1
|
Gu R, Wu T, Fu J, Sun YJ, Sun XX. Advances in the genetic etiology of female infertility. J Assist Reprod Genet 2024; 41:3261-3286. [PMID: 39320554 PMCID: PMC11707141 DOI: 10.1007/s10815-024-03248-w] [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: 04/19/2024] [Accepted: 08/07/2024] [Indexed: 09/26/2024] Open
Abstract
Human reproduction is a complex process involving gamete maturation, fertilization, embryo cleavage and development, blastocyst formation, implantation, and live birth. If any of these processes are abnormal or arrest, reproductive failure will occur. Infertility is a state of reproductive dysfunction caused by various factors. Advances in molecular genetics, including cell and molecular genetics, and high-throughput sequencing technologies, have found that genetic factors are important causes of infertility. Genetic variants have been identified in infertile women or men and can cause gamete maturation arrest, poor quality gametes, fertilization failure, and embryonic developmental arrest during assisted reproduction technology (ART), and thus reduce the clinical success rates of ART. This article reviews clinical studies on repeated in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) failures caused by ovarian dysfunction, oocyte maturation defects, oocyte abnormalities, fertilization disorders, and preimplantation embryonic development arrest due to female genetic etiology, the accumulation of pathogenic genes and gene pathogenic loci, and the functional mechanism and clinical significance of pathogenic genes in gametogenesis and early embryonic development.
Collapse
Affiliation(s)
- Ruihuan Gu
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China
| | - Tianyu Wu
- Institute of Pediatrics, State Key Laboratory of Genetic Engineering, Institutes of BiomedicalSciences, Shanghai Key Laboratory of Medical Epigenetics, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Jing Fu
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China
| | - Yi-Juan Sun
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China.
| | - Xiao-Xi Sun
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China.
| |
Collapse
|
2
|
Gu B, Le GH, Herrera S, Blair SJ, Meissner TB, Strominger JL. HLA-C expression in extravillous trophoblasts is determined by an ELF3-NLRP2/NLRP7 regulatory axis. Proc Natl Acad Sci U S A 2024; 121:e2404229121. [PMID: 39052836 PMCID: PMC11295039 DOI: 10.1073/pnas.2404229121] [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: 03/01/2024] [Accepted: 06/03/2024] [Indexed: 07/27/2024] Open
Abstract
The distinct human leukocyte antigen (HLA) class I expression pattern of human extravillous trophoblasts (EVT) endows them with unique tolerogenic properties that enable successful pregnancy. Nevertheless, how this process is elaborately regulated remains elusive. Previously, E74 like ETS transcription factor 3 (ELF3) was identified to govern high-level HLA-C expression in EVT. In the present study, ELF3 is found to bind to the enhancer region of two adjacent NOD-like receptor (NLR) genes, NLR family pyrin domain-containing 2 and 7 (NLRP2, NLRP7). Notably, our analysis of ELF3-deficient JEG-3 cells, a human choriocarcinoma cell line widely used to study EVT biology, suggests that ELF3 transactivates NLRP7 while suppressing the expression of NLRP2. Moreover, we find that NLRP2 and NLRP7 have opposing effects on HLA-C expression, thus implicating them in immune evasion at the maternal-fetal interface. We confirmed that NLRP2 suppresses HLA-C levels and described a unique role for NLRP7 in promoting HLA-C expression in JEG-3. These results suggest that these two NLR genes, which arose via gene duplication in primates, are fine-tuned by ELF3 yet have acquired divergent functions to enable proper expression levels of HLA-C in EVT, presumably through modulating the degradation kinetics of IkBα. Targeting the ELF3-NLRP2/NLRP7-HLA-C axis may hold therapeutic potential for managing pregnancy-related disorders, such as recurrent hydatidiform moles and fetal growth restriction, and thus improve placental development and pregnancy outcomes.
Collapse
Affiliation(s)
- Bowen Gu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Gia-Han Le
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Sebastian Herrera
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Steven J. Blair
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Torsten B. Meissner
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA02115
- Department of Surgery, Harvard Medical School, Boston, MA02115
| | - Jack L. Strominger
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| |
Collapse
|
3
|
Zhang T, Xing F, Qu M, Yang Z, Liu Y, Yao Y, Xing N. NLRP2 in health and disease. Immunology 2024; 171:170-180. [PMID: 37735978 DOI: 10.1111/imm.13699] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023] Open
Abstract
NLR family pyrin domain containing 2 (NLRP2) is a novel member of the Nod-like receptor (NLR) family. However, our understanding of NLRP2 has long been ambiguous. NLRP2 may have a role in the innate immune response, but its 'specific' functions remain controversial. Although NLRP2 can initiate inflammasome and promote inflammation, it can also downregulate inflammatory signals. Additionally, NLRP2 has been reported to function in the reproductive system and shows high expression in the placenta. However, the exact role of NLRP2 in the reproductive system is unclear. Here, we highlight the most current progress on NLRP2 in inflammasome activation, effector function and regulation of nuclear factor-κB. And we discuss functions of NLRP2 in inflammatory diseases, reproductive disorders and the potential implication of NLRP2 in human diseases.
Collapse
Affiliation(s)
- Tongtong Zhang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fei Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan, China
| | - Mingcui Qu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhihu Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yafei Liu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yongchao Yao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Coombs JR, Zamoshnikova A, Holley CL, Maddugoda MP, Teo DET, Chauvin C, Poulin LF, Vitak N, Ross CM, Mellacheruvu M, Coll RC, Heinz LX, Burgener SS, Emming S, Chamaillard M, Boucher D, Schroder K. NLRP12 interacts with NLRP3 to block the activation of the human NLRP3 inflammasome. Sci Signal 2024; 17:eabg8145. [PMID: 38261657 DOI: 10.1126/scisignal.abg8145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 01/04/2024] [Indexed: 01/25/2024]
Abstract
Inflammasomes are multiprotein complexes that drive inflammation and contribute to protective immunity against pathogens and immune pathology in autoinflammatory diseases. Inflammasomes assemble when an inflammasome scaffold protein senses an activating signal and forms a signaling platform with the inflammasome adaptor protein ASC. The NLRP subfamily of NOD-like receptors (NLRs) includes inflammasome nucleators (such as NLRP3) and also NLRP12, which is genetically linked to familial autoinflammatory disorders that resemble diseases caused by gain-of-function NLRP3 mutants that generate a hyperactive NLRP3 inflammasome. We performed a screen to identify ASC inflammasome-nucleating proteins among NLRs that have the canonical pyrin-NACHT-LRR domain structure. Only NLRP3 and NLRP6 could initiate ASC polymerization to form "specks," and NLRP12 failed to nucleate ASC polymerization. However, wild-type NLRP12 inhibited ASC inflammasome assembly induced by wild-type and gain-of-function mutant NLRP3, an effect not seen with disease-associated NLRP12 mutants. The capacity of NLRP12 to suppress NLRP3 inflammasome assembly was limited to human NLRP3 and was not observed for wild-type murine NLRP3. Furthermore, peripheral blood mononuclear cells from patients with an NLRP12 mutant-associated inflammatory disorder produced increased amounts of the inflammatory cytokine IL-1β in response to NLRP3 stimulation. Thus, our findings provide insights into NLRP12 biology and suggest that NLRP3 inhibitors in clinical trials for NLRP3-driven diseases may also be effective in treating NLRP12-associated autoinflammatory diseases.
