n = 120) and Western blot analyses (n = 28). The protein SPTB was selected for further biological behavior experiments in vitro. Our data suggest that SPTB maintains trophoblast cell proliferation, angiogenesis, cell motility, and the cytoskeleton network. This study provides a comprehensive proteomic portrait and identifies potential diagnostic biomarkers. These findings enhance our understanding of EPL pathogenesis and offer novel targets for diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Yating Zhao
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Yingjiqiong Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Zhejiang Province, PR China; Biomedical Big Data Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Luya Cai
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Limeng Cai
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Bo Huang
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Peilin Han
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Xiaofei Zhang
- Department of Pathology, The Zhejiang University School of Medicine Women's Hospital, Hangzhou, China
| | - Huifang Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhen Chen
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangang Yin
- Department of Diagnosis, Ningbo Clinical Pathology Diagnosis Center, Ningbo, China
| | - Ping Duan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huafeng Shou
- Department of Gynecology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Xiaoxu Zhu
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Zhe Wang
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Qihong Wan
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Jinyan Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Zhejiang Province, PR China; Biomedical Big Data Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China; Zhejiang University Cancer Center, Zhejiang University, Zhejiang Province, PR China.
| | - Jianhua Qian
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.
| |
Collapse
|
4
|
Friedrich C, Tüttelmann F. Genetics of female and male infertility. MED GENET-BERLIN 2024; 36:161-170. [PMID: 39253719 PMCID: PMC11380935 DOI: 10.1515/medgen-2024-2040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Infertility is defined as the inability to conceive within one year of unprotected intercourse, and the causes are equally distributed between both sexes. Genetics play a crucial role in couple infertility and respective diagnostic testing should follow available guidelines. Appropriate tiered genetic analyses require comprehensive physical examination of both partners in an infertile couple. A wide range of chromosomal and monogenic variants can be the underlying genetic cause of infertility in both women and men. Accurate clinical phenotyping, together with identification of the genetic origin, helps to recommend the proper treatment and to counsel couples on the success rates and potential risks for offspring.
Collapse
Affiliation(s)
- Corinna Friedrich
- University and University Hospital Münster Centre of Medical Genetics, Institute of Reproductive Genetics Vesaliusweg 12-14 48149 Münster Germany
| | - Frank Tüttelmann
- University and University Hospital Münster Centre of Medical Genetics, Institute of Reproductive Genetics Vesaliusweg 12-14 48149 Münster Germany
| |
Collapse
|
5
|
Bahutair SNM, Dube R, Kuruba MGB, Salama RAA, Patni MAMF, Kar SS, Kar R. Molecular Basis of Hydatidiform Moles-A Systematic Review. Int J Mol Sci 2024; 25:8739. [PMID: 39201425 PMCID: PMC11354253 DOI: 10.3390/ijms25168739] [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: 07/06/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Gestational trophoblastic diseases (GTDs) encompass a spectrum of conditions characterized by abnormal trophoblastic cell growth, ranging from benign molar pregnancies to malignant trophoblastic neoplasms. This systematic review explores the molecular underpinnings of GTDs, focusing on genetic and epigenetic factors that influence disease progression and clinical outcomes. Based on 71 studies identified through systematic search and selection criteria, key findings include dysregulations in tumor suppressor genes such as p53, aberrant apoptotic pathways involving BCL-2 (B-cell lymphoma), and altered expression of growth factor receptors and microRNAs (micro-ribose nucleic acid). These molecular alterations not only differentiate molar pregnancies from normal placental development but also contribute to their clinical behavior, from benign moles to potentially malignant forms. The review synthesizes insights from immunohistochemical studies and molecular analyses to provide a comprehensive understanding of GTD pathogenesis and implications for personalized care strategies.
Collapse
Affiliation(s)
- Shadha Nasser Mohammed Bahutair
- Department of Obstetrics and Gynecology, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Rajani Dube
- Department of Obstetrics and Gynecology, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Manjunatha Goud Bellary Kuruba
- Department of Biochemistry, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Rasha Aziz Attia Salama
- Department of Community Medicine, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates; (R.A.A.S.); (M.A.M.F.P.)
- Department of Public Health and Community Medicine, Kasr El Ainy Faculty of Medicine, Cairo University, Cairo 12613, Egypt
| | - Mohamed Anas Mohamed Faruk Patni
- Department of Community Medicine, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates; (R.A.A.S.); (M.A.M.F.P.)
| | - Subhranshu Sekhar Kar
- Department of Pediatrics, RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Rakhee Kar
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry 605006, India;
| |
Collapse
|
6
|
Eggermann T. Human Reproduction and Disturbed Genomic Imprinting. Genes (Basel) 2024; 15:163. [PMID: 38397153 PMCID: PMC10888310 DOI: 10.3390/genes15020163] [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: 01/02/2024] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Genomic imprinting is a specific mode of gene regulation which particularly accounts for the factors involved in development. Its disturbance affects the fetus, the course of pregnancy and even the health of the mother. In children, aberrant imprinting signatures are associated with imprinting disorders (ImpDis). These alterations also affect the function of the placenta, which has consequences for the course of the pregnancy. The molecular causes of ImpDis comprise changes at the DNA level and methylation disturbances (imprinting defects/ImpDefs), and there is an increasing number of reports of both pathogenic fetal and maternal DNA variants causing ImpDefs. These ImpDefs can be inherited, but prediction of the pregnancy complications caused is difficult, as they can cause miscarriages, aneuploidies, health issues for the mother and ImpDis in the child. Due to the complexity of imprinting regulation, each pregnancy or patient with suspected altered genomic imprinting requires a specific workup to identify the precise molecular cause and also careful clinical documentation. This review will cover the current knowledge on the molecular causes of aberrant imprinting signatures and illustrate the need to identify this basis as the prerequisite for personalized genetic and reproductive counselling of families.
Collapse
Affiliation(s)
- Thomas Eggermann
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH University Aachen, Pauwelsstr. 3, D-52074 Aachen, Germany
| |
Collapse
|
7
|
Florea A, Caba L, Grigore AM, Antoci LM, Grigore M, Gramescu MI, Gorduza EV. Hydatidiform Mole-Between Chromosomal Abnormality, Uniparental Disomy and Monogenic Variants: A Narrative Review. Life (Basel) 2023; 13:2314. [PMID: 38137915 PMCID: PMC10744706 DOI: 10.3390/life13122314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
A hydatidiform mole (HM) or molar pregnancy is the most common benign form of gestational trophoblastic disease characterized by a proliferation of the trophoblastic epithelium and villous edema. Hydatidiform moles are classified into two forms: complete and partial hydatidiform moles. These two types of HM present morphologic, histopathologic and cytogenetic differences. Usually, hydatidiform moles are a unique event, but some women present a recurrent form of complete hydatidiform moles that can be sporadic or familial. The appearance of hydatidiform moles is correlated with some genetic events (like uniparental disomy, triploidy or diandry) specific to meiosis and is the first step of embryo development. The familial forms are determined by variants in some genes, with NLRP7 and KHDC3L being the most important ones. The identification of different types of hydatidiform moles and their subsequent mechanisms is important to calculate the recurrence risk and estimate the method of progression to a malign form. This review synthesizes the heterogeneous mechanisms and their implications in genetic counseling.
Collapse
Affiliation(s)
- Andreea Florea
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.F.); (A.-M.G.); (L.-M.A.); (M.I.G.); (E.V.G.)
| | - Lavinia Caba
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.F.); (A.-M.G.); (L.-M.A.); (M.I.G.); (E.V.G.)
| | - Ana-Maria Grigore
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.F.); (A.-M.G.); (L.-M.A.); (M.I.G.); (E.V.G.)
| | - Lucian-Mihai Antoci
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.F.); (A.-M.G.); (L.-M.A.); (M.I.G.); (E.V.G.)
| | - Mihaela Grigore
- Department of Obstetrics and Gynecology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Mihaela I. Gramescu
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.F.); (A.-M.G.); (L.-M.A.); (M.I.G.); (E.V.G.)
| | - Eusebiu Vlad Gorduza
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.F.); (A.-M.G.); (L.-M.A.); (M.I.G.); (E.V.G.)
| |
Collapse
|
8
|
Salima S, Wibowo MH, Dewayani BM, Nisa AS, Alkaff FF. Recurrent Partial Hydatidiform Mole: A Case Report of Seven Consecutive Molar Pregnancies. Int J Womens Health 2023; 15:1239-1244. [PMID: 37554967 PMCID: PMC10406109 DOI: 10.2147/ijwh.s421386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
Hydatidiform mole (HM) is an aberrant pregnancy characterized by atypical trophoblastic hyperplasia, hydropic chorionic villi, and deprived fetal development. There are two types of HM, ie, complete (CHM) and partial (PHM). Both CHM and PHM can recur; however, the recurrence of PHM is very scarce compared to CHM. In this report, we present a case of a 33-year-old woman with recurrent PHM for 7 times without any normal pregnancy in-between. PHM was determined by histology examination. The patient underwent suction curettage and was followed up with serial β-hCG levels. Recurrent PHM, although rare, is associated with an increased incidence of malignancy. A series of clinical and β-hCG evaluation should be warranted because of the possibility of gestational trophoblastic neoplasia development.
Collapse
Affiliation(s)
- Siti Salima
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, Indonesia
| | - Mulyohadi Hadi Wibowo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, Indonesia
| | - Birgitta M Dewayani
- Department of Pathology of Anatomy, Faculty of Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, Indonesia
| | - Aisyah Shofiatun Nisa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, Indonesia
| | - Firas Farisi Alkaff
- Division of Pharmacology and Therapy, Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
9
|
Musfee FI, Oluwafemi OO, Agopian A, Hakonarson H, Goldmuntz E, Mitchell LE. Maternal Effect Genes as Risk Factors for Congenital Heart Defects. HGG ADVANCES 2022; 3:100098. [PMID: 35345810 PMCID: PMC8957044 DOI: 10.1016/j.xhgg.2022.100098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/04/2022] [Indexed: 11/09/2022] Open
Abstract
Maternal effect genes (MEGs) encode factors (e.g., RNA) in the oocyte that control embryonic development prior to activation of the embryonic genome. Over 80 mammalian MEGs have been identified, including several that have been associated with phenotypes in humans. Maternal variation in MEGs is associated with a range of adverse outcomes, which, in humans, include hydatidiform moles, zygotic cleavage failure, and offspring with multi-locus imprinting disorders. In addition, data from both animal models and humans suggest that the MEGs may be associated with structural birth defects such as congenital heart defects (CHDs). To further investigate the association between MEGs and CHDs, we conducted gene-level and gene-set analyses of known mammalian MEGs (n = 82) and two common groups of CHDs: conotruncal heart defects and left ventricular outflow tract defects. We identified 14 candidate CHD-related MEGs. These 14 MEGs include three (CDC20, KHDC3L, and TRIP13) of the 11 known human MEGs, as well as one (DNMT3A) of the eight MEGs that have been associated with structural birth defects in animal models. Our analyses add to the growing evidence that MEGs are associated with structural birth defects, in particular CHDs. Given the large proportion of individuals with structural birth defects for whom etiology of their condition is unknown, further investigations of MEGs as potential risk factors for structural birth defects are strongly warranted.
