1
|
Latham KE. Paternal Effects in Mammalian Reproduction: Functional, Environmental, and Clinical Relevance of Sperm Components in Early Embryos and Beyond. Mol Reprod Dev 2025; 92:e70020. [PMID: 40123230 PMCID: PMC11931271 DOI: 10.1002/mrd.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/21/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
In addition to widely recognized contributions of the paternal genome, centriole, and oocyte-activation factors, sperm deliver a wide range of macromolecules to the fertilized embryo. The impacts of these factors on the embryo, progeny, and even subsequent generations have become increasingly apparent, along with an understanding of an extensive potential for male health and environmental exposures to exert both immediate and long-term impacts on mammalian reproduction. Available data reveal that sperm factors interact with and regulate the actions of oocyte factors as well as exerting additional direct effects on the early embryo. This review provides a summary of the nature and mechanisms of paternal effects in early mammalian embryos, long-term effects in progeny, susceptibility of sperm components to diverse environmental factors, and potential approaches to mitigate adverse effects of such exposures.
Collapse
Affiliation(s)
- Keith E. Latham
- Department of Animal ScienceMichigan State UniversityEast LansingMichiganUSA
- Department of Obstetrics, Gynecology and Reproductive BiologyMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
2
|
Naveed M, Shen Z, Bao J. Sperm-borne small non-coding RNAs: potential functions and mechanisms as epigenetic carriers. Cell Biosci 2025; 15:5. [PMID: 39825433 PMCID: PMC11740426 DOI: 10.1186/s13578-025-01347-4] [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: 08/18/2024] [Accepted: 01/10/2025] [Indexed: 01/20/2025] Open
Abstract
Over the past two decades, the study of sperm-borne small non-coding RNAs (sncRNAs) has garnered substantial growth. Once considered mere byproducts during germ cell maturation, these sncRNAs have now been recognized as crucial carriers of epigenetic information, playing a significant role in transmitting acquired traits from paternal to offspring, particularly under environmental influences. A growing body of evidence highlights the pivotal role of these sncRNAs in facilitating epigenetic inheritance across generations. However, the exact mechanisms through which these paternally supplied epigenetic carriers operate remain unclear and are under hot debate. This concise review presents the most extensive evidence to date on environmentally-responsive sperm-borne sncRNAs, encompassing brief summary of their origin, dynamics, compartmentalization, characteristics, as well as in-depth elaboration of their functional roles in epigenetic and transgenerational inheritance. Additionally, the review delves into the potential mechanisms by which sperm-delivered sncRNAs may acquire and transmit paternally acquired traits to offspring, modulating zygotic gene expression and influencing early embryonic development.
Collapse
Affiliation(s)
- Muhammad Naveed
- Center for Reproduction and Genetics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Laboratory for Physical Sciences at Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China (USTC), Hefei, Anhui, China
| | - Zhaokang Shen
- Center for Reproduction and Genetics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Laboratory for Physical Sciences at Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China (USTC), Hefei, Anhui, China
| | - Jianqiang Bao
- Center for Reproduction and Genetics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Laboratory for Physical Sciences at Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China (USTC), Hefei, Anhui, China.
| |
Collapse
|
3
|
Zhao Z, Yang T, Li F. Sperm RNA code in spermatogenesis and male infertility. Reprod Biomed Online 2024; 49:104375. [PMID: 39481211 DOI: 10.1016/j.rbmo.2024.104375] [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: 02/27/2024] [Revised: 05/22/2024] [Accepted: 07/05/2024] [Indexed: 11/02/2024]
Abstract
Spermatozoa are traditionally thought to be transcriptionally inert, but recent studies have revealed the presence of sperm RNA, some of which is derived from the residues of spermatocyte transcription and some from epididymosomes. Paternal sperm RNA can be affected by external factors and further modified at the post-transcriptional level, for example N6-methyladenosine (m6A), thus shaping spermatogenesis and reproductive outcome. This review briefly introduces the origin of sperm RNA and, on this basis, summarizes the current knowledge on RNA modifications and their functional role in spermatogenesis and male infertility. The bottlenecks and knowledge gaps in the current research on RNA modification in male reproduction have also been indicated. Further investigations are needed to elucidate the functional consequences of these modifications, providing new therapeutic and preventive strategies for reproductive health and genetic inheritance.
Collapse
Affiliation(s)
- Zhongyi Zhao
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Tingting Yang
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.
| | - Fuping Li
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.
| |
Collapse
|
4
|
Yilmaz Sukranli Z, Korkmaz Bayram K, Taheri S, Cuzin F, Ozkul Y, Rassoulzadegan M. Experimentally altering microRNA levels in embryos alters adult phenotypes. Sci Rep 2024; 14:19014. [PMID: 39152124 PMCID: PMC11329699 DOI: 10.1038/s41598-024-63692-7] [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: 06/16/2023] [Accepted: 05/31/2024] [Indexed: 08/19/2024] Open
Abstract
We previously identified a unique genetic feature of Autism Spectrum Disorder (ASD) in human patients and established mouse models, a low to very low level of six microRNAs, miR-19a-3p, miR-361-5p, miR-3613-3p, miR-150-5p, miR-126-3p and miR-499a-5p. We attempted to interfere experimentally in mice with two of them, miR19a-3p and miR499a-5p by microinjecting into zygote pronuclei either the complementary sequence or an excess of the microRNA. Both resulted in low levels in the tissues and sperm of the targeted microRNAs and their pri and pre precursors. This method stably modify predetermined levels of miRNAs and identify miRNA alterations that cause changes in autistic behavior and predispose the individual to an inherited disease. Excess miRNA results in single-stranded miRNA variations in both free and DNA-bound RNA (R-loop) fractions in mouse models thus appearing to affect their own transcription. Analysis of miRNAs fractions in human patients blood samples confirm low level of six microRNAs also in R-loop fractions.
Collapse
Affiliation(s)
- Zeynep Yilmaz Sukranli
- Betul-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Keziban Korkmaz Bayram
- Betul-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
- Department of Medical Genetics, Faculty of Medicine, Yıldırım Beyazıt University, Ankara, Turkey
| | - Serpil Taheri
- Betul-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | | | - Yusuf Ozkul
- Betul-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Minoo Rassoulzadegan
- Betul-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey.