Collapse
Affiliation(s)
- Jared R Coombs
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Alina Zamoshnikova
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Caroline L Holley
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Madhavi P Maddugoda
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Daniel Eng Thiam Teo
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Camille Chauvin
- U1019, Institut Pasteur de Lille, University of Lille, Centre National de la Recherche Scientifique, INSERM, Centre Hospitalo-Universitaire Lille, Lille 59019, France
| | - Lionel F Poulin
- Laboratory of Cell Physiology, INSERM U1003, University of Lille, Lille 59000, France
| | - Nazarii Vitak
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia 4072, Australia
| | - Connie M Ross
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia 4072, Australia
| | - Manasa Mellacheruvu
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Rebecca C Coll
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Leonhard X Heinz
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Sabrina S Burgener
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Stefan Emming
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Mathias Chamaillard
- U1019, Institut Pasteur de Lille, University of Lille, Centre National de la Recherche Scientifique, INSERM, Centre Hospitalo-Universitaire Lille, Lille 59019, France
| | - Dave Boucher
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| |
Collapse
|
5
|
Ozturk S. Genetic variants underlying developmental arrests in human preimplantation embryos. Mol Hum Reprod 2023; 29:gaad024. [PMID: 37335858 DOI: 10.1093/molehr/gaad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 06/03/2023] [Indexed: 06/21/2023] Open
Abstract
Developmental arrest in preimplantation embryos is one of the major causes of assisted reproduction failure. It is briefly defined as a delay or a failure of embryonic development in producing viable embryos during ART cycles. Permanent or partial developmental arrest can be observed in the human embryos from one-cell to blastocyst stages. These arrests mainly arise from different molecular biological defects, including epigenetic disturbances, ART processes, and genetic variants. Embryonic arrests were found to be associated with a number of variants in the genes playing key roles in embryonic genome activation, mitotic divisions, subcortical maternal complex formation, maternal mRNA clearance, repairing DNA damage, transcriptional, and translational controls. In this review, the biological impacts of these variants are comprehensively evaluated in the light of existing studies. The creation of diagnostic gene panels and potential ways of preventing developmental arrests to obtain competent embryos are also discussed.
Collapse
Affiliation(s)
- Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
6
|
Han J, Zhang N, Cao Q, Shi X, Wang C, Rui X, Ding J, Zhao C, Zhang J, Ling X, Li H, Guan Y, Meng Q, Huo R. NLRP7 participates in the human subcortical maternal complex and its variants cause female infertility characterized by early embryo arrest. J Mol Med (Berl) 2023:10.1007/s00109-023-02322-7. [PMID: 37148315 DOI: 10.1007/s00109-023-02322-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
Successful human reproduction requires normal oocyte maturation, fertilization, and early embryo development. Early embryo arrest is a common phenomenon leading to female infertility, but the genetic basis is largely unknown. NLR family pyrin domain-containing 7 (NLRP7) is a member of the NLRP subfamily. Previous studies have shown that variants of NLRP7 are one of the crucial causes of female recurrent hydatidiform mole, but whether NLRP7 variants can directly affect early embryo development is unclear. We performed whole-exome sequencing in patients who experienced early embryo arrest, and five heterozygous variants (c.251G > A, c.1258G > A, c.1441G > A, c. 2227G > A, c.2323C > T) of NLRP7 were identified in affected individuals. Plasmids of NLRP7 and subcortical maternal complex components were overexpressed in 293 T cells, and Co-IP experiments showed that NLRP7 interacted with NLRP5, TLE6, PADI6, NLRP2, KHDC3L, OOEP, and ZBED3. Injecting complementary RNAs in mouse oocytes and early embryos showed that NLRP7 variants influenced the oocyte quality and some of the variants significantly affected early embryo development. These findings contribute to our understanding of the role of NLRP7 in human early embryo development and provide a new genetic marker for clinical early embryo arrest patients. KEY MESSAGES: Five heterozygous variants of NLRP7 (c.1441G > A; 2227G > A; c.251G > A; c.1258G > A; c.2323C > T) were identified in five infertile patients who experienced early embryo arrest. NLRP7 is a component of human subcortical maternal complex. NLRP7 variants lead to poor quality of oocytes and early embryo development arrest. This study provides a new genetic marker for clinical early embryo arrest patients.
Collapse
Affiliation(s)
- Jian Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Nana Zhang
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiqi Cao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xiaodan Shi
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Congjing Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Ximan Rui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jie Ding
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- Reproductive Genetic Center, Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Chun Zhao
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Junqiang Zhang
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xiufeng Ling
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Hong Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- Reproductive Genetic Center, Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Yichun Guan
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Qingxia Meng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China.
- Reproductive Genetic Center, Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China.
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China.
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
7
|
Reynaud D, Alfaidy N, Collet C, Lemaitre N, Sergent F, Miege C, Soleilhac E, Assi AA, Murthi P, Courtois G, Fauvarque MO, Slim R, Benharouga M, Abi Nahed R. NLRP7 Enhances Choriocarcinoma Cell Survival and Camouflage in an Inflammasome Independent Pathway. Cells 2023; 12:857. [PMID: 36980199 PMCID: PMC10099745 DOI: 10.3390/cells12060857] [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: 01/17/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Gestational choriocarcinoma (GC) is a highly malignant trophoblastic tumor that often develops from a complete hydatidiform mole (HM). NLRP7 is the major gene responsible for recurrent HM and is involved in the innate immune response, inflammation and apoptosis. NLRP7 can function in an inflammasome-dependent or -independent pathway. Recently, we have demonstrated that NLRP7 is highly expressed in GC tumor cells and contributes to their tumorigenesis. However, the underlying mechanisms are still unknown. Here, we investigated the mechanism by which NLRP7 controls these processes in malignant (JEG-3) and non-tumor (HTR8/SVneo) trophoblastic cells. Cell survival, dedifferentiation, camouflage, and aggressiveness were compared between normal JEG-3 cells or knockdown for NLRP7, JEG-3 Sh NLRP7. In addition, HTR8/SVneo cells overexpressing NLRP7 were used to determine the impact of NLRP7 overexpression on non-tumor cells. NLRP7 involvement in tumor cell growth and tolerance was further characterized in vivo using the metastatic mouse model of GC. RESULTS We demonstrate that NLRP7 (i) functions in an inflammasome-dependent and -independent manners in HTR8/SVneo and JEG-3 cells, respectively; (ii) differentially regulates the activity of NF-κB in tumor and non-tumor cells; (iii) increases malignant cell survival, dedifferentiation, and camouflage; and (iv) facilitates tumor cells colonization of the lungs in the preclinical model of GC. CONCLUSIONS This study demonstrates for the first time the mechanism by which NLRP7, independently of its inflammasome machinery, contributes to GC growth and tumorigenesis. The clinical relevance of NLRP7 in this rare cancer highlights its potential therapeutic promise as a molecular target to treat resistant GC patients.