Collapse
|
10
|
Picchetta L, Caroselli S, Figliuzzi M, Cogo F, Zambon P, Costa M, Pergher I, Patassini C, Cortellessa F, Zuccarello D, Poli M, Capalbo A. Molecular tools for the genomic assessment of oocyte’s reproductive competence. J Assist Reprod Genet 2022; 39:847-860. [PMID: 35124783 PMCID: PMC9050973 DOI: 10.1007/s10815-022-02411-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
The most important factor associated with oocytes' developmental competence has been widely identified as the presence of chromosomal abnormalities. However, growing application of genome-wide sequencing (GS) in population diagnostics has enabled the identification of multifactorial genetic predispositions to sub-lethal pathologies, including those affecting IVF outcomes and reproductive fitness. Indeed, GS analysis in families with history of isolated infertility has recently led to the discovery of new genes and variants involved in specific human infertility endophenotypes that impact the availability and the functionality of female gametes by altering unique mechanisms necessary for oocyte maturation and early embryo development. Ongoing advancements in analytical and bioinformatic pipelines for the study of the genetic determinants of oocyte competence may provide the biological evidence required not only for improving the diagnosis of isolated female infertility but also for the development of novel preventive and therapeutic approaches for reproductive failure. Here, we provide an updated discussion and review of the progresses made in preconception genomic medicine in the identification of genetic factors associated with oocyte availability, function, and competence.
Collapse
|
11
|
Abi Nahed R, Elkhoury Mikhael M, Reynaud D, Collet C, Lemaitre N, Michy T, Hoffmann P, Sergent F, Marquette C, Murthi P, Raia-Barjat T, Alfaidy N, Benharouga M. Role of NLRP7 in Normal and Malignant Trophoblast Cells. Biomedicines 2022; 10:252. [PMID: 35203462 PMCID: PMC8868573 DOI: 10.3390/biomedicines10020252] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/04/2022] [Accepted: 01/19/2022] [Indexed: 02/04/2023] Open
Abstract
Gestational choriocarcinoma (CC) is an aggressive cancer that develops upon the occurrence of abnormal pregnancies such as Hydatidiform moles (HMs) or upon non-molar pregnancies. CC cells often metastasize in multiple organs and can cause maternal death. Recent studies have established an association between recurrent HMs and mutations in the Nlrp7 gene. NLRP7 is a member of a new family of proteins that contributes to innate immune processes. Depending on its level of expression, NLRP7 can function in an inflammasome-dependent or independent pathway. To date, the role of NLRP7 in normal and in malignant human placentation remains to be elucidated. We have recently demonstrated that NLRP7 is overexpressed in CC trophoblast cells and may contribute to their acquisition of immune tolerance via the regulation of key immune tolerance-associated factors, namely HLA family, βCG and PD-L1. We have also demonstrated that NLRP7 increases trophoblast proliferation and decreases their differentiation, both in normal and tumor conditions. Actual findings suggest that NLRP7 expression may ensure a strong tolerance of the trophoblast by the maternal immune system during normal pregnancy and may directly affect the behavior and aggressiveness of malignant trophoblast cells. The proposed review summarizes recent advances in the understanding of the significance of NLRP7 overexpression in CC and discusses its multifaceted roles, including its function in an inflammasome-dependent or independent pathways.
Collapse
Affiliation(s)
- Roland Abi Nahed
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Maya Elkhoury Mikhael
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
| | - Deborah Reynaud
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Constance Collet
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Nicolas Lemaitre
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Thierry Michy
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Pascale Hoffmann
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Frederic Sergent
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Christel Marquette
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Padma Murthi
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia;
- Department of Obstetrics and Gynecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Tiphaine Raia-Barjat
- Department of Gynecology and Obstetrics, University Hospital, 42100 Saint Etienne, France;
| | - Nadia Alfaidy
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Mohamed Benharouga
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38054 Grenoble, France; (R.A.N.); (M.E.M.); (D.R.); (C.C.); (N.L.); (T.M.); (P.H.); (F.S.); (C.M.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| |
Collapse
|
12
|
Abstract
Maternal effect genes (MEGs) encode factors (e.g., RNA) that are present in the oocyte and required for early embryonic development. Hence, while these genes and gene products are of maternal origin, their phenotypic consequences result from effects on the embryo. The first mammalian MEGs were identified in the mouse in 2000 and were associated with early embryonic loss in the offspring of homozygous null females. In humans, the first MEG was identified in 2006, in women who had experienced a range of adverse reproductive outcomes, including hydatidiform moles, spontaneous abortions, and stillbirths. Over 80 mammalian MEGs have subsequently been identified, including several that have been associated with phenotypes in humans. In general, pathogenic variants in MEGs or the absence of MEG products are associated with a spectrum of adverse outcomes, which in humans range from zygotic cleavage failure to offspring with multi-locus imprinting disorders. Although less established, there is also evidence that MEGs are associated with structural birth defects (e.g., craniofacial malformations, congenital heart defects). This review provides an updated summary of mammalian MEGs reported in the literature through early 2021, as well as an overview of the evidence for a link between MEGs and structural birth defects.
Collapse
|
13
|
Xing D, Miller K, Beierl K, Ronnett BM. Loss of p57 Expression in Conceptions Other Than Complete Hydatidiform Mole: A Case Series With Emphasis on the Etiology, Genetics, and Clinical Significance. Am J Surg Pathol 2022; 46:18-32. [PMID: 34074808 PMCID: PMC9171551 DOI: 10.1097/pas.0000000000001749] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Combined p57 immunohistochemistry and DNA genotyping refines classification of products of conception specimens into specific types of hydatidiform moles and various nonmolar entities that can simulate them. p57 expression is highly correlated with genotyping and in practice can reliably be used to identify virtually all complete hydatidiform moles (CHM), but aberrant retained or lost p57 expression in rare CHMs and partial hydatidiform moles (PHM), as well as loss in some nonmolar abortuses, has been reported. Among a series of 2329 products of conceptions, we identified 10 cases for which loss of p57 expression was inconsistent with genotyping results (none purely androgenetic). They displayed a spectrum of generally mild abnormal villous morphology but lacked better developed features of CHMs/early CHMs, although some did suggest subtle forms of the latter. For 5 cases, genotyping (4 cases) and/or ancillary testing (1 case) determined a mechanism for the aberrant p57 results. These included 3 PHMs-2 diandric triploid and 1 triandric tetraploid-and 1 nonmolar specimen with loss of p57 expression attributable to partial or complete loss of the maternal copy of chromosome 11 and 1 nonmolar specimen with Beckwith-Wiedemann syndrome. For 5 cases, including 2 diandric triploid PHMs and 3 biparental nonmolar specimens, genotyping did not identify a mechanism, likely due to other genetic alterations which are below the resolution of or not targeted by genotyping. While overdiagnosis of a PHM as a CHM may cause less harm since appropriate follow-up with serum β-human chorionic gonadotropin levels would take place for both diagnoses, this could cause longer than necessary follow-up due to the expectation of a much greater risk of persistent gestational trophoblastic disease for CHM compared with PHM, which would be unfounded for the correct diagnosis of PHM. Overdiagnosis of a nonmolar abortus with loss of p57 expression as a CHM would lead to unnecessary follow-up and restriction on pregnancy attempts for patients with infertility. Genotyping is valuable for addressing discordance between p57 expression and morphology but cannot elucidate certain mechanisms of lost p57 expression. Future studies are warranted to determine whether chromosomal losses or gains, particularly involving imprinted genes such as p57, might play a role in modifying the risk of persistent gestational trophoblastic disease for PHMs and nonmolar conceptions that are not purely androgenetic but have some abnormal paternal imprinting of the type seen in CHMs.
Collapse
Affiliation(s)
- Deyin Xing
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Karin Miller
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Katie Beierl
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Brigitte M. Ronnett
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
14
|
Wenk RE, Peterson J, Baird M. A molecular classification of moles and its use in filiation tests. J Forensic Sci 2021; 67:328-331. [PMID: 34596233 DOI: 10.1111/1556-4029.14890] [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: 07/12/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 11/28/2022]
Abstract
Pregnancies, including ones that follow sexual assaults, occasionally produce hydatidiform moles. The alleged fathers (AFs) of moles have been tested for paternity by identifying the mole's locus phenotype-the one or two visible paternal obligate alleles (POAs) per locus. The probability that the mole inherited the POAs from the AF was divided by the probability that the mole inherited the POAs from a random man. This likelihood ratio (LR) would increase if the mole's specific genotype was known. Moles are generated in five different ways that produce five distinct genotypes. Examining a mole's multilocus STR profile reveals a mole's pathogenesis, determines locus genotypes, and increases paternity LRs.
Collapse
Affiliation(s)
- Robert E Wenk
- Forensics Division, DNA Diagnostics Corporation, Fairfield, Ohio, USA
| | - John Peterson
- Forensics Division, DNA Diagnostics Corporation, Fairfield, Ohio, USA
| | - Michael Baird
- Forensics Division, DNA Diagnostics Corporation, Fairfield, Ohio, USA
| |
Collapse
|
15
|
Genotyping diagnosis of gestational trophoblastic disease: frontiers in precision medicine. Mod Pathol 2021; 34:1658-1672. [PMID: 34088998 DOI: 10.1038/s41379-021-00831-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/28/2022]
Abstract
Investigations in recent decades have exploited tissue DNA genotyping as a powerful ancillary tool for the precision diagnosis and subclassification of gestational trophoblastic disease. As lesions of gestational origin, the inherited paternal genome, with or without copy number alterations, is the fundamental molecular basis for the diagnostic applications of DNA genotyping. Genotyping is now considered the gold standard in the confirmation and subtyping of sporadic hydatidiform moles. Although a precise diagnosis of partial mole requires DNA genotyping, prognostic stratification according to distinct genetic zygosity in complete moles has recently gained significant clinical relevance for patient care. Beyond hydatidiform moles, DNA genotyping has fundamental applications in the diagnosis or prognostic assessment of gestational trophoblastic tumors, in particular gestational choriocarcinoma. DNA genotyping provides a decisive tool in the separation of gestational trophoblastic neoplasia from non-gestational counterparts/mimics of either germ cell or somatic origin. The FIGO/WHO prognostic scoring scheme requires ascertaining the precise index gestational event and the time interval between the tumor and index gestation, where DNA genotyping can provide highly relevant information. With rapid acquisition of molecular diagnostic capabilities in the clinical practice, DNA genotyping has become closely integrated into the routine diagnostic workup of various forms of gestational trophoblastic disease.