- INSERM-CNRS, Université de Nice, Nice, France.
| |
Collapse
|
5
|
Kulikova DA, Bespalova AV, Zelentsova ES, Evgen'ev MB, Funikov SY. Epigenetic Phenomenon of Paramutation in Plants and Animals. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1429-1450. [PMID: 39245454 DOI: 10.1134/s0006297924080054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 09/10/2024]
Abstract
The phenomenon of paramutation describes the interaction between two alleles, in which one allele initiates inherited epigenetic conversion of another allele without affecting the DNA sequence. Epigenetic transformations due to paramutation are accompanied by the change in DNA and/or histone methylation patterns, affecting gene expression. Studies of paramutation in plants and animals have identified small non-coding RNAs as the main effector molecules required for the initiation of epigenetic changes in gene loci. Due to the fact that small non-coding RNAs can be transmitted across generations, the paramutation effect can be inherited and maintained in a population. In this review, we will systematically analyze examples of paramutation in different living systems described so far, highlighting common and different molecular and genetic aspects of paramutation between organisms, and considering the role of this phenomenon in evolution.
Collapse
Affiliation(s)
- Dina A Kulikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Alina V Bespalova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Elena S Zelentsova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Mikhail B Evgen'ev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Sergei Yu Funikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
6
|
Deans NC, Talbot JERB, Li M, Sáez-González C, Hövel I, Heavens D, Stam M, Hollick JB. Paramutation at the maize pl1 locus is associated with RdDM activity at distal tandem repeats. PLoS Genet 2024; 20:e1011296. [PMID: 38814980 PMCID: PMC11166354 DOI: 10.1371/journal.pgen.1011296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/11/2024] [Accepted: 05/08/2024] [Indexed: 06/01/2024] Open
Abstract
Exceptions to Mendelian inheritance often highlight novel chromosomal behaviors. The maize Pl1-Rhoades allele conferring plant pigmentation can display inheritance patterns deviating from Mendelian expectations in a behavior known as paramutation. However, the chromosome features mediating such exceptions remain unknown. Here we show that small RNA production reflecting RNA polymerase IV function within a distal downstream set of five tandem repeats is coincident with meiotically-heritable repression of the Pl1-Rhoades transcription unit. A related pl1 haplotype with three, but not one with two, repeat units also displays the trans-homolog silencing typifying paramutations. 4C interactions, CHD3a-dependent small RNA profiles, nuclease sensitivity, and polyadenylated RNA levels highlight a repeat subregion having regulatory potential. Our comparative and mutant analyses show that transcriptional repression of Pl1-Rhoades correlates with 24-nucleotide RNA production and cytosine methylation at this subregion indicating the action of a specific DNA-dependent RNA polymerase complex. These findings support a working model in which pl1 paramutation depends on trans-chromosomal RNA-directed DNA methylation operating at a discrete cis-linked and copy-number-dependent transcriptional regulatory element.
Collapse
Affiliation(s)
- Natalie C. Deans
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio, United States of America
- Centers for Applied Plant Sciences and RNA Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Joy-El R. B. Talbot
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio, United States of America
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, United States of America
| | - Mowei Li
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio, United States of America
- Centers for Applied Plant Sciences and RNA Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Cristian Sáez-González
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio, United States of America
| | - Iris Hövel
- Swammerdam Institute for Life Sciences, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | | | - Maike Stam
- Swammerdam Institute for Life Sciences, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | - Jay B. Hollick
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio, United States of America
- Centers for Applied Plant Sciences and RNA Biology, The Ohio State University, Columbus, Ohio, United States of America
- Department of Plant and Microbial Biology, University of California, Berkeley, California, United States of America
| |
Collapse
|
7
|
Pavelka J, Poláková S, Pavelková V, Galeta P. An epigenetic change in a moth is generated by temperature and transmitted to many subsequent generations mediated by RNA. PLoS One 2024; 19:e0292179. [PMID: 38451888 PMCID: PMC10919628 DOI: 10.1371/journal.pone.0292179] [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: 10/28/2022] [Accepted: 09/14/2023] [Indexed: 03/09/2024] Open
Abstract
Epigenetic changes in sexually reproducing animals may be transmitted usually only through a few generations. Here we discovered a case where epigenetic change lasts 40 generations. This epigenetic phenomenon occurs in the short antennae (sa) mutation of the flour moth (Ephestia kuehniella). We demonstrate that is probably determined by a small RNA (e.g., piRNA, miRNA, tsRNA) and transmitted in this way to subsequent generations through the male and female gametes. The observed epigenetic change cancels sa mutation and creates a wild phenotype (a moth that appears to have no mutation). It persists for many generations (40 recorded). This epigenetic transgenerational effect (suppression homozygous mutation for short antennae) in the flour moth is induced by changes during ontogenetic development, such as increased temperature on pupae development, food, different salts in food, or injection of RNA from the sperm of already affected individuals into the eggs. The epigenetic effect may occasionally disappear in some individuals and/or progeny of a pair in the generation chain in which the effect transfers. We consider that the survival of RNA over many generations has adaptive consequences. It is mainly a response to environmental change that is transmitted to offspring via RNA. In this study, we test an interesting epigenetic effect with an unexpected length after 40 generations and test what is its cause. Such transfer of RNA to subsequent generations may have a greater evolutionary significance than previously thought. Based on some analogies, we also discuss of the connection with the SIR2 gene.
Collapse
Affiliation(s)
- Jaroslav Pavelka
- University of West Bohemia, Centre of Biology, Pilsen, Czech Republic
| | - Simona Poláková
- Ministry of the Environment of the Czech Republic, Praha, Czech Republic
| | - Věra Pavelková
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Patrik Galeta
- Department of Anthropology, University of West Bohemia, Pilsen, Czech Republic
| |
Collapse
|
8
|
Wang H, Wang Z, Zhou T, Morris D, Chen S, Li M, Wang Y, Zheng H, Fu W, Yan W. Small RNA shuffling between murine sperm and their cytoplasmic droplets during epididymal maturation. Dev Cell 2023; 58:779-790.e4. [PMID: 37023748 PMCID: PMC10627525 DOI: 10.1016/j.devcel.2023.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 12/18/2022] [Accepted: 03/10/2023] [Indexed: 04/08/2023]
Abstract
Reports that mouse sperm gain small RNAs from the epididymosomes secreted by epididymal epithelial cells and that these "foreign" small RNAs act as an epigenetic information carrier mediating the transmission of acquired paternal traits have drawn great attention because the findings suggest that heritable information can flow from soma to germ line, thus invalidating the long-standing Weismann's barrier theory on heritable information flow. Using small RNA sequencing (sRNA-seq), northern blots, sRNA in situ hybridization, and immunofluorescence, we detected substantial changes in the small RNA profile in murine caput epididymal sperm (sperm in the head of the epididymis), and we further determined that the changes resulted from sperm exchanging small RNAs, mainly tsRNAs and rsRNAs, with cytoplasmic droplets rather than the epididymosomes. Moreover, the murine sperm-borne small RNAs were mainly derived from the nuclear small RNAs in late spermatids. Thus, caution is needed regarding sperm gaining foreign small RNAs as an underlying mechanism of epigenetic inheritance.