Collapse
Affiliation(s)
- Déborah Reynaud
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38043 Grenoble, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique, University Grenoble Alpes and Centre Hospitalo-Universitaire Grenoble Alpes, CS 10217, CEDEX 9, 38043 Grenoble, France
| | - Nadia Alfaidy
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38043 Grenoble, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique, University Grenoble Alpes and Centre Hospitalo-Universitaire Grenoble Alpes, CS 10217, CEDEX 9, 38043 Grenoble, France
| | - Constance Collet
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38043 Grenoble, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique, University Grenoble Alpes and Centre Hospitalo-Universitaire Grenoble Alpes, CS 10217, CEDEX 9, 38043 Grenoble, France
| | - Nicolas Lemaitre
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38043 Grenoble, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique, University Grenoble Alpes and Centre Hospitalo-Universitaire Grenoble Alpes, CS 10217, CEDEX 9, 38043 Grenoble, France
| | - Frederic Sergent
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38043 Grenoble, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique, University Grenoble Alpes and Centre Hospitalo-Universitaire Grenoble Alpes, CS 10217, CEDEX 9, 38043 Grenoble, France
| | - Céline Miege
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38043 Grenoble, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique, University Grenoble Alpes and Centre Hospitalo-Universitaire Grenoble Alpes, CS 10217, CEDEX 9, 38043 Grenoble, France
| | | | - Alaa Al Assi
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), Univeristy Grenoble Alpes, Inserm, 38000 Grenoble, France
| | - Padma Murthi
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne VIC 3800, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women’s Hospital, Parkville, VIC 3502, Australia
| | - Gilles Courtois
- University Grenoble Alpes, Inserm, CEA, UA13 BGE, 38000 Grenoble, France
| | | | - Rima Slim
- Departments of Human Genetics and Obstetrics and Gynecology, McGill University Health Centre Research Institute, Montréal, QC H4A 3J1, Canada
| | - Mohamed Benharouga
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38043 Grenoble, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique, University Grenoble Alpes and Centre Hospitalo-Universitaire Grenoble Alpes, CS 10217, CEDEX 9, 38043 Grenoble, France
| | - Roland Abi Nahed
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38043 Grenoble, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique, University Grenoble Alpes and Centre Hospitalo-Universitaire Grenoble Alpes, CS 10217, CEDEX 9, 38043 Grenoble, France
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), Univeristy Grenoble Alpes, Inserm, 38000 Grenoble, France
| |
Collapse
|
8
|
Modulation of Nod-like Receptor Expression in the Thymus during Early Pregnancy in Ewes. Vaccines (Basel) 2022; 10:vaccines10122128. [PMID: 36560538 PMCID: PMC9781860 DOI: 10.3390/vaccines10122128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/24/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Nucleotide-binding oligomerization domain receptors (NOD-like receptors, NLRs) are involved in modulating the innate immune responses of the trophoblast and the placenta in normal pregnancy. The thymus participates in regulation of innate and adaptive immune responses. However, it is unclear whether expression of NLR is modulated in the maternal thymus during early pregnancy. In this study, thymuses were sampled at day 16 of the estrous cycle, and at days 13, 16 and 25 of gestation (n = 6 for each group) from ewes after slaughter. Different stages were chosen because the maternal thymus was under the different effects of interferon-tau and/or progesterone or not. RT-qPCR, Western blot and immunohistochemistry analysis were used to analyze the expression of the NLR family, including NOD1; NOD2; major histocompatibility complex class II transactivator (CIITA); NLR family apoptosis inhibitory protein (NAIP); nucleotide-binding oligomerization domain and Leucine-rich repeat and Pyrin domain containing protein 1 (NLRP1), NLRP3 and NLRP7. The results showed that expression level of NOD1 was changed with the pregnancy stages, and expression levels of NOD2, CIITA, NAIP, NLRP1, NLRP3 and NLRP7 mRNA and proteins were peaked at day 13 of pregnancy. The levels of NOD2 and CIITA were increased during early pregnancy. The stainings for NOD2 and NLRP7 proteins were located in epithelial reticular cells, capillaries and thymic corpuscles. In summary, pregnancy stages changed expression of NLR family in the maternal thymus, which may be related to the modulation of maternal thymic immune responses, and beneficial for normal pregnancy in sheep.
Collapse
|
9
|
Marini HR, Micali A, Squadrito G, Puzzolo D, Freni J, Antonuccio P, Minutoli L. Nutraceuticals: A New Challenge against Cadmium-Induced Testicular Injury. Nutrients 2022; 14:663. [PMID: 35277022 PMCID: PMC8838120 DOI: 10.3390/nu14030663] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 12/04/2022] Open
Abstract
Cadmium (Cd) is a widespread heavy metal and a ubiquitous environmental toxicant. For the general population, the principal causes of Cd exposure are cigarette smoking, air pollution and contaminated water and food consumption, whereas occupational exposure usually involves humans working in mines or manufacturing batteries and pigments that utilize Cd. The aim of the present review is to evaluate recent data regarding the mechanisms of Cd-induced testicular structural and functional damages and the state of the art of the therapeutic approaches. Additionally, as the current literature demonstrates convincing associations between diet, food components and men's sexual health, a coherent nutraceutical supplementation may be a new valid therapeutic strategy for both the prevention and alleviation of Cd-induced testicular injury. The toxic effects on testes induced by Cd include many specific mechanisms, such as oxidative stress, inflammation and apoptosis. As no specific therapy for the prevention or treatment of the morbidity and mortality associated with Cd exposure is available, the development of new therapeutic agents is requested. Dietary strategies and the use of nutraceuticals, particularly abundant in fresh fruits, beans, vegetables and grains, typical of the Mediterranean diet, are recommended against Cd-induced testicular injury.
Collapse
Affiliation(s)
- Herbert Ryan Marini
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (G.S.); (L.M.)
| | - Antonio Micali
- Department of Human Pathology of Adult and Childhood, University of Messina, 98125 Messina, Italy; (A.M.); (P.A.)
| | - Giovanni Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (G.S.); (L.M.)
| | - Domenico Puzzolo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy;
| | - José Freni
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy;
| | - Pietro Antonuccio
- Department of Human Pathology of Adult and Childhood, University of Messina, 98125 Messina, Italy; (A.M.); (P.A.)
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (G.S.); (L.M.)
| |
Collapse
|
10
|
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
|
11
|
Li G, Tian X, Lv D, Zhang L, Zhang Z, Wang J, Yang M, Tao J, Ma T, Wu H, Ji P, Wu Y, Lian Z, Cui W, Liu G. NLRP7 is expressed in the ovine ovary and associated with in vitro pre-implantation embryo development. Reproduction 2020; 158:415-427. [PMID: 31505467 PMCID: PMC6826174 DOI: 10.1530/rep-19-0081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 09/10/2019] [Indexed: 12/13/2022]
Abstract
NLRP (NACHT, LRR and PYD domain-containing proteins) family plays pivotal roles in mammalian reproduction. Mutation of NLRP7 is often associated with human recurrent hydatidiform moles. Few studies regarding the functions of NLRP7 have been performed in other mammalian species rather than humans. In the current study, for the first time, the function of NLRP7 has been explored in ovine ovary. NLRP7 protein was mainly located in ovarian follicles and in in vitro pre-implantation embryos. To identify its origin, 763 bp partial CDS of NLRP7 deriving from sheep cumulus oocyte complexes (COCs) was cloned, it showed a great homology with Homo sapiens. The high levels of mRNA and protein of NLRP7 were steadily expressed in oocytes, parthenogenetic embryos or IVF embryos. NLRP7 knockdown by the combination of siRNA and shRNA jeopardized both the parthenogenetic and IVF embryo development. These results strongly suggest that NLRP7 plays an important role in ovine reproduction. The potential mechanisms of NLRP7 will be fully investigated in the future.