Collapse
|
16
|
Kopelman ZA, Hope ER. High-Risk Gestational Trophoblastic Neoplasia from a Homozygous NLRP7 Mutation. Gynecol Oncol Rep 2021; 37:100803. [PMID: 34189227 PMCID: PMC8215180 DOI: 10.1016/j.gore.2021.100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/18/2021] [Accepted: 05/31/2021] [Indexed: 11/15/2022] Open
Abstract
NLRP7 germline mutation can result in high risk gestational trophoblastic neoplasia. No successful reproductive outcomes have been reported with homozygous NLRP7 mutation. Germline testing should be considered for patients presenting with recurrent gestational trophoblastic disease. Once an NLRP7 mutation is diagnosed, consultation with reproductive endocrinology is necessary to discuss future fertility. Further research is needed in rare cases regarding gestational trophoblastic neoplasia recurrence and reproductive outcomes.
Collapse
Affiliation(s)
- Zachary A Kopelman
- Department of Obstetrics and Gynecology, Brooke Army Medical Center, 3551 Roger Brooke Drive, Fort Sam Houston, TX 78234, USA
| | - Erica R Hope
- Department of Obstetrics and Gynecology, Brooke Army Medical Center, 3551 Roger Brooke Drive, Fort Sam Houston, TX 78234, USA
| |
Collapse
|
17
|
Xing D, Adams E, Huang J, Ronnett BM. Refined diagnosis of hydatidiform moles with p57 immunohistochemistry and molecular genotyping: updated analysis of a prospective series of 2217 cases. Mod Pathol 2021; 34:961-982. [PMID: 33024305 DOI: 10.1038/s41379-020-00691-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023]
Abstract
Immunohistochemical analysis of p57 expression and molecular genotyping accurately subclassify molar specimens into complete hydatidiform mole (CHM) and partial hydatidiform mole (PHM) and distinguish these from nonmolar specimens. Characteristics of a prospective series of potentially molar specimens analyzed in a large gynecologic pathology practice are summarized. Of 2217 cases (2160 uterine, 57 ectopic), 2080 (94%) were successfully classified: 571 CHMs (570 uterine, 1 ectopic), 498 PHMs (497 uterine, 1 ectopic), 900 nonmolar (including 147 trisomies, 19 digynic triploids, and 4 donor egg conceptions), and 56 androgenetic/biparental mosaics; 137 were complex or unsatisfactory and not definitively classified. CHMs dominated in patients aged < 21 and >45 years and were the only kind of molar conception found in the latter group. Of 564 successfully immunostained CHMs, 563 (99.8%) were p57-negative (1 p57-positive [retained maternal chromosome 11] androgenetic by genotyping). Of 153 genotyped CHMs, 148 (96.7%) were androgenetic (85% monospermic) and 5 were biparental, the latter likely familial biparental hydatidiform moles. Of 486 successfully immunostained PHMs, 481 (99%) were p57-positive (3 p57-negative [loss of maternal chromosome 11], 2 unknown mechanism). Of 497 genotyped PHMs, 484 (97%) were diandric triploid (99% dispermic) and 13 were triandric tetraploid (all at least dispermic). Of 56 androgenetic/biparental mosaics, 37 had a p57-negative complete molar component (16 confirmed as androgenetic by genotyping). p57 expression is highly correlated with genotyping, serving as a reliable marker for CHMs, and identifies molar components and androgenetic cell lines in mosaic conceptions. Correlation of morphology, p57 expression, genotyping data, and history are required to recognize familial biparental hydatidiform moles and donor egg conceptions, as the former can be misclassified as nonmolar and the latter can be misclassified as dispermic CHM on the basis of isolated genotyping results.
Collapse
Affiliation(s)
- Deyin Xing
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Emily Adams
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Jialing Huang
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Brigitte M Ronnett
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA.
- Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
18
|
The genetics of recurrent hydatidiform moles in Mexico: further evidence of a strong founder effect for one mutation in NLRP7 and its widespread. J Assist Reprod Genet 2021; 38:1879-1886. [PMID: 33751332 DOI: 10.1007/s10815-021-02132-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To investigate the frequency of a founder mutation in NLRP7, L750V, in independent cohorts of Mexican patients with recurrent hydatidiform moles (RHMs). METHODS Mutation analysis was performed by Sanger sequencing on DNA from 44 unrelated Mexican patients with RHMs and seven molar tissues from seven additional unrelated patients. RESULTS L750V was present in homozygous or heterozygous state in 37 (86%) patients and was transmitted on the same haplotype to patients from different states of Mexico. We also identified a second founder mutation, c.2810+2T>G in eight (18.1%) patients, and a novel premature stop-codon mutation W653*. CONCLUSION Our data confirm the strong founder effect for L750V, which appears to be the most common mutation in NLRP7. We also report on six healthy live births to five patients with biallelic NLRP7 mutations, two from spontaneous conceptions and four from donated ovum and discuss our recommendations for DNA testing and genetic counseling.
Collapse
|
19
|
Joneborg U, Coopmans L, van Trommel N, Seckl M, Lok CAR. Fertility and pregnancy outcome in gestational trophoblastic disease. Int J Gynecol Cancer 2021; 31:399-411. [PMID: 33649007 DOI: 10.1136/ijgc-2020-001784] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/01/2020] [Indexed: 12/28/2022] Open
Abstract
The aim of this review is to provide an overview of existing literature and current knowledge on fertility rates and reproductive outcomes after gestational trophoblastic disease. A systematic literature search was performed to retrieve all available studies on fertility rates and reproductive outcomes after hydatidiform mole pregnancy, low-risk gestational trophoblastic neoplasia, high- and ultra-high-risk gestational trophoblastic neoplasia, and the rare placental site trophoblastic tumor and epithelioid trophoblastic tumor forms of gestational trophoblastic neoplasia. The effects of single-agent chemotherapy, multi-agent including high-dose chemotherapy, and immunotherapy on fertility, pregnancy wish, and pregnancy outcomes were evaluated and summarized. After treatment for gestational trophoblastic neoplasia, most, but not all, women want to achieve another pregnancy. Age and extent of therapy determine if there is a risk of loss of fertility. Single-agent treatment does not affect fertility and subsequent pregnancy outcome. Miscarriage occurs more often in women who conceive within 6 months of follow-up after chemotherapy. Multi-agent chemotherapy hastens the natural menopause by three years and commonly induces a temporary amenorrhea, but in young women rarely causes permanent ovarian failure or infertility. Subsequent pregnancies have a high chance of ending with live healthy babies. In contrast, high-dose chemotherapy typically induces permanent amenorrhea, and no pregnancies have been reported after high-dose chemotherapy for gestational trophoblastic neoplasia. Immunotherapy is promising and may give better outcomes than multiple schedules of chemotherapy or even high-dose chemotherapy. The first pregnancy after immunotherapy has recently been described. Data on fertility-sparing treatment in placental site trophoblastic tumor and epithelioid trophoblastic tumor are still scarce, and this option should be offered with caution. In general, patients with gestational trophoblastic neoplasia may be reassured about their future fertility and pregnancy outcome. Detailed registration of high-risk gestational trophoblastic neoplasia is still indispensable to obtain more complete data to better inform patients in the future.
Collapse
Affiliation(s)
- Ulrika Joneborg
- Department of Pelvic Cancer, Karolinska University Hospital, Karolinska Institute Department of Women's and Children's Health, Stockholm, Sweden
| | - Leonoor Coopmans
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Amsterdam, Noord-Holland, The Netherlands
| | - Nienke van Trommel
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Amsterdam, Noord-Holland, The Netherlands
| | - Michael Seckl
- Department of Medical Oncology, Hammersmith Hospitals; Imperial College London, London, Pennsylvania, UK
| | - Christianne A R Lok
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Amsterdam, Noord-Holland, The Netherlands
| |
Collapse
|
20
|
Wu Q, Chen L, Song Y, Zhu J, Chen Y, Sun P. Expression pattern Nanog is associated with the development of gestational trophoblastic neoplasia in Chinese women from Fujian province: a case-control study. Gynecol Endocrinol 2021; 37:108-112. [PMID: 32584203 DOI: 10.1080/09513590.2020.1781080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
BACKGROUND Gestational trophoblastic disease (GTD) is a group of interrelated but distinct diseases and has a serious impact on the reproductive health of women. To analyze the expression of Nanog in GTD and to evaluate its potential to predict the development of gestational trophoblastic neoplasia (GTN). METHODS The study included 41 normal first-trimester placentas matched by gestational age to 53 regressed-hydatidiform-moles (rHMs), 56 malignant-HMs (mHMs) and 17 choriocarcinomas (CCAs) and evaluated the Nanog expression by immunohistochemistry. The chi-square test, ANOVA, Fisher's exact test and logistic regression were performed to assess the Nanog expression and clinical prognostic factors in GTD. RESULTS Compared to normal placenta levels, the Nanog expression was increased in GTD samples (p < .05). In HMs, Nanog expression was positively correlated with serum β-hCG levels,uterine size and theca-lutein cysts (p < .05). Compared with the low-risk metastatic group (Federation of Gynecology and Obstetrics (FIGO) score ≤ 6), the high-risk metastatic group (FIGO score >7) had higher Nanog expression (p = .030). Moreover, logistic regression analysis showed that the positive expression of Nanog had the highest risk of developing into GTN (OR = 4.764, p < .001). CONCLUSIONS Nanog is an independent predictor of clinical outcomes. It can also be a reliable predictor for GTN development from GTD.