Collapse
Affiliation(s)
- Hetan Wang
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, CA 90502, USA; Department of Medical Genetics, China Medical University, Shenyang 110122, China
| | - Zhuqing Wang
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, CA 90502, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Dayton Morris
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, CA 90502, USA
| | - Sheng Chen
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, CA 90502, USA
| | - Musheng Li
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Yue Wang
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, CA 90502, USA
| | - Huili Zheng
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, CA 90502, USA
| | - Weineng Fu
- Department of Medical Genetics, China Medical University, Shenyang 110122, China
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, CA 90502, USA; Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
9
|
Cabej NR. On the origin and nature of nongenetic information in eumetazoans. Ann N Y Acad Sci 2023. [PMID: 37154677 DOI: 10.1111/nyas.15001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Nongenetic information implies all the forms of biological information not related to genes and DNA in general. Despite the deep scientific relevance of the concept, we currently lack reliable knowledge about its carriers and origins; hence, we still do not understand its true nature. Given that genes are the targets of nongenetic information, it appears that a parsimonious approach to find the ultimate source of that information is to trace back the sequential steps of the causal chain upstream of the target genes up to the ultimate link as the source of the nongenetic information. From this perspective, I examine seven nongenetically determined phenomena: placement of locus-specific epigenetic marks on DNA and histones, changes in snRNA expression patterns, neural induction of gene expression, site-specific alternative gene splicing, predator-induced morphological changes, and cultural inheritance. Based on the available evidence, I propose a general model of the common neural origin of all these forms of nongenetic information in eumetazoans.
Collapse
Affiliation(s)
- Nelson R Cabej
- Department of Biology, University of Tirana, Tirana, Albania
| |
Collapse
|
10
|
Mo J, Wan MT, Au DWT, Shi J, Tam N, Qin X, Cheung NKM, Lai KP, Winkler C, Kong RYC, Seemann F. Transgenerational bone toxicity in F3 medaka (Oryzias latipes) induced by ancestral benzo[a]pyrene exposure: Cellular and transcriptomic insights. J Environ Sci (China) 2023; 127:336-348. [PMID: 36522066 DOI: 10.1016/j.jes.2022.04.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 06/17/2023]
Abstract
Benzo[a]pyrene (BaP), a ubiquitous pollutant, raises environmental health concerns due to induction of bone toxicity in the unexposed offspring. Exposure of F0 ancestor medaka (Oryzias latipes) to 1 µg/L BaP for 21 days causes reduced vertebral bone thickness in the unexposed F3 male offspring. To reveal the inherited modifications, osteoblast (OB) abundance and molecular signaling pathways of transgenerational BaP-induced bone thinning were assessed. Histomorphometric analysis showed a reduction in OB abundance. Analyses of the miRNA and mRNA transcriptomes revealed the dysregulation of Wnt signaling (frzb/ola-miR-1-3p, sfrp5/ola-miR-96-5p/miR-455-5p) and bone morphogenetic protein (Bmp) signaling (bmp3/ola-miR-96-5p/miR-181b-5p/miR-199a-5p/miR-205-5p/miR-455-5p). Both pathways are major indicators of impaired bone formation, while the altered Rank signaling in osteoclasts (c-fos/miR-205-5p) suggests a potentially augmented bone resorption. Interestingly, a typical BaP-responsive pathway, the Nrf2-mediated oxidative stress response (gst/ola-miR-181b-5p/miR-199a-5p/miR-205), was also affected. Moreover, mRNA levels of epigenetic modification enzymes (e.g., hdac6, hdac7, kdm5b) were found dysregulated. The findings indicated that epigenetic factors (e.g., miRNAs, histone modifications) may directly regulate the expression of genes associated with transgenerational BaP bone toxicity and warrants further studies. The identified candidate genes and miRNAs may serve as potential biomarkers for BaP-induced bone disease and as indicators of historic exposures in wild fish for conservation purposes.
Collapse
Affiliation(s)
- Jiezhang Mo
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510000, China; Shenzhen Key Laboratory for the Sustainable Use of Marine Biodiversity, Research Centre for the Oceans and Human Health, Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China; State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Miles Teng Wan
- State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Doris Wai-Ting Au
- Shenzhen Key Laboratory for the Sustainable Use of Marine Biodiversity, Research Centre for the Oceans and Human Health, Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China; State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Jingchun Shi
- State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Nathan Tam
- State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Xian Qin
- State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Napo K M Cheung
- State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Keng Po Lai
- State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China; Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin 541004, China
| | - Christoph Winkler
- Department of Biological Sciences, National University of Singapore, 119077, Singapore
| | - Richard Yuen-Chong Kong
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510000, China; Shenzhen Key Laboratory for the Sustainable Use of Marine Biodiversity, Research Centre for the Oceans and Human Health, Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China; State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China.
| | - Frauke Seemann
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 510000, China; Center for Coastal Studies and Department of Life Sciences, Texas A&M University-Corpus Christi, Corpus Christi, Texas 78412, USA.
| |
Collapse
|
11
|
Chen Q. Sperm RNA-mediated epigenetic inheritance in mammals: challenges and opportunities. Reprod Fertil Dev 2022; 35:118-124. [PMID: 36592983 PMCID: PMC9827497 DOI: 10.1071/rd22218] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Emerging evidence now shows that in addition to delivering a haploid DNA, the mammalian sperm also carry various types of RNAs that respond to the paternal environment, which can mediate the intergenerational transmission of certain phenotypes to the offspring relating to the paternal environmental exposures (e.g. diet, mental stress). Improved analytical tools are beginning to decipher the complexity of sperm RNAs, RNA modifications and their spatial compartmentalisation, which support the concept of 'sperm RNA code' in programming specific offspring phenotypes during embryonic development. In this commentary article, I discuss the challenges and opportunities in solidifying the field of mammalian sperm RNA-mediated epigenetic inheritance, including the identification of the key sperm RNAs that are responsible for the paternal phenotype transmission, and the cellular and molecular events that are triggered by sperm RNAs during embryo development. I also discuss the translational application potential by harnessing the knowledge of sperm RNA code to improve farm animal production and human health.