Collapse
Affiliation(s)
- Guangdong Li
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiuzhi Tian
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Dongying Lv
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lu Zhang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhenzhen Zhang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jing Wang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Minghui Yang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jingli Tao
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Teng Ma
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hao Wu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Pengyun Ji
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yingjie Wu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhengxing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Wei Cui
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Guoshi Liu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
12
|
Li Z, Tang J, Wen W, Wu W, Wang J, Xu J, Yu Y, He Z, Pan X, Wei H, Zhu Y, Hu S, Cao J, Shen H, Que J, Wang W, Zhu Q, Chen L. Systematic analysis of genetic variants in cancer-testis genes identified two novel lung cancer susceptibility loci in Chinese population. J Cancer 2020; 11:1985-1993. [PMID: 32194810 PMCID: PMC7052880 DOI: 10.7150/jca.40002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/25/2019] [Indexed: 12/29/2022] Open
Abstract
Cancer-testis (CT) genes played important roles in the progression of malignant tumors and were recognized as promising therapeutic targets. However, the roles of genetic variants in CT genes in lung cancer susceptibility have not been well depicted. This study aimed to evaluate the associations between genetic variants in CT genes and lung cancer risk in Chinese population. A total of 22,556 qualified SNPs from 268 lung cancer associated CT genes were initially evaluated based on our previous lung cancer GWAS (Genome-wide association studies) with 2,331 cases and 3,077 controls. As a result, 17 candidate SNPs were further genotyped in 1,056 cases and 1,053 controls using Sequenom platform. Two variants (rs6941653, OPRM1, T > C, screening: OR = 1.24, 95%CI: 1.12-1.38, P = 2.40×10-5; validation: OR = 1.18, 95%CI: 1.01-1.37, P = 0.039 and rs402969, NLRP8, C > T, screening: OR = 1.15, 95%CI: 1.04-1.26, P = 0.006; validation: OR = 1.16, 95%CI: 1.02-1.33, P = 0.028) were identified as novel lung cancer susceptibility variants. Stratification analysis indicated that the effect of rs6941653 was stronger in lung squamous cell carcinoma (OR = 1.36) than that in lung adenocarcinoma (OR = 1.15, I2 = 77%, P = 0.04). Finally, functional annotations, differential gene expression analysis, pathway and gene ontology analyses were performed to suggest the potential functions of our identified variants and genes. In conclusion, this study identified two novel lung cancer risk variants in Chinese population and provided deeper insight into the roles of CT genes in lung tumorigenesis.
Collapse
Affiliation(s)
- Zhihua Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jianwei Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wei Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Weibing Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jun Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jing Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yue Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhicheng He
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xianglong Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Haixing Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yining Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shuo Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jing Cao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, International Joint Research Center, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center of Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Jun Que
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Quan Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
13
|
Abstract
Imprinting disorders are a group of congenital diseases caused by dysregulation of genomic imprinting, affecting prenatal and postnatal growth, neurocognitive development, metabolism and cancer predisposition. Aberrant expression of imprinted genes can be achieved through different mechanisms, classified into epigenetic - if not involving DNA sequence change - or genetic in the case of altered genomic sequence. Despite the underlying mechanism, the phenotype depends on the parental allele affected and opposite phenotypes may result depending on the involvement of the maternal or the paternal chromosome. Imprinting disorders are largely underdiagnosed because of the broad range of clinical signs, the overlap of presentation among different disorders, the presence of mild phenotypes, the mitigation of the phenotype with age and the limited availability of molecular techniques employed for diagnosis. This review briefly illustrates the currently known human imprinting disorders, highlighting endocrinological aspects of pediatric interest.
Collapse
Affiliation(s)
- Diana Carli
- University of Torino, Department of Pediatric and Public Health Sciences, Torino, Italy
| | - Evelise Riberi
- University of Torino, Department of Pediatric and Public Health Sciences, Torino, Italy
| | | | - Alessandro Mussa
- University of Torino, Department of Pediatric and Public Health Sciences, Torino, Italy,* Address for Correspondence: University of Torino, Department of Pediatric and Public Health Sciences, Torino, Italy Phone: +39-011-313-1985 E-mail:
| |
Collapse
|
14
|
Lim RR, Wieser ME, Ganga RR, Barathi VA, Lakshminarayanan R, Mohan RR, Hainsworth DP, Chaurasia SS. NOD-like Receptors in the Eye: Uncovering Its Role in Diabetic Retinopathy. Int J Mol Sci 2020; 21:E899. [PMID: 32019187 PMCID: PMC7037099 DOI: 10.3390/ijms21030899] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/22/2020] [Accepted: 01/27/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetic retinopathy (DR) is an ocular complication of diabetes mellitus (DM). International Diabetic Federations (IDF) estimates up to 629 million people with DM by the year 2045 worldwide. Nearly 50% of DM patients will show evidence of diabetic-related eye problems. Therapeutic interventions for DR are limited and mostly involve surgical intervention at the late-stages of the disease. The lack of early-stage diagnostic tools and therapies, especially in DR, demands a better understanding of the biological processes involved in the etiology of disease progression. The recent surge in literature associated with NOD-like receptors (NLRs) has gained massive attraction due to their involvement in mediating the innate immune response and perpetuating inflammatory pathways, a central phenomenon found in the pathogenesis of ocular diseases including DR. The NLR family of receptors are expressed in different eye tissues during pathological conditions suggesting their potential roles in dry eye, ocular infection, retinal ischemia, cataract, glaucoma, age-related macular degeneration (AMD), diabetic macular edema (DME) and DR. Our group is interested in studying the critical early components involved in the immune cell infiltration and inflammatory pathways involved in the progression of DR. Recently, we reported that NLRP3 inflammasome might play a pivotal role in the pathogenesis of DR. This comprehensive review summarizes the findings of NLRs expression in the ocular tissues with special emphasis on its presence in the retinal microglia and DR pathogenesis.
Collapse
Affiliation(s)
- Rayne R. Lim
- Ocular Immunology and Angiogenesis Lab, University of Missouri, Columbia, MO 652011, USA; (R.R.L.); (M.E.W.); (R.R.M.)
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 652011, USA
- Ophthalmology, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 652011, USA
| | - Margaret E. Wieser
- Ocular Immunology and Angiogenesis Lab, University of Missouri, Columbia, MO 652011, USA; (R.R.L.); (M.E.W.); (R.R.M.)
| | - Rama R. Ganga
- Surgery, University of Missouri, Columbia, MO 652011, USA;
| | | | | | - Rajiv R. Mohan
- Ocular Immunology and Angiogenesis Lab, University of Missouri, Columbia, MO 652011, USA; (R.R.L.); (M.E.W.); (R.R.M.)
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 652011, USA
- Ophthalmology, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 652011, USA
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 652011, USA;
| | - Dean P. Hainsworth
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 652011, USA;
| | - Shyam S. Chaurasia
- Ocular Immunology and Angiogenesis Lab, University of Missouri, Columbia, MO 652011, USA; (R.R.L.); (M.E.W.); (R.R.M.)
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 652011, USA
- Ophthalmology, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 652011, USA
| |
Collapse
|
15
|
Tsai PY, Chen KR, Li YC, Kuo PL. NLRP7 Is Involved in the Differentiation of the Decidual Macrophages. Int J Mol Sci 2019; 20:E5994. [PMID: 31795138 PMCID: PMC6929161 DOI: 10.3390/ijms20235994] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 01/14/2023] Open
Abstract
Macrophage polarization, regulated appropriately, may play important roles in successful pregnancy. In the face of the vital roles of decidua macrophages in pregnancy, it is insufficient to recognize the trigger of macrophage differentiation and polarization. We aimed to explore the link between the NLRP7 gene and macrophage polarization in human deciduas. Here, we enrolled the endometrial tissues from eight pregnant women in the first trimester. We found that NLRP7 was abundant in endometrial tissues and that NLRP7 was expressed in decidual macrophages of the first-trimester pregnancy. NLRP7 was predominately expressed in the decidual M2 macrophages, as compared with the M1 macrophages. Furthermore, our results suggest that NLRP7 is associated with decidual macrophage differentiation. NLRP7 over-expression suppresses the expression of M1 markers and enhances the expression of the M2 markers. Considering that NLRP7 relates to decidualization and macrophage differentiation, we propose that NLRP7 is a primate-specific multitasking gene to maintain endometrial hemostasis and reproductive success. This finding may pave the way for therapies of pathological pregnancies.