Collapse
Affiliation(s)
- Qibin Wu
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lihua Chen
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yiyi Song
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jianfang Zhu
- Department of Pathology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yaojia Chen
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Pengming Sun
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
21
|
Saito MK. Elucidation of the Pathogenesis of Autoinflammatory Diseases Using iPS Cells. CHILDREN-BASEL 2021; 8:children8020094. [PMID: 33535645 PMCID: PMC7912798 DOI: 10.3390/children8020094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/20/2021] [Accepted: 01/29/2021] [Indexed: 11/22/2022]
Abstract
Autoinflammatory diseases are a disease entity caused by the dysregulation of innate immune cells. Typical autoinflammatory diseases are monogenic disorders and often very rare. As a result, there is a relative lack of understanding of the pathogenesis, poor diagnosis and little available treatment. Induced pluripotent stem (iPS) cells are a new technology being applied to in vitro disease modeling. These models are especially useful for the analysis of rare and intractable diseases including autoinflammatory diseases. In this review, I will provide a general overview of iPS cell models for autoinflammatory diseases and a brief description of the results obtained from individual reports.
Collapse
Affiliation(s)
- Megumu K Saito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 6068507, Japan
| |
Collapse
|
22
|
Capozzi VA, Butera D, Armano G, Monfardini L, Gaiano M, Gambino G, Sozzi G, Merisio C, Berretta R. Obstetrics outcomes after complete and partial molar pregnancy: Review of the literature and meta-analysis. Eur J Obstet Gynecol Reprod Biol 2021; 259:18-25. [PMID: 33550107 DOI: 10.1016/j.ejogrb.2021.01.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 01/25/2023]
Abstract
The hydatidiform mole is a rare gynecological disease rising from the trophoblastic. Post-molar pregnancies have an extremely variable course, varying from repeated abortions, stillbirths, preterm births, live births, or recurring in further molar pregnancies. Literature on obstetric outcomes following molar pregnancy is poor, often including monocentric studies, and with data collected from national databases. This review and meta-analysis aim to analyze the obstetric outcomes after conservative management of complete (CHM) and partial (PHM) molar pregnancies. The meta-analysis was performed following the Meta-analysis Of Observational Studies in Epidemiology (MOOSE) and the preferred reporting items for systematic reviews and meta-analyses statement (PRISMA). Six studies met the inclusion. Of the total 25,222 patients, 13,129 complete (52.1 %) and 12,093 partial (47.9 %) molar pregnancies were included. Live births rate after CHM was statistically higher (p = 0.002) compared to the live births after PHM (53.6 % vs. 51.0 %, 3266 vs. 1807 cases, respectively). Studies showed heterogeneity I2 = 57.7 %, pooled proportion = 0.2 %, and 95 % Confidence Interval (CI) 0.6 to 0.9. No statistically significant difference was demonstrated for ectopic pregnancies (p = 0.633), miscarriage (p = 0.637), preterm birth (p = 0.865), stillbirth (p = 0.911), termination of pregnancy (p = 0.572), and complete molar recurrence (p = 0.580) after CHM and PHM. Partial molar recurrence occurred more frequently after PHM than CHM (0.4 % vs. 0.3 %, 52 vs. 37 cases, respectively, p = 0.002). Careful counseling on the obstetric subsequent pregnancies outcomes should be provided to patients eager for further pregnancy and further studies are needed to confirm these results.
Collapse
Affiliation(s)
- Vito Andrea Capozzi
- Department of Gynecology and Obstetrics, University of Parma, Via Gramsci, 43125, Parma, Italy.
| | - Diana Butera
- Department of Gynecology and Obstetrics, University of Parma, Via Gramsci, 43125, Parma, Italy
| | - Giulia Armano
- Department of Gynecology and Obstetrics, University of Parma, Via Gramsci, 43125, Parma, Italy
| | - Luciano Monfardini
- Department of Gynecology and Obstetrics, University of Parma, Via Gramsci, 43125, Parma, Italy
| | - Michela Gaiano
- Department of Gynecology and Obstetrics, University of Parma, Via Gramsci, 43125, Parma, Italy
| | - Giulia Gambino
- Department of Gynecology and Obstetrics, University of Parma, Via Gramsci, 43125, Parma, Italy
| | - Giulio Sozzi
- Department of Gynecologic Oncology, University of Palermo, Piazzale N. Leotta, 90139, Palermo, Italy
| | - Carla Merisio
- Department of Gynecology and Obstetrics, University of Parma, Via Gramsci, 43125, Parma, Italy
| | - Roberto Berretta
- Department of Gynecology and Obstetrics, University of Parma, Via Gramsci, 43125, Parma, Italy
| |
Collapse
|
23
|
Sebire NJ. Recurrent GTD and GTD coexisting with normal twin pregnancy. Best Pract Res Clin Obstet Gynaecol 2020; 74:122-130. [PMID: 33451920 DOI: 10.1016/j.bpobgyn.2020.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/04/2020] [Accepted: 12/01/2020] [Indexed: 10/22/2022]
Abstract
Hydatidiform mole (HM) affects around 1/1000 pregnancies, and in such cases the recurrence risk is around 1%, being greater for those with complete HM (CHM). Whilst most cases appear sporadic with unknown mechanisms, there is a distinct subgroup of patients who suffer recurrent pregnancy loss, including multiple recurrent CHM (familial recurrent biparental HM syndrome). The majority of these cases are related to maternal genetic mutations in genes related to the control of imprinting, specifically NALP7 and KHDC3L. Oocyte donation is an effective treatment allowing these patients to have successful pregnancies. Approximately 1 in 50,000 pregnancies are complicated by twin pregnancy comprising normal foetus and HM, the majority of reported cases being CHM. Such pregnancies are at significantly increased risk of complications, including pregnancy loss, early-onset preeclampsia and severe preterm delivery, but when managed conservatively the delivery of a liveborn healthy infant occurs in around one-third of cases. Regardless of management, the risk of persistent GTD in such cases appears similar to that following singleton CHM. Rarely, other conditions mimic prenatal ultrasound appearances of twin pregnancy with HM, CHM mosaicism and placental mesenchymal dysplasia, both of which have distinctive histological and genetic features.
Collapse
Affiliation(s)
- Neil J Sebire
- Trophoblastic Disease Unit, Department of Histopathology, Charing Cross Hospital Imperial Nhs Trust, London, UK.
| |
Collapse
|
24
|
Capalbo A, Poli M, Riera-Escamilla A, Shukla V, Kudo Høffding M, Krausz C, Hoffmann ER, Simon C. Preconception genome medicine: current state and future perspectives to improve infertility diagnosis and reproductive and health outcomes based on individual genomic data. Hum Reprod Update 2020; 27:254-279. [PMID: 33197264 DOI: 10.1093/humupd/dmaa044] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/13/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Our genetic code is now readable, writable and hackable. The recent escalation of genome-wide sequencing (GS) applications in population diagnostics will not only enable the assessment of risks of transmitting well-defined monogenic disorders at preconceptional stages (i.e. carrier screening), but also facilitate identification of multifactorial genetic predispositions to sub-lethal pathologies, including those affecting reproductive fitness. Through GS, the acquisition and curation of reproductive-related findings will warrant the expansion of genetic assessment to new areas of genomic prediction of reproductive phenotypes, pharmacogenomics and molecular embryology, further boosting our knowledge and therapeutic tools for treating infertility and improving women's health. OBJECTIVE AND RATIONALE In this article, we review current knowledge and potential development of preconception genome analysis aimed at detecting reproductive and individual health risks (recessive genetic disease and medically actionable secondary findings) as well as anticipating specific reproductive outcomes, particularly in the context of IVF. The extension of reproductive genetic risk assessment to the general population and IVF couples will lead to the identification of couples who carry recessive mutations, as well as sub-lethal conditions prior to conception. This approach will provide increased reproductive autonomy to couples, particularly in those cases where preimplantation genetic testing is an available option to avoid the transmission of undesirable conditions. In addition, GS on prospective infertility patients will enable genome-wide association studies specific for infertility phenotypes such as predisposition to premature ovarian failure, increased risk of aneuploidies, complete oocyte immaturity or blastocyst development failure, thus empowering the development of true reproductive precision medicine. SEARCH METHODS Searches of the literature on PubMed Central included combinations of the following MeSH terms: human, genetics, genomics, variants, male, female, fertility, next generation sequencing, genome exome sequencing, expanded carrier screening, secondary findings, pharmacogenomics, controlled ovarian stimulation, preconception, genetics, genome-wide association studies, GWAS. OUTCOMES Through PubMed Central queries, we identified a total of 1409 articles. The full list of articles was assessed for date of publication, limiting the search to studies published within the last 15 years (2004 onwards due to escalating research output of next-generation sequencing studies from that date). The remaining articles' titles were assessed for pertinence to the topic, leaving a total of 644 articles. The use of preconception GS has the potential to identify inheritable genetic conditions concealed in the genome of around 4% of couples looking to conceive. Genomic information during reproductive age will also be useful to anticipate late-onset medically actionable conditions with strong genetic background in around 2-4% of all individuals. Genetic variants correlated with differential response to pharmaceutical treatment in IVF, and clear genotype-phenotype associations are found for aberrant sperm types, oocyte maturation, fertilization or pre- and post-implantation embryonic development. All currently known capabilities of GS at the preconception stage are reviewed along with persisting and forthcoming barriers for the implementation of precise reproductive medicine. WIDER IMPLICATIONS The expansion of sequencing analysis to additional monogenic and polygenic traits may enable the development of cost-effective preconception tests capable of identifying underlying genetic causes of infertility, which have been defined as 'unexplained' until now, thus leading to the development of a true personalized genomic medicine framework in reproductive health.