Collapse
Affiliation(s)
- Qi Chen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA.,Correspondence to: Qi Chen, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA,
| |
Collapse
|
12
|
Lee GS, Conine CC. The Transmission of Intergenerational Epigenetic Information by Sperm microRNAs. EPIGENOMES 2022; 6:12. [PMID: 35466187 PMCID: PMC9036291 DOI: 10.3390/epigenomes6020012] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/19/2022] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Epigenetic information is transmitted from one generation to the next, modulating the phenotype of offspring non-genetically in organisms ranging from plants to mammals. For intergenerational non-genetic inheritance to occur, epigenetic information must accumulate in germ cells. The three main carriers of epigenetic information-histone post-translational modifications, DNA modifications, and RNAs-all exhibit dynamic patterns of regulation during germ cell development. For example, histone modifications and DNA methylation are extensively reprogrammed and often eliminated during germ cell maturation and after fertilization during embryogenesis. Consequently, much attention has been given to RNAs, specifically small regulatory RNAs, as carriers of inherited epigenetic information. In this review, we discuss examples in which microRNAs have been implicated as key players in transmitting paternal epigenetic information intergenerationally.
Collapse
Affiliation(s)
- Grace S. Lee
- Pharmacology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA;
| | - Colin C. Conine
- Departments of Genetics and Pediatrics—Penn Epigenetics Institute, Institute of Regenerative Medicine, and Center for Reproduction and Women’s Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Small Noncoding RNAs in Reproduction and Infertility. Biomedicines 2021; 9:biomedicines9121884. [PMID: 34944700 PMCID: PMC8698561 DOI: 10.3390/biomedicines9121884] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/20/2022] Open
Abstract
Infertility has been reported as one of the most common reproductive impairments, affecting nearly one in six couples worldwide. A large proportion of infertility cases are diagnosed as idiopathic, signifying a deficit in information surrounding the pathology of infertility and necessity of medical intervention such as assisted reproductive therapy. Small noncoding RNAs (sncRNAs) are well-established regulators of mammalian reproduction. Advanced technologies have revealed the dynamic expression and diverse functions of sncRNAs during mammalian germ cell development. Mounting evidence indicates sncRNAs in sperm, especially microRNAs (miRNAs) and transfer RNA (tRNA)-derived small RNAs (tsRNAs), are sensitive to environmental changes and mediate the inheritance of paternally acquired metabolic and mental traits. Here, we review the critical roles of sncRNAs in mammalian germ cell development. Furthermore, we highlight the functions of sperm-borne sncRNAs in epigenetic inheritance. We also discuss evidence supporting sncRNAs as promising biomarkers for fertility and embryo quality in addition to the present limitations of using sncRNAs for infertility diagnosis and treatment.
Collapse
|
14
|
Liang K, Yao L, Wang S, Zheng L, Qian Z, Ge Y, Chen L, Cheng X, Ma R, Li C, Jing J, Yang Y, Yu W, Xue T, Chen Q, Cao S, Ma J, Yao B. miR-125a-5p increases cellular DNA damage of aging males and perturbs stage-specific embryo development via Rbm38-p53 signaling. Aging Cell 2021; 20:e13508. [PMID: 34751998 PMCID: PMC8672779 DOI: 10.1111/acel.13508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/26/2021] [Accepted: 10/09/2021] [Indexed: 11/29/2022] Open
Abstract
An increasing number of men are fathering children at an older age than in the past. While advanced maternal age has long been recognized as a risk factor for adverse reproductive outcomes, the influence of paternal age on reproduction is incompletely comprehended. Herein, we found that miR‐125a‐5p was upregulated in the sperm of aging males and was related to inferior sperm DNA integrity as an adverse predictor. Moreover, we demonstrated that miR‐125a‐5p suppressed mitochondrial function and increased cellular DNA damage in GC2 cells. We also found that miR‐125a‐5p perturbed embryo development at specific morula/blastocyst stages. Mechanistically, we confirmed that miR‐125a‐5p disturbed the mitochondrial function by targeting Rbm38 and activating the p53 damage response pathway, and induced a developmental delay in a p21‐dependent manner. Our study revealed an important role of miR‐125a‐5p in sperm function and early embryo development of aging males, and provided a fresh view to comprehend the aging process in sperm.
Collapse
Affiliation(s)
- Kuan Liang
- Center of Reproductive Medicine Nanjing Jinling Hospital The First School of Clinical Medicine Southern Medical University Nanjing China
| | - Liangyu Yao
- Department of Urology The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Shuxian Wang
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Lu Zheng
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Zhang Qian
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Yifeng Ge
- Center of Reproductive Medicine Nanjing Jinling Hospital The First School of Clinical Medicine Southern Medical University Nanjing China
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Li Chen
- Center of Reproductive Medicine Nanjing Jinling Hospital The First School of Clinical Medicine Southern Medical University Nanjing China
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Xi Cheng
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Rujun Ma
- Center of Reproductive Medicine Nanjing Jinling Hospital The First School of Clinical Medicine Southern Medical University Nanjing China
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Chuwei Li
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Jun Jing
- Center of Reproductive Medicine Nanjing Jinling Hospital The First School of Clinical Medicine Southern Medical University Nanjing China
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Yang Yang
- Basic Medical Laboratory Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Wanwan Yu
- Department of Emergency medicine Jinling Hospital, Medical School of Nanjing University Nanjing China
| | - Tongmin Xue
- Department Reproductive Medical Center Jinling Hospital Nanjing Medicine University Nanjing China
| | - Qiwei Chen
- Center of Reproductive Medicine Nanjing Jinling Hospital The First School of Clinical Medicine Southern Medical University Nanjing China
| | - Siyuan Cao
- School of Life Science Nanjing Normal University Nanjing China
| | - Jinzhao Ma
- Center of Reproductive Medicine Nanjing Jinling Hospital The First School of Clinical Medicine Southern Medical University Nanjing China
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
| | - Bing Yao
- Center of Reproductive Medicine Nanjing Jinling Hospital The First School of Clinical Medicine Southern Medical University Nanjing China
- Center of Reproductive Medicine Nanjing Jinling Hospital Clinical School of Medical College Nanjing University Nanjing China
- Department Reproductive Medical Center Jinling Hospital Nanjing Medicine University Nanjing China
- School of Life Science Nanjing Normal University Nanjing China
| |
Collapse
|
15
|
Paternal environmental exposure-induced spermatozoal small noncoding RNA alteration meditates the intergenerational epigenetic inheritance of multiple diseases. Front Med 2021; 16:176-184. [PMID: 34515940 DOI: 10.1007/s11684-021-0885-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/30/2021] [Indexed: 10/20/2022]
Abstract
Studies of human and mammalian have revealed that environmental exposure can affect paternal health conditions as well as those of the offspring. However, studies that explore the mechanisms that meditate this transmission are rare. Recently, small noncoding RNAs (sncRNAs) in sperm have seemed crucial to this transmission due to their alteration in sperm in response to environmental exposure, and the methodology of microinjection of isolated total RNA or sncRNAs or synthetically identified sncRNAs gradually lifted the veil of sncRNA regulation during intergenerational inheritance along the male line. Hence, by reviewing relevant literature, this study intends to answer the following research concepts: (1) paternal environmental factors that can be passed on to offspring and are attributed to spermatozoal sncRNAs, (2) potential role of paternal spermatozoal sncRNAs during the intergenerational inheritance process, and (3) the potential mechanism by which spermatozoal sncRNAs meditate intergenerational inheritance. In summary, increased attention highlights the hidden wonder of spermatozoal sncRNAs during intergenerational inheritance. Therefore, in the future, more studies should focus on the origin of RNA alteration, the target of RNA regulation, and how sncRNA regulation during embryonic development can be sustained even in adult offspring.