Collapse
Affiliation(s)
- Pei-Yin Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70401, Taiwan; (P.-Y.T.); (K.-R.C.)
| | - Kuan-Ru Chen
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70401, Taiwan; (P.-Y.T.); (K.-R.C.)
| | - Yueh-Chun Li
- Laboratory of cytogenetic research, Lee Women’s Hospital, Taichung 40652, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70401, Taiwan; (P.-Y.T.); (K.-R.C.)
| |
Collapse
|
16
|
Assrawi E, Louvrier C, Lepelletier C, Georgin-Lavialle S, Bouaziz JD, Awad F, Moinet F, Moguelet P, Vignon-Pennamen MD, Piterboth W, Jumeau C, Cobret L, El Khouri E, Copin B, Duquesnoy P, Legendre M, Grateau G, Karabina SA, Amselem S, Giurgea I. Somatic Mosaic NLRP3 Mutations and Inflammasome Activation in Late-Onset Chronic Urticaria. J Invest Dermatol 2019; 140:791-798.e2. [PMID: 31513803 DOI: 10.1016/j.jid.2019.06.153] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 01/20/2023]
Abstract
Chronic urticaria is a common skin disorder with heterogeneous causes. In the absence of physical triggers, chronic urticarial rash is called idiopathic or spontaneous. The objective of this study was to identify the molecular and cellular bases of a disease condition displayed by two unrelated patients aged over 60 years who presented for two decades with a chronic urticaria resistant to standard therapy that occurred in the context of systemic inflammation not triggered by cold. In both patients, a targeted sequencing approach using a next generation technology identified somatic mosaic mutations in NLRP3, a gene encoding a key inflammasome component. The study of several of both patients' cell types showed that, despite the late onset of the disease, NLRP3 mutations were not found to be restricted to myelomonocytic cells. Rather, the data obtained strongly suggested that the mutational event occurred very early, during embryonic development. As shown by functional studies, the identified mutations-an in-frame deletion and a recurrent NLRP3 missense mutation-have a gain-of-function effect on NLRP3-inflammasome activation. Consistently, a complete remission was obtained in both patients with anti-IL-1 receptor antagonists. This study unveils that in late-onset chronic urticaria, the search for autoinflammatory markers and somatic mosaic NLRP3 mutations may have important diagnostic and therapeutic consequences.
Collapse
Affiliation(s)
- Eman Assrawi
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France
| | - Camille Louvrier
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France; Unité Fonctionnelle de génétique moléculaire, Assistance Publique-Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - Clémence Lepelletier
- Service de dermatologie, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
| | - Sophie Georgin-Lavialle
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France; Service de médecine interne, Assistance Publique-Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Jean-David Bouaziz
- Service de dermatologie, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
| | - Fawaz Awad
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France; Current address: Al-Quds University, Faculty of Medicine, Biochemistry and Molecular Biology Department, Abu Deis, Jerusalem, Palestine
| | - Florence Moinet
- Service de médecine interne, Centre Hospitalier Universitaire de Martinique, Martinique, Fort de France, France
| | - Philippe Moguelet
- Unité d'anatomie et cytologie pathologiques, Assistance Publique-Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | | | - William Piterboth
- Unité Fonctionnelle de génétique moléculaire, Assistance Publique-Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - Claire Jumeau
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France
| | - Laetitia Cobret
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France
| | - Elma El Khouri
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France
| | - Bruno Copin
- Unité Fonctionnelle de génétique moléculaire, Assistance Publique-Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - Philippe Duquesnoy
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France
| | - Marie Legendre
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France; Unité Fonctionnelle de génétique moléculaire, Assistance Publique-Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - Gilles Grateau
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France; Service de médecine interne, Assistance Publique-Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Sonia A Karabina
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France
| | - Serge Amselem
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France; Unité Fonctionnelle de génétique moléculaire, Assistance Publique-Hôpitaux de Paris, Hôpital Trousseau, Paris, France.
| | - Irina Giurgea
- Sorbonne Université, INSERM, Hôpital Trousseau, Maladies génétiques d'expression pédiatrique, Paris, France; Unité Fonctionnelle de génétique moléculaire, Assistance Publique-Hôpitaux de Paris, Hôpital Trousseau, Paris, France.
| |
Collapse
|
17
|
Mu J, Wang W, Chen B, Wu L, Li B, Mao X, Zhang Z, Fu J, Kuang Y, Sun X, Li Q, Jin L, He L, Sang Q, Wang L. Mutations in NLRP2 and NLRP5 cause female infertility characterised by early embryonic arrest. J Med Genet 2019; 56:471-480. [PMID: 30877238 DOI: 10.1136/jmedgenet-2018-105936] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/23/2019] [Accepted: 02/12/2019] [Indexed: 11/03/2022]
Abstract
BACKGROUND Successful human reproduction requires normal spermatogenesis, oogenesis, fertilisation and early embryonic development, and abnormalities in any of these processes will result in infertility. Early embryonic arrest is commonly observed in infertile patients with recurrent failure of assisted reproductive technology (ART). However, the genetic basis for early embryonic arrest is largely unknown. OBJECTIVE We aim to identify genetic causes of infertile patients characterised by early embryonic arrest. METHODS We pursued exome sequencing in a proband with embryonic arrest from the consanguineous family. We further screened candidate genes in a cohort of 496 individuals diagnosed with early embryonic arrest by Sanger sequencing. Effects of mutations were investigated in HeLa cells, oocytes and embryos. RESULTS We identified five independent individuals carrying biallelic mutations in NLRP2. We also found three individuals from two families carrying biallelic mutations in NLRP5. These mutations in NLRP2 and NLRP5 caused decreased protein expression in vitro and in oocytes and embryos. CONCLUSIONS NLRP2 and NLRP5 are novel mutant genes responsible for human early embryonic arrest. This finding provides additional potential diagnostic markers for patients with recurrent failure of ART and helps us to better understand the genetic basis of female infertility characterised by early embryonic arrest.
Collapse
Affiliation(s)
- Jian Mu
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, China
| | - Wenjing Wang
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, China
| | - Biaobang Chen
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, China
| | - Ling Wu
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Li
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyan Mao
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhihua Zhang
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Fu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yanping Kuang
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Qiaoli Li
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, China
| | - Lin He
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Sang
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, China
| | - Lei Wang
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, China
- Shanghai Center for Women and Children's Health, Shanghai, China
| |
Collapse
|
18
|
Lacey CJ, Doudney K, Bridgman PG, George PM, Mulder RT, Zarifeh JJ, Kimber B, Cadzow MJ, Black MA, Merriman TR, Lehnert K, Bickley VM, Pearson JF, Cameron VA, Kennedy MA. Copy number variants implicate cardiac function and development pathways in earthquake-induced stress cardiomyopathy. Sci Rep 2018; 8:7548. [PMID: 29765130 PMCID: PMC5954162 DOI: 10.1038/s41598-018-25827-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 04/25/2018] [Indexed: 02/07/2023] Open
Abstract
The pathophysiology of stress cardiomyopathy (SCM), also known as takotsubo syndrome, is poorly understood. SCM usually occurs sporadically, often in association with a stressful event, but clusters of cases are reported after major natural disasters. There is some evidence that this is a familial condition. We have examined three possible models for an underlying genetic predisposition to SCM. Our primary study cohort consists of 28 women who suffered SCM as a result of two devastating earthquakes that struck the city of Christchurch, New Zealand, in 2010 and 2011. To seek possible underlying genetic factors we carried out exome analysis, genotyping array analysis, and array comparative genomic hybridization on these subjects. The most striking finding was the observation of a markedly elevated rate of rare, heterogeneous copy number variants (CNV) of uncertain clinical significance (in 12/28 subjects). Several of these CNVs impacted on genes of cardiac relevance including RBFOX1, GPC5, KCNRG, CHODL, and GPBP1L1. There is no physical overlap between the CNVs, and the genes they impact do not appear to be functionally related. The recognition that SCM predisposition may be associated with a high rate of rare CNVs offers a novel perspective on this enigmatic condition.