Collapse
Affiliation(s)
- Antonio Capalbo
- Igenomix Italy, Marostica, Italy.,Igenomix Foundation, INCLIVA, Valencia, Spain
| | | | - Antoni Riera-Escamilla
- Andrology Department, Fundació Puigvert, Universitat Autònoma de Barcelona, Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Vallari Shukla
- Department of Cellular and Molecular Medicine, DRNF Center for Chromosome Stability, University of Copenhagen, Copenhagen, Denmark
| | - Miya Kudo Høffding
- Department of Cellular and Molecular Medicine, DRNF Center for Chromosome Stability, University of Copenhagen, Copenhagen, Denmark
| | - Csilla Krausz
- Andrology Department, Fundació Puigvert, Universitat Autònoma de Barcelona, Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, Spain.,Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Centre of Excellence DeNothe, University of Florence, Florence, Italy
| | - Eva R Hoffmann
- Department of Cellular and Molecular Medicine, DRNF Center for Chromosome Stability, University of Copenhagen, Copenhagen, Denmark
| | - Carlos Simon
- Igenomix Foundation, INCLIVA, Valencia, Spain.,Department of Obstetrics and Gynecology, University of Valencia, Valencia, Spain.,Department of Obstetrics and Gynecology BIDMC, Harvard University, Cambridge, MA, USA
| |
Collapse
|
25
|
Amoushahi M, Sunde L, Lykke-Hartmann K. The pivotal roles of the NOD-like receptors with a PYD domain, NLRPs, in oocytes and early embryo development†. Biol Reprod 2020; 101:284-296. [PMID: 31201414 DOI: 10.1093/biolre/ioz098] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/29/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
Nucleotide-binding oligomerization domain (NOD)-like receptors with a pyrin domain (PYD), NLRPs, are pattern recognition receptors, well recognized for their important roles in innate immunity and apoptosis. However, several NLRPs have received attention for their new, specialized roles as maternally contributed genes important in reproduction and embryo development. Several NLRPs have been shown to be specifically expressed in oocytes and preimplantation embryos. Interestingly, and in line with divergent functions, NLRP genes reveal a complex evolutionary divergence. The most pronounced difference is the human-specific NLRP7 gene, not identified in rodents. However, mouse models have been extensively used to study maternally contributed NLRPs. The NLRP2 and NLRP5 proteins are components of the subcortical maternal complex (SCMC), which was recently identified as essential for mouse preimplantation development. The SCMC integrates multiple proteins, including KHDC3L, NLRP5, TLE6, OOEP, NLRP2, and PADI6. The NLRP5 (also known as MATER) has been extensively studied. In humans, inactivating variants in specific NLRP genes in the mother are associated with distinct phenotypes in the offspring, such as biparental hydatidiform moles (BiHMs) and preterm birth. Maternal-effect recessive mutations in KHDC3L and NLRP5 (and NLRP7) are associated with reduced reproductive outcomes, BiHM, and broad multilocus imprinting perturbations. The precise mechanisms of NLRPs are unknown, but research strongly indicates their pivotal roles in the establishment of genomic imprints and post-zygotic methylation maintenance, among other processes. Challenges for the future include translations of findings from the mouse model into human contexts and implementation in therapies and clinical fertility management.
Collapse
Affiliation(s)
| | - Lone Sunde
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
26
|
Fallahi J, Alashti SK, Aliabadi BE, Mohammadi S, Fardaei M. Recurrent pregnancy loss in the female with a heterozygous mutation in KHDC3L gene. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
27
|
NLRP7 plays a functional role in regulating BMP4 signaling during differentiation of patient-derived trophoblasts. Cell Death Dis 2020; 11:658. [PMID: 32814763 PMCID: PMC7438493 DOI: 10.1038/s41419-020-02884-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 01/03/2023]
Abstract
Complete hydatidiform mole (HM) is a gestational trophoblastic disease resulting in hyperproliferation of trophoblast cells and absence of embryo development. Mutations in the maternal-effect gene NLRP7 are the major cause of familial recurrent complete HM. Here, we established an in vitro model of HM using patient-specific induced pluripotent stem cells (iPSCs) derived trophoblasts harboring NLRP7 mutations. Using whole transcriptome profiling during trophoblast differentiation, we showed that impaired NLRP7 expression results in precocious downregulation of pluripotency factors, activation of trophoblast lineage markers, and promotes maturation of differentiated extraembryonic cell types such as syncytiotrophoblasts. Interestingly, we found that these phenotypes are dependent on BMP4 signaling and BMP pathway inhibition corrected the excessive trophoblast differentiation of patient-derived iPSCs. Our human iPSC model of a genetic placental disease recapitulates aspects of trophoblast biology, highlights the broad utility of iPSC-derived trophoblasts for modeling human placental diseases and identifies NLRP7 as an essential modulator of key developmental cell fate regulators.
Collapse
|
28
|
Hafezi M, Chekini Z, Zamanian M. Which One Is More Prominent in Recurrent Hydatidiform Mole, Ovum or Sperm? INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2020; 14:154-158. [PMID: 32681629 PMCID: PMC7382686 DOI: 10.22074/ijfs.2020.6017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 01/14/2020] [Indexed: 11/04/2022]
Abstract
Recurrent hydatidiform mole is defined as episodes of two molar pregnancies in a female. Often, complete moles only derive androgenic nuclear genome. We described two cases with repeated molar pregnancies attempted to prevent future episodes by performing intracytoplasmic sperm injection (ICSI) and preimplantation genetic diagnosis (PGD) to assess genetic disorders. The first patient had previously six complete molar pregnancies and advised to carry out ICSI with ovum donation to achieve a normal pregnancy. The second case had previously five molar pregnancies and no XY embryos from the ICSI/PGD process. We had to (at the insistence of the patient) transfer XX embryos in this patient which resulted in a complete hydatidiform mole (CHM). Hence, available data based on our patients and previous studies demonstrated that oocyte might play a critical role in the pathophysiology of recurrent hydatidiform mole, while it has not been often considered.
Collapse
Affiliation(s)
- Maryam Hafezi
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran. Electronic Address:
| | - Zahra Chekini
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mohammadreza Zamanian
- Department of Genetics, Reproductive Biomedicine Research Center , Royan Institute for Reproductive Biomedicine, ACECR, Tehran , Iran
| |
Collapse
|
29
|
Cinkornpumin JK, Kwon SY, Guo Y, Hossain I, Sirois J, Russett CS, Tseng HW, Okae H, Arima T, Duchaine TF, Liu W, Pastor WA. Naive Human Embryonic Stem Cells Can Give Rise to Cells with a Trophoblast-like Transcriptome and Methylome. Stem Cell Reports 2020; 15:198-213. [PMID: 32619492 PMCID: PMC7363941 DOI: 10.1016/j.stemcr.2020.06.003] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 01/01/2023] Open
Abstract
Human embryonic stem cells (hESCs) readily differentiate to somatic or germ lineages but have impaired ability to form extra-embryonic lineages such as placenta or yolk sac. Here, we demonstrate that naive hESCs can be converted into cells that exhibit the cellular and molecular phenotypes of human trophoblast stem cells (hTSCs) derived from human placenta or blastocyst. The resulting "transdifferentiated" hTSCs show reactivation of core placental genes, acquisition of a placenta-like methylome, and the ability to differentiate to extravillous trophoblasts and syncytiotrophoblasts. Modest differences are observed between transdifferentiated and placental hTSCs, most notably in the expression of certain imprinted loci. These results suggest that naive hESCs can differentiate to extra-embryonic lineage and demonstrate a new way of modeling human trophoblast specification and placental methylome establishment.
Collapse
Affiliation(s)
| | - Sin Young Kwon
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Yixin Guo
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, 718 East Haizhou Road, Haining 314400, China
| | - Ishtiaque Hossain
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Jacinthe Sirois
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Colleen S Russett
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Hsin-Wei Tseng
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Hiroaki Okae
- Department of Informative Genetics, Environment and Genome Research Centre, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Centre, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Thomas F Duchaine
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Wanlu Liu
- Department of Orthopedic of the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China; Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, 718 East Haizhou Road, Haining 314400, China
| | - William A Pastor
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada.
| |
Collapse
|
30
|
Germline NLRP7 mutations: genomic imprinting and hydatidiform mole. Virchows Arch 2020; 477:175-176. [PMID: 32577811 DOI: 10.1007/s00428-020-02802-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/05/2020] [Accepted: 03/18/2020] [Indexed: 10/24/2022]
|
31
|
Zhang W, Chen Z, Zhang D, Zhao B, Liu L, Xie Z, Yao Y, Zheng P. KHDC3L mutation causes recurrent pregnancy loss by inducing genomic instability of human early embryonic cells. PLoS Biol 2019; 17:e3000468. [PMID: 31609975 PMCID: PMC6812846 DOI: 10.1371/journal.pbio.3000468] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 10/24/2019] [Accepted: 09/26/2019] [Indexed: 12/28/2022] Open
Abstract
Recurrent pregnancy loss (RPL) is an important complication in reproductive health. About 50% of RPL cases are unexplained, and understanding the genetic basis is essential for its diagnosis and prognosis. Herein, we report causal KH domain containing 3 like (KHDC3L) mutations in RPL. KHDC3L is expressed in human epiblast cells and ensures their genome stability and viability. Mechanistically, KHDC3L binds to poly(ADP-ribose) polymerase 1 (PARP1) to stimulate its activity. In response to DNA damage, KHDC3L also localizes to DNA damage sites and facilitates homologous recombination (HR)-mediated DNA repair. KHDC3L dysfunction causes PARP1 inhibition and HR repair deficiency, which is synthetically lethal. Notably, we identified two critical residues, Thr145 and Thr156, whose phosphorylation by Ataxia-telangiectasia mutated (ATM) is essential for KHDC3L’s functions. Importantly, two deletions of KHDC3L (p.E150_V160del and p.E150_V172del) were detected in female RPL patients, both of which harbor a common loss of Thr156 and are impaired in PARP1 activation and HR repair. In summary, our study reveals both KHDC3L as a new RPL risk gene and its critical function in DNA damage repair pathways. Recurrent pregnancy loss is an important complication in reproductive health, and about 50% of cases remain unexplained. This study shows that KHDC3L safeguards the genomic stability of human early embryonic cells, and damaging mutations in its gene cause recurrent pregnancy loss in humans.
Collapse
Affiliation(s)
- Weidao Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Zhongliang Chen
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Dengfeng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Bo Zhao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Lu Liu
- Department of Obstetrics and Gynaecology, Yan An Hospital, Kunming Medical University, Kunming, China
| | - Zhengyuan Xie
- Yunnan Key Laboratory for Fertility Regulation and Birth Health of Minority Nationalities, Key Laboratory of Preconception Health in Western China, NHFPC, Population and Family Planning Institute of Yunnan Province, Kunming, China
| | - Yonggang Yao
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ping Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
- * E-mail:
| |
Collapse
|
32
|
King JR, Wilson ML, Hetey S, Kiraly P, Matsuo K, Castaneda AV, Toth E, Krenacs T, Hupuczi P, Mhawech-Fauceglia P, Balogh A, Szilagyi A, Matko J, Papp Z, Roman LD, Cortessis VK, Than NG. Dysregulation of Placental Functions and Immune Pathways in Complete Hydatidiform Moles. Int J Mol Sci 2019; 20:E4999. [PMID: 31658584 PMCID: PMC6829352 DOI: 10.3390/ijms20204999] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022] Open
Abstract
Gene expression studies of molar pregnancy have been limited to a small number of candidate loci. We analyzed high-dimensional RNA and protein data to characterize molecular features of complete hydatidiform moles (CHMs) and corresponding pathologic pathways. CHMs and first trimester placentas were collected, histopathologically examined, then flash-frozen or paraffin-embedded. Frozen CHMs and control placentas were subjected to RNA-Seq, with resulting data and published placental RNA-Seq data subjected to bioinformatics analyses. Paraffin-embedded tissues from CHMs and control placentas were used for tissue microarray (TMA) construction, immunohistochemistry, and immunoscoring for galectin-14. Of the 14,022 protein-coding genes expressed in all samples, 3,729 were differentially expressed (DE) in CHMs, of which 72% were up-regulated. DE genes were enriched in placenta-specific genes (OR = 1.88, p = 0.0001), of which 79% were down-regulated, imprinted genes (OR = 2.38, p = 1.54 × 10-6), and immune genes (OR = 1.82, p = 7.34 × 10-18), of which 73% were up-regulated. DNA methylation-related enzymes and histone demethylases were dysregulated. "Cytokine-cytokine receptor interaction" was the most impacted of 38 dysregulated pathways, among which 17 were immune-related pathways. TMA-based immunoscoring validated the lower expression of galectin-14 in CHM. In conclusion, placental functions were down-regulated, imprinted gene expression was altered, and immune pathways were activated, indicating complex dysregulation of placental developmental and immune processes in CHMs.