Collapse
|
16
|
He T, Guo H, Shen X, Wu X, Xia L, Jiang X, Xu Y, Chen D, Zhang Y, Tan D, Tan Y. Hypoxia-induced alteration of RNA modifications in the mouse testis and sperm. Biol Reprod 2021; 105:1171-1178. [PMID: 34296257 DOI: 10.1093/biolre/ioab142] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/26/2021] [Accepted: 07/15/2021] [Indexed: 12/30/2022] Open
Abstract
Hypobaric hypoxia as an extreme environment in a plateau may have deleterious effects on human health. Studies have indicated that rush entry into a plateau may reduce male fertility and manifest in decreased sperm counts and weakened sperm motility. RNA modifications are sensitive to environmental changes and have recently emerged as novel post-transcriptional regulators in male spermatogenesis and intergenerational epigenetic inheritance. In the present study, we generated a mouse hypoxia model simulating the environment of 5500 meters in altitude for 35 days, which led to compromised spermatogenesis, decreased sperm counts, and an increased sperm deformation rate. Using this hypoxia model, we further applied our recently developed high-throughput RNA modification quantification platform based on LC-MS/MS, which exhibited the capacity to simultaneously examine 25 types of RNA modifications. Our results revealed an altered sperm RNA modifications signature in the testis (6 types) and mature sperm (11 types) under the hypoxia model, with 4 types showing overlap (Am, Gm, m7G, and m22G). Our data first drew the signature of RNA modification profiles and comprehensively analyzed the alteration of RNA modification levels in mouse testis and sperm under a mouse hypoxia model. These data may be highly related to human conditions under a similar hypoxia environment.
Collapse
Affiliation(s)
- Tong He
- Laboratory Animal Center, Chongqing Medical University, Yixueyuan Road 1, Yuzhong District, Chongqing, 400016, P. R. China.,Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Huanping Guo
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Xipeng Shen
- Laboratory Animal Center, Chongqing Medical University, Yixueyuan Road 1, Yuzhong District, Chongqing, 400016, P. R. China.,Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Xiao Wu
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Lin Xia
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Xuelin Jiang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Yinying Xu
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Dan Chen
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Yunfang Zhang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Dongmei Tan
- Laboratory Animal Center, Chongqing Medical University, Yixueyuan Road 1, Yuzhong District, Chongqing, 400016, P. R. China
| | - Yi Tan
- Laboratory Animal Center, Chongqing Medical University, Yixueyuan Road 1, Yuzhong District, Chongqing, 400016, P. R. China
| |
Collapse
|
17
|
Cabej NR. A mechanism of inheritance of acquired traits in animals. Dev Biol 2021; 475:106-117. [PMID: 33741349 DOI: 10.1016/j.ydbio.2021.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 01/11/2023]
Abstract
Observational and experimental evidence for the inheritance of acquired traits in animals is slowly, but steadily accumulating. The onset and transmission of acquired traits implies the acquisition and transmission from parents to progeny of new information, which is different from the genetic information contained in DNA. The new non-genetic information most commonly is passed on from parents to the offspring via gamete(s), but how it is precisely transmitted to the successive generations is still unknown. Based on adequate empirical evidence presented herein, a hypothesis is proposed of the inheritance of acquired traits in animals and the flow of the relevant parental information to the offspring.
Collapse
Affiliation(s)
- Nelson R Cabej
- University of Tirana Faculty of Medicine, Universiteti i Mjekesise Tirane, Department of Biology, 147 Manhattan Terrace, Dumont, 07628, USA.
| |
Collapse
|
18
|
Zhu L, Marjani SL, Jiang Z. The Epigenetics of Gametes and Early Embryos and Potential Long-Range Consequences in Livestock Species-Filling in the Picture With Epigenomic Analyses. Front Genet 2021; 12:557934. [PMID: 33747031 PMCID: PMC7966815 DOI: 10.3389/fgene.2021.557934] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 02/04/2021] [Indexed: 12/31/2022] Open
Abstract
The epigenome is dynamic and forged by epigenetic mechanisms, such as DNA methylation, histone modifications, chromatin remodeling, and non-coding RNA species. Increasing lines of evidence support the concept that certain acquired traits are derived from environmental exposure during early embryonic and fetal development, i.e., fetal programming, and can even be "memorized" in the germline as epigenetic information and transmitted to future generations. Advances in technology are now driving the global profiling and precise editing of germline and embryonic epigenomes, thereby improving our understanding of epigenetic regulation and inheritance. These achievements open new avenues for the development of technologies or potential management interventions to counteract adverse conditions or improve performance in livestock species. In this article, we review the epigenetic analyses (DNA methylation, histone modification, chromatin remodeling, and non-coding RNAs) of germ cells and embryos in mammalian livestock species (cattle, sheep, goats, and pigs) and the epigenetic determinants of gamete and embryo viability. We also discuss the effects of parental environmental exposures on the epigenetics of gametes and the early embryo, and evidence for transgenerational inheritance in livestock.
Collapse
Affiliation(s)
- Linkai Zhu
- AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Sadie L. Marjani
- Department of Biology, Central Connecticut State University, New Britain, CT, United States
| | - Zongliang Jiang
- AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
19
|
Epigenetic Mechanisms of Paternal Stress in Offspring Development and Diseases. Int J Genomics 2021; 2021:6632719. [PMID: 33532485 PMCID: PMC7837765 DOI: 10.1155/2021/6632719] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/28/2020] [Accepted: 01/08/2021] [Indexed: 01/10/2023] Open
Abstract
The major biological function of the sperm cell is to transmit the paternal genetic and epigenetic information to the embryo as well as the following offspring. Sperm has a unique epigenome. An increasing body of epidemiological study supports that paternal stress induced by environmental exposures and lifestyle can modulate the sperm epigenome (including histone modification, DNA methylation, and noncoding RNA expression), sperm-egg fusion, embryo development, and offspring health. Based on the existing literature, we have summarized the paternal exposure on sperm epigenome along with the representative phenotypes of offspring and the possible mechanism involved.