Collapse
Affiliation(s)
- Cameron J Lacey
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand.
| | - Kit Doudney
- Molecular Pathology Laboratory, Canterbury Health Laboratories, Canterbury District Health Board, Christchurch, New Zealand
| | - Paul G Bridgman
- Department of Cardiology, Christchurch Hospital, Christchurch, New Zealand
| | - Peter M George
- Molecular Pathology Laboratory, Canterbury Health Laboratories, Canterbury District Health Board, Christchurch, New Zealand
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Roger T Mulder
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Julie J Zarifeh
- Psychiatric Consultation Service, Christchurch Hospital, Canterbury District Health Board, Christchurch, New Zealand
| | - Bridget Kimber
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Murray J Cadzow
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Michael A Black
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Klaus Lehnert
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Vivienne M Bickley
- Molecular Pathology Laboratory, Canterbury Health Laboratories, Canterbury District Health Board, Christchurch, New Zealand
| | - John F Pearson
- Biostatistics and Computational Biology Unit, University of Otago, Christchurch, New Zealand
| | - Vicky A Cameron
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Martin A Kennedy
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
19
|
Zhai R, Yuan Y, Jiao F, Hao F, Fang X, Zhang Y, Qian X. Facile synthesis of magnetic metal organic frameworks for highly efficient proteolytic digestion used in mass spectrometry-based proteomics. Anal Chim Acta 2017; 994:19-28. [DOI: 10.1016/j.aca.2017.08.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/23/2017] [Accepted: 08/26/2017] [Indexed: 12/12/2022]
|
20
|
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
|
21
|
Meunier E, Broz P. Evolutionary Convergence and Divergence in NLR Function and Structure. Trends Immunol 2017; 38:744-757. [PMID: 28579324 DOI: 10.1016/j.it.2017.04.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/13/2017] [Accepted: 04/21/2017] [Indexed: 12/11/2022]
Abstract
The recognition of cellular damage caused by either pathogens or abiotic stress is essential for host defense in all forms of life in the plant and animal kingdoms. The NOD-like receptors (NLRs) represent a large family of multidomain proteins that were initially discovered for their role in host defense in plants and vertebrates. Over recent years the wide distribution of NLRs among metazoans has become apparent and their origins have begun to emerge. Moreover, intense study of NLR function has shown that they play essential roles beyond pathogen recognition - in the regulation of antigen presentation, cell death, inflammation, and even in embryonic development. We summarize here the latest insights into NLR biology and discuss examples of converging and diverging evolution of NLR function and structure.
Collapse
Affiliation(s)
- Etienne Meunier
- Focal Area Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland; Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, France
| | - Petr Broz
- Focal Area Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
22
|
Sanchez-Delgado M, Riccio A, Eggermann T, Maher ER, Lapunzina P, Mackay D, Monk D. Causes and Consequences of Multi-Locus Imprinting Disturbances in Humans. Trends Genet 2016; 32:444-455. [PMID: 27235113 DOI: 10.1016/j.tig.2016.05.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/20/2022]
Abstract
Eight syndromes are associated with the loss of methylation at specific imprinted loci. There has been increasing evidence that these methylation defects in patients are not isolated events occurring at a given disease-associated locus but that some of these patients may have multi-locus imprinting disturbances (MLID) affecting additional imprinted regions. With the recent advances in technology, methylation profiling has revealed that imprinted loci represent only a small fraction of the methylation differences observed between the gametes. To figure out how imprinting anomalies occur at multiple imprinted domains, we have to understand the interplay between DNA methylation and histone modifications in the process of selective imprint protection during pre-implantation reprogramming, which, if disrupted, leads to these complex imprinting disorders (IDs).
Collapse
Affiliation(s)
- Marta Sanchez-Delgado
- Imprinting and Cancer group, Cancer Epigenetic and Biology Program, Institut d'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona, Spain
| | - Andrea Riccio
- DiSTABiF, Seconda Università degli Studi di Napoli, Caserta; Institute of Genetics and Biophysics - ABT, CNR, Napoli, Italy
| | - Thomas Eggermann
- Institute of Human Genetics University Hospital Aachen, Aachen, Germany
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Pablo Lapunzina
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain; CIBERER, Centro deInvestigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Deborah Mackay
- Human Genetics and Genomic Medicine, Faculty of Medicine University of Southampton, Southampton, UK
| | - David Monk
- Imprinting and Cancer group, Cancer Epigenetic and Biology Program, Institut d'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona, Spain.
| |
Collapse
|
23
|
Minutoli L, Antonuccio P, Irrera N, Rinaldi M, Bitto A, Marini H, Pizzino G, Romeo C, Pisani A, Santoro G, Puzzolo D, Magno C, Squadrito F, Micali A, Altavilla D. NLRP3 Inflammasome Involvement in the Organ Damage and Impaired Spermatogenesis Induced by Testicular Ischemia and Reperfusion in Mice. J Pharmacol Exp Ther 2015; 355:370-80. [PMID: 26407722 DOI: 10.1124/jpet.115.226936] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/24/2015] [Indexed: 01/05/2023] Open
Abstract
We investigated the role of the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome during testis ischemia and reperfusion injury (TI/R) in wild-type (WT) and NLRP3 knock-out (KO) mice. WT and KO mice underwent 1 hour testicular ischemia followed by 4 hours and 1 and 7 days of reperfusion or a sham TI/R. Furthermore, two groups of WT mice were treated at the beginning of reperfusion and up to 7 days with two inflammasome inhibitors, BAY 11-7082 (20 mg/kg i.p.) or Brilliant Blue G (45.5 mg/kg i.p.), or vehicle. Animals were killed with a pentobarbital sodium overdose at 4 hours and 1 and 7 days, and bilateral orchidectomies were performed. Biochemical and morphologic studies were carried out in all groups. TI/R in WT mice significantly increased caspase-1 and interleukin (IL)-1β mRNA after 4 hours and IL-18 mRNA at 1 day of reperfusion (P ≤ 0.05). There was also a significant increase in caspase-3 and terminal deoxynucleotidyl transferase-mediated digoxigenin-deoxyuridine nick-end labeling-positive cells, marked histologic damage, and altered spermatogenesis in WT mice in both testes after 1 and 7 days of reperfusion. KO TI/R mice, WT TI/R BAY 11-7082, and Brilliant Blue G treated mice showed a significant reduced IL-1β and IL-18 mRNA expression, blunted caspase-1 and -3 expression, minor histologic damages, low terminal deoxynucleotidyl transferase-mediated digoxigenin-deoxyuridine nick-end labeling activity, and preserved spermatogenesis. These data suggest that the activation of NLRP3 plays a key role in TI/R, and its inhibition might represent a therapeutic target for the management of patients with unilateral testicular torsion.