Collapse
Affiliation(s)
- Jennifer R King
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Melissa L Wilson
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Szabolcs Hetey
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary.
| | - Peter Kiraly
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary.
| | - Koji Matsuo
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Antonio V Castaneda
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Eszter Toth
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary.
| | - Tibor Krenacs
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary.
| | - Petronella Hupuczi
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary.
| | - Paulette Mhawech-Fauceglia
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary.
| | - Andras Szilagyi
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary.
| | - Janos Matko
- Department of Immunology, Institute of Biology, Eotvos Lorand University, H-1117 Budapest, Hungary.
| | - Zoltan Papp
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary.
- Department of Obstetrics and Gynecology, Semmelweis University, H-1088 Budapest, Hungary.
| | - Lynda D Roman
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Victoria K Cortessis
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Nandor Gabor Than
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary.
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary.
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary.
| |
Collapse
|
33
|
Matsushima N, Takatsuka S, Miyashita H, Kretsinger RH. Leucine Rich Repeat Proteins: Sequences, Mutations, Structures and Diseases. Protein Pept Lett 2019; 26:108-131. [PMID: 30526451 DOI: 10.2174/0929866526666181208170027] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 12/18/2022]
Abstract
Mutations in the genes encoding Leucine Rich Repeat (LRR) containing proteins are associated with over sixty human diseases; these include high myopia, mitochondrial encephalomyopathy, and Crohn's disease. These mutations occur frequently within the LRR domains and within the regions that shield the hydrophobic core of the LRR domain. The amino acid sequences of fifty-five LRR proteins have been published. They include Nod-Like Receptors (NLRs) such as NLRP1, NLRP3, NLRP14, and Nod-2, Small Leucine Rich Repeat Proteoglycans (SLRPs) such as keratocan, lumican, fibromodulin, PRELP, biglycan, and nyctalopin, and F-box/LRR-repeat proteins such as FBXL2, FBXL4, and FBXL12. For example, 363 missense mutations have been identified. Replacement of arginine, proline, or cysteine by another amino acid, or the reverse, is frequently observed. The diverse effects of the mutations are discussed based on the known structures of LRR proteins. These mutations influence protein folding, aggregation, oligomerization, stability, protein-ligand interactions, disulfide bond formation, and glycosylation. Most of the mutations cause loss of function and a few, gain of function.
Collapse
Affiliation(s)
- Norio Matsushima
- Center for Medical Education, Sapporo Medical University, Sapporo 060-8556, Japan.,Institute of Tandem Repeats, Noboribetsu 059-0464, Japan
| | - Shintaro Takatsuka
- Center for Medical Education, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Hiroki Miyashita
- Institute of Tandem Repeats, Noboribetsu 059-0464, Japan.,Hokubu Rinsho Co., Ltd, Sapporo 060-0061, Japan
| | - Robert H Kretsinger
- Department of Biology, University of Virginia, Charlottesville, VA 22904, United States
| |
Collapse
|
34
|
Yatsenko SA, Rajkovic A. Genetics of human female infertility†. Biol Reprod 2019; 101:549-566. [PMID: 31077289 PMCID: PMC8127036 DOI: 10.1093/biolre/ioz084] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/17/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
About 10% of women of reproductive age are unable to conceive or carry a pregnancy to term. Female factors alone account for at least 35% of all infertility cases and comprise a wide range of causes affecting ovarian development, maturation of oocytes, and fertilization competence, as well as the potential of a fertilized egg for preimplantation development, implantation, and fetal growth. Genetic abnormalities leading to infertility in females comprise large chromosome abnormalities, submicroscopic chromosome deletion and duplications, and DNA sequence variations in the genes that control numerous biological processes implicated in oogenesis, maintenance of ovarian reserve, hormonal signaling, and anatomical and functional development of female reproductive organs. Despite the great number of genes implicated in reproductive physiology by the study of animal models, only a subset of these genes is associated with human infertility. In this review, we mainly focus on genetic alterations identified in humans and summarize recent knowledge on the molecular pathways of oocyte development and maturation, the crucial role of maternal-effect factors during embryogenesis, and genetic conditions associated with ovarian dysgenesis, primary ovarian insufficiency, early embryonic lethality, and infertility.
Collapse
Affiliation(s)
- Svetlana A Yatsenko
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Magee-Womens Research Institute, Pittsburgh, PA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Aleksandar Rajkovic
- Department of Pathology, University of California San Francisco, San Francisco, CA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA
- Institute of Human Genetics, University of California San Francisco, San Francisco, CA
| |
Collapse
|
35
|
Wesseler K, Kraft F, Eggermann T. Molecular and Clinical Opposite Findings in 11p15.5 Associated Imprinting Disorders: Characterization of Basic Mechanisms to Improve Clinical Management. Int J Mol Sci 2019; 20:ijms20174219. [PMID: 31466347 PMCID: PMC6747273 DOI: 10.3390/ijms20174219] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/26/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
Silver-Russell and Beckwith-Wiedemann syndromes (SRS, BWS) are rare congenital human disorders characterized by opposite growth disturbances. With the increasing knowledge on the molecular basis of SRS and BWS, it has become obvious that the disorders mirror opposite alterations at the same genomic loci in 11p15.5. In fact, these changes directly or indirectly affect the expression of IGF2 and CDKN1C and their associated pathways, and thereby, cause growth disturbances as key features of both diseases. The increase of knowledge has become possible with the development and implementation of new and comprehensive assays. Whereas, in the beginning molecular testing was restricted to single chromosomal loci, many tests now address numerous loci in the same run, and the diagnostic implementation of (epi)genome wide assays is only a question of time. These high-throughput approaches will be complemented by the analysis of other omic datasets (e.g., transcriptome, metabolome, proteome), and it can be expected that the integration of these data will massively improve the understanding of the pathobiology of imprinting disorders and their diagnostics. Especially long-read sequencing methods, e.g., nanopore sequencing, allowing direct detection of native DNA modification, will strongly contribute to a better understanding of genomic imprinting in the near future. Thereby, new genomic loci and types of pathogenic variants will be identified, resulting in more precise discrimination into different molecular subgroups. These subgroups serve as the basis for (epi)genotype-phenotype correlations, allowing a more directed prognosis, counseling, and therapy. By deciphering the pathophysiological consequences of SRS and BWS and their molecular disturbances, future therapies will be available targeting the basic cause of the disease and respective pathomechanisms and will complement conventional therapeutic strategies.
Collapse
Affiliation(s)
- Katharina Wesseler
- Institute of Human Genetics, University Hospital, Technical University Aachen (RWTH), 52074 Aachen, Germany
| | - Florian Kraft
- Institute of Human Genetics, University Hospital, Technical University Aachen (RWTH), 52074 Aachen, Germany
| | - Thomas Eggermann
- Institute of Human Genetics, University Hospital, Technical University Aachen (RWTH), 52074 Aachen, Germany.
| |
Collapse
|
36
|
Ji M, Shi X, Xiang Y, Cui Q, Zhao J. NLRP7 and KHDC3L variants in Chinese patients with recurrent hydatidiform moles. Jpn J Clin Oncol 2019; 49:620-627. [PMID: 31220306 DOI: 10.1093/jjco/hyz036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 01/07/2019] [Accepted: 02/26/2019] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE Recurrent hydatidiform moles are reportedly biparental complete moles and related to mutated NLRP7 and KHDC3L. This study was designed to identify mutations of gene NLRP7 and KHDC3L in biparental complete moles. METHODS In this study, we have screened NLRP7 and KHDC3L mutations in five patients with recurrent moles and five with sporadic moles. Molar tissues and blood samples were collected from patients and their partners. Genotypes of the molar tissues were determined based on short tandem repeat polymorphism. The coding exons of NLRP7 and KHDC3L were sequenced. RESULTS Two patients with recurrent moles had biparental complete moles, while all other patients had androgenetic complete moles. Three non-synonymous variants in NLRP7 (c.955 G>A, c.1280 T>C and c.1441 G>A) and one in KHDC3L (c.602 C>G) were identified in patients with recurrent moles. NLRP7 c.1441 G>A and c.1280 T>C were mutations found in the Chinese population, while c.1441 G>A was only detected in patients with biparental complete moles in this study. CONCLUSIONS Genotyping can be used to differentiate biparental complete moles from androgenetic moles and to predict the risk of recurrent moles in future pregnancies. NLRP7 c.1441 G>A may associate with biparental complete moles. Biparental complete moles exhibit genetic heterogeneity.