Collapse
|
20
|
Le Blévec E, Muroňová J, Ray PF, Arnoult C. Paternal epigenetics: Mammalian sperm provide much more than DNA at fertilization. Mol Cell Endocrinol 2020; 518:110964. [PMID: 32738444 DOI: 10.1016/j.mce.2020.110964] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022]
Abstract
The spermatozoon is a highly differentiated cell with unique characteristics: it is mobile, thanks to its flagellum, and is very compact. The sperm cytoplasm is extremely reduced, containing no ribosomes, and therefore does not allow translation, and its nucleus contains very closed chromatin, preventing transcription. This DNA compaction is linked to the loss of nucleosomes and the replacement of histones by protamines. Based on these characteristics, sperm was considered to simply deliver paternal DNA to the oocyte. However, some parts of the sperm DNA remain organized in a nucleosomal format, and bear epigenetic information. In addition, the nucleus and the cytoplasm contain a multitude of RNAs of different types, including non-coding RNAs (ncRNAs) which also carry epigenetic information. For a long time, these RNAs were considered residues of spermatogenesis. After briefly describing the mechanisms of compaction of sperm DNA, we focus this review on the origin and function of the different ncRNAs. We present studies demonstrating the importance of these RNAs in embryonic development and transgenerational adaptation to stress. We also look at other epigenetic marks, such as DNA methylation or post-translational modifications of histones, and show that they are sensitive to environmental stress and transmissible to offspring. The post-fertilization role of certain sperm-borne proteins is also discussed.
Collapse
Affiliation(s)
- Emilie Le Blévec
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France; IMV Technologies, ZI N° 1 Est, L'Aigle, F-61300, France
| | - Jana Muroňová
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France
| | - Pierre F Ray
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France; CHU de Grenoble, UM GI-DPI, Grenoble, F-38000, France
| | - Christophe Arnoult
- Université Grenoble Alpes, Grenoble, F-38000, France; Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble, F-38000, France.
| |
Collapse
|
21
|
Galan C, Krykbaeva M, Rando OJ. Early life lessons: The lasting effects of germline epigenetic information on organismal development. Mol Metab 2020; 38:100924. [PMID: 31974037 PMCID: PMC7300385 DOI: 10.1016/j.molmet.2019.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND An organism's metabolic phenotype is primarily affected by its genotype, its lifestyle, and the nutritional composition of its food supply. In addition, it is now clear from studies in many different species that ancestral environments can also modulate metabolism in at least one to two generations of offspring. SCOPE OF REVIEW We limit ourselves here to paternal effects in mammals, primarily focusing on studies performed in inbred rodent models. Although hundreds of studies link paternal diets and offspring metabolism, the mechanistic basis by which epigenetic information in sperm programs nutrient handling in the next generation remains mysterious. Our goal in this review is to provide a brief overview of paternal effect paradigms and the germline epigenome. We then pivot to exploring one key mystery in this literature: how do epigenetic changes in sperm, most of which are likely to act transiently in the early embryo, ultimately direct a long-lasting physiological response in offspring? MAJOR CONCLUSIONS Several potential mechanisms exist by which transient epigenetic modifications, such as small RNAs or methylation states erased shortly after fertilization, could be transferred to more durable heritable information. A detailed mechanistic understanding of this process will provide deep insights into early development, and could be of great relevance for human health and disease.
Collapse
Affiliation(s)
- Carolina Galan
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Marina Krykbaeva
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Oliver J Rando
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
22
|
Genetic control of non-genetic inheritance in mammals: state-of-the-art and perspectives. Mamm Genome 2020; 31:146-156. [PMID: 32529318 PMCID: PMC7369129 DOI: 10.1007/s00335-020-09841-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022]
Abstract
Thought to be directly and uniquely dependent from genotypes, the ontogeny of individual phenotypes is much more complicated. Individual genetics, environmental exposures, and their interaction are the three main determinants of individual's phenotype. This picture has been further complicated a decade ago when the Lamarckian theory of acquired inheritance has been rekindled with the discovery of epigenetic inheritance, according to which acquired phenotypes can be transmitted through fertilization and affect phenotypes across generations. The results of Genome-Wide Association Studies have also highlighted a big degree of missing heritability in genetics and have provided hints that not only acquired phenotypes, but also individual's genotypes affect phenotypes intergenerationally through indirect genetic effects. Here, we review available examples of indirect genetic effects in mammals, what is known of the underlying molecular mechanisms and their potential impact for our understanding of missing heritability, phenotypic variation. and individual disease risk.
Collapse
|
23
|
Ma J, Chen Q, Wang S, Ma R, Jing J, Yang Y, Feng Y, Zou Z, Zhang Y, Ge X, Xue T, Liang K, Cao S, Wang D, Chen L, Yao B. Mitochondria-related miR-574 reduces sperm ATP by targeting ND5 in aging males. Aging (Albany NY) 2020; 12:8321-8338. [PMID: 32381753 PMCID: PMC7244036 DOI: 10.18632/aging.103141] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/31/2020] [Indexed: 12/28/2022]
Abstract
Couples are delaying childbearing in recent decades. While women experience a notable decrease in oocyte production in their late thirties, the effect of advanced paternal age on reproduction is incompletely understood. Herein, we observed that numerous miRNAs, including miR-574, increased in the sperm of aging males, as indicated by high-throughput sequencing. We demonstrated that miR-574 was upregulated in the sperm of two aging mouse models and was related to inferior sperm motility as an adverse predictor. Moreover, we proved that miR-574 suppressed mitochondrial function and reduced cellular ATP production in GC2 cells. Mechanistically, we demonstrated that miR-574 regulated mitochondrial function by directly targeting mt-ND5. Our study revealed an important role of miR-574 in sperm function in aging males and provided a fresh view to comprehend the aging process in sperm.