Collapse
Affiliation(s)
- Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Pietro Antonuccio
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Mariagrazia Rinaldi
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Herbert Marini
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Gabriele Pizzino
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Carmelo Romeo
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Antonina Pisani
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Giuseppe Santoro
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Domenico Puzzolo
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Carlo Magno
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Antonio Micali
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| | - Domenica Altavilla
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico "G. Martino", Messina, Italy (C.M.)
| |
Collapse
|
24
|
Sanchez-Delgado M, Martin-Trujillo A, Tayama C, Vidal E, Esteller M, Iglesias-Platas I, Deo N, Barney O, Maclean K, Hata K, Nakabayashi K, Fisher R, Monk D. Absence of Maternal Methylation in Biparental Hydatidiform Moles from Women with NLRP7 Maternal-Effect Mutations Reveals Widespread Placenta-Specific Imprinting. PLoS Genet 2015; 11:e1005644. [PMID: 26544189 PMCID: PMC4636177 DOI: 10.1371/journal.pgen.1005644] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 10/12/2015] [Indexed: 11/18/2022] Open
Abstract
Familial recurrent hydatidiform mole (RHM) is a maternal-effect autosomal recessive disorder usually associated with mutations of the NLRP7 gene. It is characterized by HM with excessive trophoblastic proliferation, which mimics the appearance of androgenetic molar conceptuses despite their diploid biparental constitution. It has been proposed that the phenotypes of both types of mole are associated with aberrant genomic imprinting. However no systematic analyses for imprinting defects have been reported. Here, we present the genome-wide methylation profiles of both spontaneous androgenetic and biparental NLRP7 defective molar tissues. We observe total paternalization of all ubiquitous and placenta-specific differentially methylated regions (DMRs) in four androgenetic moles; namely gain of methylation at paternally methylated loci and absence of methylation at maternally methylated regions. The methylation defects observed in five RHM biopsies from NLRP7 defective patients are restricted to lack-of-methylation at maternal DMRs. Surprisingly RHMs from two sisters with the same missense mutations, as well as consecutive RHMs from one affected female show subtle allelic methylation differences, suggesting inter-RHM variation. These epigenotypes are consistent with NLRP7 being a maternal-effect gene and involved in imprint acquisition in the oocyte. In addition, bioinformatic screening of the resulting methylation datasets identified over sixty loci with methylation profiles consistent with imprinting in the placenta, of which we confirm 22 as novel maternally methylated loci. These observations strongly suggest that the molar phenotypes are due to defective placenta-specific imprinting and over-expression of paternally expressed transcripts, highlighting that maternal-effect mutations of NLRP7 are associated with the most severe form of multi-locus imprinting defects in humans.
Collapse
Affiliation(s)
- Marta Sanchez-Delgado
- Imprinting and Cancer Group, Cancer Epigenetic and Biology Program, Institut d’Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona, Spain
| | - Alejandro Martin-Trujillo
- Imprinting and Cancer Group, Cancer Epigenetic and Biology Program, Institut d’Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona, Spain
| | - Chiharu Tayama
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Enrique Vidal
- Cancer Epigenetics Group, Cancer Epigenetic and Biology Program, Institut d’Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Cancer Epigenetic and Biology Program, Institut d’Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona, Spain
- Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Spain
- Institucio Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Isabel Iglesias-Platas
- Servicio de Neonatología, Hospital Sant Joan de Déu, Fundació Sant Joan de Déu, Barcelona, Spain
| | - Nandita Deo
- Whipps Cross University Hospital, Barts Health NHS Trust, Leytonstone, London, United Kingdom
| | - Olivia Barney
- Leicester Royal Infirmary, Leicester, United Kingdom
| | | | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Rosemary Fisher
- Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- Trophoblastic Tumour Screening and Treatment Centre, Department of Oncology, Imperial College London, London, United Kingdom
| | - David Monk
- Imprinting and Cancer Group, Cancer Epigenetic and Biology Program, Institut d’Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona, Spain
- * E-mail:
| |
Collapse
|
25
|
Elvers I, Turner-Maier J, Swofford R, Koltookian M, Johnson J, Stewart C, Zhang CZ, Schumacher SE, Beroukhim R, Rosenberg M, Thomas R, Mauceli E, Getz G, Palma FD, Modiano JF, Breen M, Lindblad-Toh K, Alföldi J. Exome sequencing of lymphomas from three dog breeds reveals somatic mutation patterns reflecting genetic background. Genome Res 2015; 25:1634-45. [PMID: 26377837 PMCID: PMC4617960 DOI: 10.1101/gr.194449.115] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/16/2015] [Indexed: 12/11/2022]
Abstract
Lymphoma is the most common hematological malignancy in developed countries. Outcome is strongly determined by molecular subtype, reflecting a need for new and improved treatment options. Dogs spontaneously develop lymphoma, and the predisposition of certain breeds indicates genetic risk factors. Using the dog breed structure, we selected three lymphoma predisposed breeds developing primarily T-cell (boxer), primarily B-cell (cocker spaniel), and with equal distribution of B- and T-cell lymphoma (golden retriever), respectively. We investigated the somatic mutations in B- and T-cell lymphomas from these breeds by exome sequencing of tumor and normal pairs. Strong similarities were evident between B-cell lymphomas from golden retrievers and cocker spaniels, with recurrent mutations in TRAF3-MAP3K14 (28% of all cases), FBXW7 (25%), and POT1 (17%). The FBXW7 mutations recurrently occur in a specific codon; the corresponding codon is recurrently mutated in human cancer. In contrast, T-cell lymphomas from the predisposed breeds, boxers and golden retrievers, show little overlap in their mutation pattern, sharing only one of their 15 most recurrently mutated genes. Boxers, which develop aggressive T-cell lymphomas, are typically mutated in the PTEN-mTOR pathway. T-cell lymphomas in golden retrievers are often less aggressive, and their tumors typically showed mutations in genes involved in cellular metabolism. We identify genes with known involvement in human lymphoma and leukemia, genes implicated in other human cancers, as well as novel genes that could allow new therapeutic options.
Collapse
Affiliation(s)
- Ingegerd Elvers
- Broad Institute, Cambridge, Massachusetts 02142, USA; Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala SE 751 23, Sweden
| | | | - Ross Swofford
- Broad Institute, Cambridge, Massachusetts 02142, USA
| | | | | | - Chip Stewart
- Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Cheng-Zhong Zhang
- Broad Institute, Cambridge, Massachusetts 02142, USA; Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Steven E Schumacher
- Broad Institute, Cambridge, Massachusetts 02142, USA; Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Rameen Beroukhim
- Broad Institute, Cambridge, Massachusetts 02142, USA; Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | | | - Rachael Thomas
- North Carolina State University, Raleigh, North Carolina 27695, USA
| | - Evan Mauceli
- Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Gad Getz
- Broad Institute, Cambridge, Massachusetts 02142, USA; Harvard Medical School, Boston, Massachusetts 02115, USA; Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | - Jaime F Modiano
- Animal Cancer Care and Research Program, College of Veterinary Medicine, and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Matthew Breen
- North Carolina State University, Raleigh, North Carolina 27695, USA; University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina 27514, USA
| | - Kerstin Lindblad-Toh
- Broad Institute, Cambridge, Massachusetts 02142, USA; Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala SE 751 23, Sweden
| | | |
Collapse
|
26
|
Monk D. Germline-derived DNA methylation and early embryo epigenetic reprogramming: The selected survival of imprints. Int J Biochem Cell Biol 2015; 67:128-38. [PMID: 25966912 DOI: 10.1016/j.biocel.2015.04.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/27/2015] [Indexed: 12/27/2022]
Abstract
DNA methylation is an essential epigenetic mechanism involved in many essential cellular processes. During development epigenetic reprograming takes place during gametogenesis and then again in the pre-implantation embryo. These two reprograming windows ensure genome-wide removal of methylation in the primordial germ cells so that sex-specific signatures can be acquired in the sperm and oocyte. Following fertilization the majority of this epigenetic information is erased to give the developing embryo an epigenetic profile coherent with pluripotency. It is estimated that ∼65% of the genome is differentially methylated between the gametes, however following embryonic reprogramming only parent-of-origin methylation at known imprinted loci remains. This suggests that trans-acting factors such as Zfp57 can discriminate imprinted differentially methylated regions (DMRs) from the thousands of CpG rich regions that are differentially marked in the gametes. Recently transient imprinted DMRs have been identified suggesting that these loci are also protected from pre-implantation reprograming but succumb to de novo remethylation at the implantation stage. This highlights that "ubiquitous" imprinted loci are also resilient to gaining methylation by protecting their unmethylated alleles. In this review I examine the processes involved in epigenetic reprograming and the mechanisms that ensure allelic methylation at imprinted loci is retained throughout the life of the organism, discussing the critical differences between mouse and humans. This article is part of a Directed Issue entitled: Epigenetics Dynamics in development and disease.