Collapse
Affiliation(s)
- Mingliang Ji
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Xiaohua Shi
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Yang Xiang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| | - Quancai Cui
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Jun Zhao
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
37
|
Fallahi J, Razban V, Momtahan M, Akbarzadeh-Jahromi M, Namavar-Jahromi B, Anvar Z, Fardaei M. A Novel Mutation in NLRP7 Related to Recurrent Hydatidiform Mole and Reproductive Failure. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2019; 13:135-138. [PMID: 31037924 PMCID: PMC6500085 DOI: 10.22074/ijfs.2019.5657] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 11/24/2018] [Indexed: 11/04/2022]
Abstract
Background Hydatidiform mole (HM) is an abnormal human pregnancy with excessive trophoblastic proliferation and abnormal embryonic development, dividing into two complete HM (CHM) and partial HM (PHM) groups. One subcategory of the CHMs is recurrent and familial, which is known as biparental HM (BiHMs) or recurrent HM (RHM). NLRP7, KHDC3L and PADI6 are maternal-effect genes involved in RHMs. NLRP7 is a major gene responsible for RHMs. This study was performed on patients with molar pregnancies and miscarriage. The aim of this study was to genetic screen for mutations in NLRP7 and KHDC3L genes in an affected woman with previous history of 5RHM and the sibling with history of miscarriage. Materials and Methods In this experimental study, DNA was extracted from blood samples. KHDC3L and NLRP7 were polymerase chain reaction (PCR) amplified. The PCR products were purified and Sanger sequenced. Results In this study, there is no mutation in KHDC3L gene but a novel mutation was identified in the NACHT domain of NLRP7 gene. Patient with five recurrent moles had this mutation in the homozygous state while her sister with one miscarriage and one normal child showed this mutation in the heterozygous state. Conclusion In this study, we identified a new mutation in NLRP7 gene of a patient with recurrent HM. Following egg donation, this patient has a normal boy. The sister of this patient with heterozygous mutation has a spontaneous abortion and one normal child that confirm the impact of a defective allele of NLRP7 on reproductive wastage in a recent finding.
Collapse
Affiliation(s)
- Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Momtahan
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojgan Akbarzadeh-Jahromi
- Fetal and Maternal Research Center, Pathology Department, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Bahia Namavar-Jahromi
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Infertility Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Anvar
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Infertility Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran.Electronic Address:
| | - Majid Fardaei
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.Electronic Address:
| |
Collapse
|
38
|
Pinar MH, Gibbins K, He M, Kostadinov S, Silver R. Early Pregnancy Losses: Review of Nomenclature, Histopathology, and Possible Etiologies. Fetal Pediatr Pathol 2018; 37:191-209. [PMID: 29737906 DOI: 10.1080/15513815.2018.1455775] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Miscarriage is a frequent complication of human pregnancy: ∼50% to 70% of spontaneous conceptions are lost prior to the second trimester. Etiology of miscarriage includes genetic abnormalities, infections, immunological and implantation disorders, uterine and endocrine abnormalities, and lifestyle factors. Given such variability, knowledge regarding causes, pathophysiological mechanisms, and morphologies of primary early pregnancy loss has significant gaps; often, pregnancy losses remain unexplained. Pathologic evaluation of miscarriage tissue is an untapped source of knowledge. Although miscarriage specimens comprise a significant part of pathologists' workload, information reported from these specimens is typically of minimal clinical utility for delineating etiology or predicting recurrence risk. Standardized terminology is available, though not universally used. We reintroduce the terminology and review new information about early pregnancy losses and their morphologies. Current clinical terminology is inconsistent, hampering research progress. This review is a resource for diagnostic pathologists studying this complex problem.
Collapse
Affiliation(s)
- M Halit Pinar
- a Perinatal and Pediatric Pathology , Warren Alpert Medical School, Brown University , Providence , Rhode Island , USA
| | - Karen Gibbins
- b Division of Maternal and Fetal Medicine , University of Utah Hospital, Obstetrics and Gynecology , Salt Lake City , Utah , USA
| | - Mai He
- c Pathology and Laboratory Medicine, Division of Pediatric Pathology , Washington University in Saint Louis School of Medicine , Saint Louis , Missouri , USA
| | - Stefan Kostadinov
- d Pathology and Laboratory Medicine, Division of Perinatal and Pediatric Pathology, Women and Infants Hospital , Brown University Warren Alpert Medical School , Providence , Rhode Island , USA
| | - Robert Silver
- e Division of Maternal Fetal Medicine , University of Utah Hospital, Obstetrics and Gynecology , Salt Lake City , Utah , USA
| |
Collapse
|
39
|
Kalogiannidis I, Kalinderi K, Kalinderis M, Miliaras D, Tarlatzis B, Athanasiadis A. Recurrent complete hydatidiform mole: where we are, is there a safe gestational horizon? Opinion and mini-review. J Assist Reprod Genet 2018; 35:967-973. [PMID: 29737470 DOI: 10.1007/s10815-018-1202-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/25/2018] [Indexed: 02/07/2023] Open
Abstract
Benign hydatidiform mole, complete or partial, is the most common type of gestational trophoblastic disease (GTD) characterised by excessive trophoblastic proliferation and abnormal embryonic development. Although most complete hydatidiform moles (CHMs) are diploid androgenetic, a few cases of CHMs are biparental, characterised by recurrence and familial clustering. In these rare cases, mutations in NLRP7 or KHDC3L genes, associated with maternal imprinting defects, have been implicated. Current data regarding future pregnancy options in hydatidiform moles are discussed and our opinion is presented based on an incidence that took place in our hospital with a woman with consecutive molar pregnancies. In recurrent hydatidiform moles, DNA testing should be performed and when NLRP7 or KHDC3L mutation are detected, oocyte donation should be proposed as an option to maximise woman's chances of having a normal pregnancy.
Collapse
Affiliation(s)
- Ioannis Kalogiannidis
- 3rd Department of Obstetrics and Gynaecology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kallirhoe Kalinderi
- 3rd Department of Obstetrics and Gynaecology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece. .,Department of General Biology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Michail Kalinderis
- Department of Obstetrics and Gynaecology, King's College Hospital NHS Foundation Trust, Princess Royal University Hospital, Farnborough Common, BR6 8ND, Orpington, UK
| | - Dimosthenis Miliaras
- Laboratory of Histology & Embryology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Basil Tarlatzis
- 1st Department of Obstetrics & Gynaecology, Papageorgiou General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Apostolos Athanasiadis
- 3rd Department of Obstetrics and Gynaecology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
40
|
Clinical and genetic-epigenetic aspects of recurrent hydatidiform mole: A review of literature. Taiwan J Obstet Gynecol 2018; 57:1-6. [DOI: 10.1016/j.tjog.2017.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2017] [Indexed: 11/19/2022] Open
|
41
|
Monk D, Sanchez-Delgado M, Fisher R. NLRPs, the subcortical maternal complex and genomic imprinting. Reproduction 2017; 154:R161-R170. [PMID: 28916717 DOI: 10.1530/rep-17-0465] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/06/2017] [Accepted: 09/15/2017] [Indexed: 01/01/2023]
Abstract
Before activation of the embryonic genome, the oocyte provides many of the RNAs and proteins required for the epigenetic reprogramming and the transition to a totipotent state. Targeted disruption of a subset of oocyte-derived transcripts in mice results in early embryonic lethality and cleavage-stage embryonic arrest as highlighted by the members of the subcortical maternal complex (SCMC). Maternal-effect recessive mutations of NLRP7, KHDC3L and NLRP5 in humans are associated with variable reproductive outcomes, biparental hydatidiform moles (BiHM) and widespread multi-locus imprinting disturbances. The precise mechanism of action of these genes is unknown, but the maternal-effect phenomenon suggests a function during early pre-implantation development, while biochemical and genetic studies implement them as SCMC members or interacting partners. In this review article, we discuss the role of the NLRP family members and the SCMC proteins in the establishment of genomic imprints and post-zygotic methylation maintenance, the recent advances made in the understanding of the biology involved in BiHM formation and the wider roles of the SCMC in mammalian reproduction.
Collapse
Affiliation(s)
- David Monk
- Imprinting and Cancer GroupCancer Epigenetic and Biology Program, Institut d'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona, Spain
| | - Marta Sanchez-Delgado
- Imprinting and Cancer GroupCancer Epigenetic and Biology Program, Institut d'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona, Spain
| | - Rosemary Fisher
- Imperial Centre for Translational and Experimental MedicineImperial College London, London, UK.,Trophoblastic Tumour Screening and Treatment CentreDepartment of Oncology, Imperial College London, London, UK
| |
Collapse
|
42
|
Huang JY, Yu PH, Li YC, Kuo PL. NLRP7 contributes to in vitro decidualization of endometrial stromal cells. Reprod Biol Endocrinol 2017; 15:66. [PMID: 28810880 PMCID: PMC5558772 DOI: 10.1186/s12958-017-0286-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/09/2017] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Nucleotide-binding oligomerization domain (NACHT), leucine rich repeat (LRR) and pyrin domain (PYD) 7 containing protein, NLRP7, is a member of the NLR family which serves as innate immune sensors. Mutations and genetic variants of NLRP7 have been found in women with infertility associated conditions, such as recurrent hydatidiform mole, recurrent miscarriage, and preeclampsia. Decidualization of endometrial stromal cells is a hallmark of tissue remodeling to support embryo implantation and proper placental development. Given defective decidualization has been implicated in miscarriage as well as preeclampsia, we aimed to explore the link between the NLRP7 gene and decidualization. METHODS Endometrial samples obtained from pregnant women in the first trimester and non-pregnant women were used to study NLRP7 expression pattern. The human telomerase reverse transcriptase (hTERT)-immortalized human endometrial stromal cells (T-HESCs) were used to study the effect of NLRP7 on decidualization. Decidualization of T-HESCs was induced with 1 μM medroxyprogesterone acetate (MPA) and 0.5 mM 8-bromoadenosine 3':5'-cyclic monophosphate (8-Br-cAMP). siRNA was used to knock down NLRP7 while lentiviral vectors were used to overexpress NLRP7 in cells. NLRP7 expression was detected by immunofluorescence, qRT-PCR, and Western blotting. Decidualization markers, Insulin-like growth factor-binding protein 1 (IGFBP-1) and prolactin (PRL), were detected by qRT-PCR and ELISA. Nuclear translocation of NLRP7 was detected by the subcellular fractionation and confocal microscopy. The effect of NLRP7 on progesterone receptor (PR) activity was evaluated by a reporter system. RESULTS NLRP7 was up-regulated in the decidual stromal cells of human first-trimester endometrium. After in vitro decidualization, T-HESCs presented with the swollen phenotype and increased expressions of IGFBP-1 and PRL. Knockdown or over-expression of NLRP7 reduced or enhanced the decidualization, respectively, according to the expression level of IGFBP-1. NLRP7 was found to translocate in the nucleus of decidualized T-HESCs and able to promote PR activity. CONCLUSIONS NLRP7 was upregulated and translocated to the nucleus of the endometrial stromal cells in an in vitro decidualization model. Overexpressed NLRP7 promoted the IGFBP-1 expression and PR reporter activation. IGFBP-1 expression decreased with the knockdown of NLRP7. Therefore, we suggest that NLRP7 contributes to in vitro decidualization of endometrial stromal cells.