Collapse
Affiliation(s)
- Jinzhao Ma
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China.,Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Qiwei Chen
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China
| | - Shuxian Wang
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Rujun Ma
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China.,Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Jun Jing
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China.,Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Yang Yang
- Institute of Laboratory Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Yuming Feng
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China.,Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Zhichuan Zou
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China.,Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Yu Zhang
- Center of Reproductive Medicine, Nanjing Jinling Hospital, School of Medicine, Jiangsu University, Zhenjiang 212002, Jiangsu, China
| | - Xie Ge
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China.,Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Tongmin Xue
- Jinling Hospital Department Reproductive Medical Center, Nanjing Medicine University, Nanjing 210002, Jiangsu, China
| | - Kuan Liang
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China
| | - Siyuan Cao
- School of Life Science, Nanjing Normal University, Nanjing 210002, Jiangsu, China
| | - Dandan Wang
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Li Chen
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China.,Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Bing Yao
- Center of Reproductive Medicine, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, Jiangsu, China.,Center of Reproductive Medicine, Nanjing Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing 210002, Jiangsu, China.,Center of Reproductive Medicine, Nanjing Jinling Hospital, School of Medicine, Jiangsu University, Zhenjiang 212002, Jiangsu, China.,Jinling Hospital Department Reproductive Medical Center, Nanjing Medicine University, Nanjing 210002, Jiangsu, China.,School of Life Science, Nanjing Normal University, Nanjing 210002, Jiangsu, China
| |
Collapse
|
24
|
Sperm RNA: Quo vadis? Semin Cell Dev Biol 2020; 97:123-130. [DOI: 10.1016/j.semcdb.2019.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 06/26/2019] [Accepted: 07/08/2019] [Indexed: 12/27/2022]
|
25
|
Liberman N, Wang SY, Greer EL. Transgenerational epigenetic inheritance: from phenomena to molecular mechanisms. Curr Opin Neurobiol 2019; 59:189-206. [PMID: 31634674 DOI: 10.1016/j.conb.2019.09.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023]
Abstract
Inherited information not encoded in the DNA sequence can regulate a variety of complex phenotypes. However, how this epigenetic information escapes the typical epigenetic erasure that occurs upon fertilization and how it regulates behavior is still unclear. Here we review recent examples of brain related transgenerational epigenetic inheritance and delineate potential molecular mechanisms that could regulate how non-genetic information could be transmitted.
Collapse
Affiliation(s)
- Noa Liberman
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston MA 02115, USA
| | - Simon Yuan Wang
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston MA 02115, USA
| | - Eric Lieberman Greer
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston MA 02115, USA.
| |
Collapse
|
26
|
Wang Q, Zhang Y, Le F, Wang N, Zhang F, Luo Y, Lou Y, Hu M, Wang L, Thurston LM, Xu X, Jin F. Alteration in the expression of the renin-angiotensin system in the myocardium of mice conceived by in vitro fertilization. Biol Reprod 2019; 99:1276-1288. [PMID: 30010728 PMCID: PMC6299247 DOI: 10.1093/biolre/ioy158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/11/2018] [Indexed: 12/25/2022] Open
Abstract
Epidemiological studies have revealed that offspring conceived by in vitro fertilization (IVF) have an elevated risk of cardiovascular malformations at birth, and are more predisposed to cardiovascular diseases. The renin-angiotensin system (RAS) plays an essential role in both the pathogenesis of congenital heart disease in fetuses and cardiovascular dysfunction in adults. This study aimed to assess the relative expression levels of genes in the RAS pathway in mice conceived using IVF, compared to natural mating with superovulation. Results demonstrated that expression of the angiotensin II receptor type 1 (AGTR1), connective tissue growth factor (CTGF), and collagen 3 (COL3), in the myocardial tissue of IVF-conceived mice, was elevated at 3 weeks, 10 weeks, and 1.5 years of age, when compared to their non-IVF counterparts. These data were supported by microRNA microarray analysis of the myocardial tissue of aged IVF-conceived mice, where miR-100, miR-297, and miR-758, which interact with COL3, AGTR1, and COL1 respectively, were upregulated when compared to naturally mated mice of the same age. Interestingly, bisulfite sequencing data indicated that IVF-conceived mice exhibited decreased methylation of CpG sites in Col1. In support of our in vivo investigations, miR-297 overexpression was shown to upregulate AGTR1 and CTGF, and increased cell proliferation in cultured H9c2 cardiomyocytes. These findings indicate that the altered expression of RAS in myocardial tissue might contribute to cardiovascular malformation and/or dysfunction in IVF-conceived offspring. Furthermore, these cardiovascular abnormalities might be the result of altered DNA methylation and abnormal regulation of microRNAs.
Collapse
Affiliation(s)
- Qijing Wang
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yue Zhang
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Fang Le
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ning Wang
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Fan Zhang
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yuqin Luo
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yiyun Lou
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang Province, China
| | - Minhao Hu
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Liya Wang
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Lisa M Thurston
- Department of Comparative Biomedical Science, Royal Veterinary College, University of London, London NW1 0TU, UK.,Academic Unit of Reproduction and Development, Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2SF, UK
| | - Xiangrong Xu
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Fan Jin
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics of National Ministry of Education, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
27
|
Sharma U. Paternal Contributions to Offspring Health: Role of Sperm Small RNAs in Intergenerational Transmission of Epigenetic Information. Front Cell Dev Biol 2019; 7:215. [PMID: 31681757 PMCID: PMC6803970 DOI: 10.3389/fcell.2019.00215] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 09/18/2019] [Indexed: 12/21/2022] Open
Abstract
The most fundamental process for the perpetuation of a species is the transfer of information from parent to offspring. Although genomic DNA contributes to the majority of the inheritance, it is now clear that epigenetic information −information beyond the underlying DNA sequence − is also passed on to future generations. However, the mechanism and extent of such inheritance are not well-understood. Here, I review some of the concepts, evidence, and mechanisms of intergenerational epigenetic inheritance via sperm small RNAs. Recent studies provide evidence that mature sperm are highly abundant in small non-coding RNAs. These RNAs are modulated by paternal environmental conditions and potentially delivered to the zygote at fertilization, where they can regulate early embryonic development. Intriguingly, sperm small RNA payload undergoes dramatic changes during testicular and post-testicular maturation, making the mature sperm epigenome highly unique and distinct from testicular germ cells. I explore the mechanism of sperm small RNA remodeling during post-testicular maturation in the epididymis, and the potential role of this reprograming in intergenerational epigenetic inheritance.
Collapse
Affiliation(s)
- Upasna Sharma
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
28
|
Kianmehr L, Khazali H, Rajabi-Maham H, Sharifi-Zarchi A, Cuzin F, Rassoulzadegan M. Genome-Wide Distribution of Nascent Transcripts in Sperm DNA, Products of a Late Wave of General Transcription. Cells 2019; 8:cells8101196. [PMID: 31623338 PMCID: PMC6830337 DOI: 10.3390/cells8101196] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 10/02/2019] [Indexed: 12/22/2022] Open
Abstract
Mature spermatozoa contain a whole repertoire of the various classes of cellular RNAs, both coding and non-coding. It was hypothesized that after fertilization they might impact development, a claim supported by experimental evidence in various systems. Despite the current increasing interest in the transgenerational maintenance of epigenetic traits and their possible determination by RNAs, little remains known about conservation in sperm and across generations and the specificities and mechanisms involved in transgenerational maintenance. We identified two distinct fractions of RNAs in mature mouse sperm, one readily extracted in the aqueous phase of the classical TRIzol procedure and a distinct fraction hybridized with homologous DNA in DNA-RNA complexes recovered from the interface, purified after DNase hydrolysis and analyzed by RNA-seq methodology. This DNA-associated RNA (D RNA) was found to represent as much as half of the cell contents in differentiated sperm, in which a major part of the cytoplasmic material has been discarded. Stable complexes were purified free of proteins and identified as hybrids (R-loops) on the basis of their sensitivity to RNase H hydrolysis. Further analysis by RNA-seq identified transcripts from all the coding and non-coding regions of the genome, thus revealing an extensive wave of transcription, prior to or concomitant with the terminal compaction of the chromatin.