Collapse
Affiliation(s)
- David Monk
- Imprinting and Cancer group, Cancer Epigenetic and Biology Program, Institut d'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona 08908, Spain.
| |
Collapse
|
27
|
Huber RG, Eibl C, Fuchs JE. Intrinsic flexibility of NLRP pyrin domains is a key factor in their conformational dynamics, fold stability, and dimerization. Protein Sci 2014; 24:174-81. [PMID: 25403012 DOI: 10.1002/pro.2601] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/09/2014] [Indexed: 01/12/2023]
Abstract
Nucleotide-binding domain leucine-rich repeat-containing receptors (NLRs) are key proteins in the innate immune system. The 14 members of the NLRP family of NLRs contain an N-terminal pyrin domain which is central for complex formation and signal transduction. Recently, X-ray structures of NLRP14 revealed an unexpected rearrangement of the α5/6 stem-helix of the pyrin domain allowing a novel symmetric dimerization mode. We characterize the conformational transitions underlying NLRP oligomerization using molecular dynamics simulations. We describe conformational stability of native NLRP14 and mutants in their monomeric and dimeric states and compare them to NLRP4, a representative of a native pyrin domain fold. Thereby, we characterize the interplay of conformational dynamics, fold stability, and dimerization in NLRP pyrin domains. We show that intrinsic flexibility of NLRP pyrin domains is a key factor influencing their behavior in physiological conditions. Additionally, we provide further evidence for the crucial importance of a charge relay system within NLRPs that critically influences their conformational ensemble in solution.
Collapse
Affiliation(s)
- Roland G Huber
- Institute for General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain, 80/82, Innsbruck, Austria; Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street #07-01 Matrix, Singapore, 138671
| | | | | |
Collapse
|
28
|
Akoury E, Zhang L, Ao A, Slim R. NLRP7 and KHDC3L, the two maternal-effect proteins responsible for recurrent hydatidiform moles, co-localize to the oocyte cytoskeleton. Hum Reprod 2014; 30:159-69. [PMID: 25358348 DOI: 10.1093/humrep/deu291] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
STUDY QUESTION What is the subcellular localization in human oocytes and preimplantation embryos, of the two maternal-effect proteins, NLRP7 and KHDC3L, responsible for recurrent hydatidiform moles (RHMs)? SUMMARY ANSWER NLRP7 and KHDC3L localize to the oocyte cytoskeleton and are polar and absent from the cell-to-cell contact region in early preimplantation embryos. WHAT IS KNOWN ALREADY NLRP7 and KHDC3L expression has been described at the RNA level in some stages of human oocytes and preimplantation embryos and at the protein level by immunohistochemistry in human and bovine ovaries. NLRP7 and KHDC3L co-localize to the microtubule organizing center and/or the Golgi apparatus in human hematopoietic cells. STUDY DESIGN, SIZE, DURATION A total of 164 spare human oocytes and embryos from patients undergoing in vitro fertilization were used. PARTICIPANTS/MATERIALS, SETTING, METHODS Oocytes and early cleavage-stage embryos were fixed, immunostained with NLRP7 and/or KHDC3L antibodies, and analyzed using high-resolution confocal immunofluorescence and electron microscopies. MAIN RESULTS AND THE ROLE OF CHANCE NLRP7 and KHDC3L localize to the cytoskeleton and are predominant at the cortical region in growing oocytes. After the first cellular division, these two maternal-effect proteins become asymmetrically confined to the outer cortical region and excluded from the cell-to-cell contact region until the blastocyst stage where NLRP7 and KHDC3L homogeneously redistribute to the cytoplasm and the nucleus, respectively. LIMITATIONS, REASONS FOR CAUTION We could not analyze fresh human oocytes and embryos. The analyzed materials were donated by patients undergoing assisted reproductive technologies and released for research 1-3 days after their collection and the transfer of embryos to the patients. WIDER IMPLICATIONS OF THE FINDINGS Our study is the first comprehensive and high-resolution localization of the only two known maternal-effect proteins, NLRP7 and KHDC3L, in human oocytes and preimplantation embryos. Our data contribute to a better understanding of the roles of these two proteins in the integrity of the oocytes, post-zygotic divisions, and cell-lineage differentiation. STUDY FUNDING/COMPETING INTERESTS This work was supported by the Canadian Institute of Health Research (86546 to R.S.); E.A. was supported by fellowships from the Research Institute of the McGill University Health Centre and a CREATE award from the Réseau Québécois en Reproduction. All authors declare no conflict of interest.
Collapse
Affiliation(s)
- Elie Akoury
- Department of Human Genetics, McGill University Health Center, Montreal, QC, Canada Department of Obstetrics and Gynecology, McGill University Health Center, Montreal, QC, Canada
| | - Li Zhang
- Department of Obstetrics and Gynecology, McGill University Health Center, Montreal, QC, Canada
| | - Asangla Ao
- Department of Obstetrics and Gynecology, McGill University Health Center, Montreal, QC, Canada
| | - Rima Slim
- Department of Human Genetics, McGill University Health Center, Montreal, QC, Canada Department of Obstetrics and Gynecology, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
29
|
Van Gorp H, Kuchmiy A, Van Hauwermeiren F, Lamkanfi M. NOD-like receptors interfacing the immune and reproductive systems. FEBS J 2014; 281:4568-82. [PMID: 25154302 DOI: 10.1111/febs.13014] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/13/2014] [Accepted: 08/19/2014] [Indexed: 12/13/2022]
Abstract
Nucleotide-binding oligomerization domain receptors (NOD-like receptors, NLRs) are intracellular proteins that are chiefly known for their critical functions in inflammatory responses and host defense against microbial pathogens. Several NLRs have been demonstrated to assemble inflammasomes or to engage transcriptional signaling cascades that result in the production of pro-inflammatory cytokines and bactericidal factors. In recent years, NLRs have also emerged as key regulators of early mammalian embryogenesis and reproduction. A subset of phylogenetically related NLRs represents a new class of maternal effect genes that are highly expressed in maturing oocytes and pre-implantation embryos. Mutations in several of these NLRs have been linked to hereditary reproductive defects and imprinting diseases. In this review, we discuss the expression profiles, the emerging functions and molecular mode of action of these NLRs with newly recognized roles at the interfaces of the immune and reproductive systems. In addition, we provide an overview of coding mutations in NLRs that have been associated with human reproductive diseases, and outline crucial outstanding questions in this emerging research field.
Collapse
Affiliation(s)
- Hanne Van Gorp
- Department of Medical Protein Research, VIB, Ghent, Belgium; Department of Biochemistry, Ghent University, Ghent, Belgium
| | | | | | | |
Collapse
|