Collapse
Affiliation(s)
- Jyun-Yuan Huang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, 138 Sheng-Li Road, Tainan, 704, Taiwan
| | - Pei-Hsiu Yu
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, 138 Sheng-Li Road, Tainan, 704, Taiwan
| | - Yueh-Chun Li
- Department of Biomedical Sciences, Chung Shan Medical University, No.110, Sec. 1, Jianguo N. Rd., South Dist, Taichung City, 402, Taiwan.
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, 138 Sheng-Li Road, Tainan, 704, Taiwan.
| |
Collapse
|
43
|
Rigante D. A systematic approach to autoinflammatory syndromes: a spelling booklet for the beginner. Expert Rev Clin Immunol 2017; 13:571-597. [PMID: 28064547 DOI: 10.1080/1744666x.2017.1280396] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hallmark of autoinflammatory syndromes (AIS) is the periodic recurrence of 'sterile' inflammatory attacks characterized by fever and organ- or tissue-specific inflammation. Basic research projects over the last two decades have boosted our understanding of pathological pathways, mainly involving interleukin (IL)-1 biosynthesis, and also revealed that their dysregulation results from genetically-heterogeneous inborn errors of innate immunity and leads to multiple inflammatory phenotypes. Starting from the evidence of poor response to IL-1 inhibitors of some patients with multi-organ inflammation, further research studies have disclosed a crucial role for nuclear factor (NF)-κB and type I interferon (IFN) in specific AIS. Presently, new genetically-defined AIS have been identified, following the in-depth analysis of molecular pathways which involve either constitutive NF-κB activation or IFN signaling. Areas covered: This review is intended as a spelling booklet to help clinicians approaching patients with AIS in a simple way, using the component of the innate immunity they mainly affect. AIS have been split into 4 groups: IL-1-mediated disorders, NF-κB-mediated disorders, IFN-mediated disorders, and syndromes with still unraveled pathogenetic mechanisms or without any dominating cytokine involved. This classification has mere scholastic purposes and does not reflect the intimate complexity of each disorder discussed herein. Expert commentary: The understanding of dysregulated molecular pathways driving specific phenotypes in most AIS has prompted numerous projects to discover therapies targeting directly cytokine-mediated manifestations in such problematic patients, hopefully aimed to decrease or cancel inflammation and lead to a drastic change in patients' lives. The future has only begun.
Collapse
Affiliation(s)
- Donato Rigante
- a Institute of Pediatrics, Periodic Fever Research Center , Fondazione Policlinico Universitario A. Gemelli, Università Cattolica Sacro Cuore , Rome , Italy
| |
Collapse
|
44
|
Eysbouts Y, Brouwer R, Ottevanger P, Massuger L, Sweep F, Thomas C, van Herwaarden A. Serum Human Chorionic Gonadotropin Normogram for the Detection of Gestational Trophoblastic Neoplasia. Int J Gynecol Cancer 2017; 27:1035-1041. [PMID: 28498241 DOI: 10.1097/igc.0000000000000966] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE The aim of this study was to develop a serum human chorionic gonadotropin (hCG) normogram for both uneventful complete and partial hydatidiform moles in the first-trimester ultrasound era. METHODS An hCG normogram for both complete and partial hydatidiform moles was constructed, based on 639 patients with uneventful serum hCG regression after evacuation between 1990 and 2014. Serum hCG was measured by an in-house-developed radioimmunoassay, detecting both intact hCG and free β-subunit. It has been in use for all serum measurements sent to the Dutch Central Registry for Hydatidiform Moles since 1977. RESULTS Since introduction of routine first-trimester ultrasonography, lower pre-evacuation and follow-up serum hCG concentrations were observed. When compared with complete hydatidiform moles, patients with a partial hydatidiform mole had significantly lower pre-evacuation serum hCG concentration (median, 4400 and 875 ng/mL, respectively; P < 0.001) and earlier hCG normalization (median, 7 and 6 weeks, respectively; P < 0.001) but higher gestational age (mean, 11.5 and 13.0 weeks, respectively; P < 0.001). For both complete and partial hydatidiform moles, 95% of patients reached normal serum hCG concentrations within 14 weeks after evacuation. CONCLUSIONS A normogram for the detection of gestational trophoblastic neoplasia was developed for complete and partial hydatidiform moles. Although interesting from a scientific perspective, the small divergence in hCG regression between complete and partial hydatidiform moles will be of little importance in clinical practice, as actual differences in regression will encompass only days. To promote clarity and unity in daily practice, we therefore propose a combined normogram to be used as a reference guideline for follow-up after evacuation of a hydatidiform mole. This normogram will be compliant with patients in today's clinical practice.
Collapse
Affiliation(s)
- Yalcke Eysbouts
- Departments of *Obstetrics and Gynecology, †Laboratory Medicine, and ‡Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Hui P, Buza N, Murphy KM, Ronnett BM. Hydatidiform Moles: Genetic Basis and Precision Diagnosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 12:449-485. [DOI: 10.1146/annurev-pathol-052016-100237] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Pei Hui
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06510;
| | - Natalia Buza
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06510;
| | | | - Brigitte M. Ronnett
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21231
| |
Collapse
|
47
|
Kuchmiy AA, D'Hont J, Hochepied T, Lamkanfi M. NLRP2 controls age-associated maternal fertility. J Exp Med 2016; 213:2851-2860. [PMID: 27881734 PMCID: PMC5154945 DOI: 10.1084/jem.20160900] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/27/2016] [Accepted: 10/24/2016] [Indexed: 11/24/2022] Open
Abstract
Kuchmiy et al. show that Nlrp2, while dispensable for regulation of inflammasome activation, controls maternal fertility with progressing age, playing an unexpected and critical role in maintaining oocyte quality later in life. Nucleotide-binding domain and leucine-rich repeat (NLR) proteins are well-known for their key roles in the immune system. Ectopically expressed NLRP2 in immortalized cell lines assembles an inflammasome and inhibits activation of the proinflammatory transcription factor NF-κB, but the physiological roles of NLRP2 are unknown. Here, we show that Nlrp2-deficient mice were born with expected Mendelian ratios and that Nlrp2 was dispensable for innate and adaptive immunity. The observation that Nlrp2 was exclusively expressed in oocytes led us to explore the role of Nlrp2 in parthenogenetic activation of oocytes. Remarkably, unlike oocytes of young adult Nlrp2-deficient mice, activated oocytes of mature adult mice developed slower and largely failed to reach the blastocyst stage. In agreement, we noted strikingly declining reproductive rates in vivo with progressing age of female Nlrp2-deficient mice. This work identifies Nlrp2 as a critical regulator of oocyte quality and suggests that NLRP2 variants with reduced activity may contribute to maternal age-associated fertility loss in humans.
Collapse
Affiliation(s)
- Anna A Kuchmiy
- Inflammation Research Center, VIB, B-9052 Zwijnaarde, Belgium.,Department of Internal Medicine, Ghent University, B-9000 Ghent, Belgium
| | - Jinke D'Hont
- Inflammation Research Center, VIB, B-9052 Zwijnaarde, Belgium.,Department of Biomedical Molecular Biology, Ghent University, B-9000 Ghent, Belgium
| | - Tino Hochepied
- Inflammation Research Center, VIB, B-9052 Zwijnaarde, Belgium.,Department of Biomedical Molecular Biology, Ghent University, B-9000 Ghent, Belgium
| | - Mohamed Lamkanfi
- Inflammation Research Center, VIB, B-9052 Zwijnaarde, Belgium .,Department of Internal Medicine, Ghent University, B-9000 Ghent, Belgium
| |
Collapse
|
48
|
Colgan TJ, Chang MC, Nanji S, Kolomietz E. A Reappraisal of the Incidence of Placental Hydatidiform Mole Using Selective Molecular Genotyping. Int J Gynecol Cancer 2016; 26:1345-50. [PMID: 27258730 DOI: 10.1097/igc.0000000000000754] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Reports on the incidence of hydatidiform mole (HM) have varied depending on study population and methodology. This institutional-based study was undertaken to identify the incidence of HM in a modern obstetric practice using advanced laboratory diagnostic techniques. METHODS A retrospective review of consecutive hospital cases of HM was conducted for a 27-month period. Pathologic diagnoses of partial mole (PM) and complete mole (CM) were based on histopathologic assessment and selective use of p57 immunohistochemistry and molecular genotyping (MG) using formalin-fixed paraffin-embedded tissues. RESULTS During the study period, 14,944 obstetric deliveries took place at our institution. Forty-nine cases of HM (18 CMs, 31 PMs) were identified. Histopathology with the selective use of p57 immunohistochemistry was used in 25 of 49 HMs (18 CMs, 7 PMs). Histopathologic features were equivocal in the remaining cases (24/49 cases), and adjunctive MG was performed; all were PMs. The incidence of HM was 3.3/1000 deliveries. Partial mole was more prevalent with a CM (PM ratio, 1:1.72). CONCLUSIONS Our observed incidence of HM is greater than previous studies and is attributable to improved detection of PM cases. Molecular genotyping and cytogenetic evidence indicate that CM is almost half as common as PM. This ratio may be useful in benchmarking laboratory diagnosis and HM registries.
Collapse
Affiliation(s)
- Terence J Colgan
- *Section of Gynecological Pathology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, †Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, and ‡Division of Diagnostic Medical Genetics, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
49
|
A Novel Genetic Mutation in a Patient With Recurrent Biparental Complete Hydatidiform Mole: A Brief Report. Int J Gynecol Cancer 2016; 26:1351-3. [DOI: 10.1097/igc.0000000000000755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
AbstractRecurrent hydatidiform moles are defined by the occurrence of two or more molar pregnancies in the same patient. Familial recurrent hydatidiform moles (FRHM) is a rare autosomal recessive condition where women have an inherited predisposition to have molar pregnancies. Genotyping demonstrated that they are diploid and biparental. We report a case of FRHM from Egypt with a history of 6 recurrent complete moles. Sequencing of the NLPR7 gene revealed a deleterious homozygous base change in exon 2, c.197G>A, which would result in a truncated protein p.W66*. To the best of our knowledge, this mutation has not been described before.
Collapse
|
50
|
Zhao S, Sebire NJ, Kaur B, Seckl MJ, Fisher RA. Molecular genotyping of placental site and epithelioid trophoblastic tumours; female predominance. Gynecol Oncol 2016; 142:501-7. [DOI: 10.1016/j.ygyno.2016.05.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 01/25/2023]
|