Collapse
Affiliation(s)
- Leila Kianmehr
- Animal Sciences and Biotechnology Department, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, G.C, Tehran 1983963113, Iran.
- Université de Nice-Sophia Antipolis, Faculté des Sciences, Parc Valrose, INSERM-CNRS, 06108 Nice CEDEX 2, France.
| | - Homayoun Khazali
- Animal Sciences and Biotechnology Department, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, G.C, Tehran 1983963113, Iran.
| | - Hassan Rajabi-Maham
- Animal Sciences and Biotechnology Department, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, G.C, Tehran 1983963113, Iran.
| | - Ali Sharifi-Zarchi
- Computer Engineering Department, Sharif University of Technology, Tehran 1458889694, Iran.
| | - François Cuzin
- Université de Nice-Sophia Antipolis, Faculté des Sciences, Parc Valrose, INSERM-CNRS, 06108 Nice CEDEX 2, France.
| | - Minoo Rassoulzadegan
- Université de Nice-Sophia Antipolis, Faculté des Sciences, Parc Valrose, INSERM-CNRS, 06108 Nice CEDEX 2, France.
| |
Collapse
|
29
|
Broecker F, Moelling K. What viruses tell us about evolution and immunity: beyond Darwin? Ann N Y Acad Sci 2019; 1447:53-68. [PMID: 31032941 PMCID: PMC6850104 DOI: 10.1111/nyas.14097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/09/2019] [Accepted: 03/20/2019] [Indexed: 12/14/2022]
Abstract
We describe mechanisms of genetic innovation mediated by viruses and related elements that, during evolution, caused major genetic changes beyond what was anticipated by Charles Darwin. Viruses and related elements introduced genetic information and have shaped the genomes and immune systems of all cellular life forms. None of these mechanisms contradict Darwin's theory of evolution but extend it by means of sequence information that has recently become available. Not only do small increments of genetic information contribute to evolution, but also do major events such as infection by viruses or bacteria, which can supply new genetic information to a host by horizontal gene transfer. Thereby, viruses and virus-like elements act as major drivers of evolution.
Collapse
Affiliation(s)
- Felix Broecker
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Karin Moelling
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland.,Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
30
|
Dupont C, Kappeler L, Saget S, Grandjean V, Lévy R. Role of miRNA in the Transmission of Metabolic Diseases Associated With Paternal Diet-Induced Obesity. Front Genet 2019; 10:337. [PMID: 31057600 PMCID: PMC6482346 DOI: 10.3389/fgene.2019.00337] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/29/2019] [Indexed: 12/25/2022] Open
Abstract
The concept of Developmental Origins of Health and Diseases (DOHaD) recognizes that an unfavorable maternal environment alters the developmental trajectory of the fetus and can lead to long-term risk of developing chronic noncommunicable diseases. More recently, the concept of a paternal transmission [Paternal Origins of Health and Diseases (POHaD)] has emerged stressing the impact of paternal overweight or obesity on offspring's health and development. While very few examples of paternal epigenetic inheritance of metabolic disorders have been evidenced in human, many experimental mouse models based on high-fat diet (HFD)-induced paternal obesity have been developed to breakdown molecular mechanisms involved in the process. Besides DNA methylation and chromatin structure, sperm short noncoding RNAs have been considered as the main epigenetic vector of inheritance of paternally environmentally induced changes. Among them, sperm miRNAs are one particular subspecies sensitive to environmental changes and obesity can modify the sperm miRNA profile. Once delivered into the zygote, these molecules might induce epigenetic modifications in the embryo, thereby leading to consequences for fetus development and offspring physical and metabolic health later on in life. Furthermore, some data also suggest that metabolic pathologies may be intergenerationally or transgenerationally transmitted.
Collapse
Affiliation(s)
- Charlotte Dupont
- Sorbonne Université, Inserm, Centre de Recherche St-Antoine, CRSA, AP-HP, Hôpital Tenon, Service de biologie de la reproduction-CECOS, Paris, France
| | - Laurent Kappeler
- Sorbonne Université, INSERM, Institute of Cardiometabolism, Centre de Recherche St-Antoine, CRSA, Paris, France
| | - Sarah Saget
- Sorbonne Université, INSERM, Institute of Cardiometabolism, Centre de Recherche St-Antoine, CRSA, Paris, France
| | - Valérie Grandjean
- Inserm U1065, Team Control of Gene Expression (10), Université Cote d’Azur, Nice, France
| | - Rachel Lévy
- Sorbonne Université, Inserm, Centre de Recherche St-Antoine, CRSA, AP-HP, Hôpital Tenon, Service de biologie de la reproduction-CECOS, Paris, France
| |
Collapse
|
31
|
Godschalk R, Remels A, Hoogendoorn C, van Benthem J, Luijten M, Duale N, Brunborg G, Olsen AK, Bouwman FG, Munnia A, Peluso M, Mariman E, van Schooten FJ. Paternal Exposure to Environmental Chemical Stress Affects Male Offspring's Hepatic Mitochondria. Toxicol Sci 2019; 162:241-250. [PMID: 29145655 DOI: 10.1093/toxsci/kfx246] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Preconceptional paternal exposures may affect offspring's health, which cannot be explained by mutations in germ cells, but by persistent changes in the regulation of gene expression. Therefore, we investigated whether pre-conceptional paternal exposure to benzo[a]pyrene (B[a]P) could alter the offspring's phenotype. Male C57BL/6 mice were exposed to B[a]P by gavage for 6 weeks, 3× per week, and were crossed with unexposed BALB-c females 6 weeks after the final exposure. The offspring was kept under normal feeding conditions and was sacrificed at 3 weeks of age. Analysis of the liver proteome by 2D-gel electrophoresis and mass spectrometry indicated that proteins involved in mitochondrial function were significantly downregulated in the offspring of exposed fathers. This down-regulation of mitochondrial proteins was paralleled by a reduction in mitochondrial DNA copy number and reduced activity of citrate synthase and β-hydroxyacyl-CoA dehydrogenase, but in male offspring only. Surprisingly, analysis of hepatic mRNA expression revealed a male-specific up-regulation of the genes, whose proteins were downregulated, including Aldh2 and Ogg1. This discrepancy could be related to several selected microRNA (miRNA)'s that regulate the translation of these proteins; miRNA-122, miRNA-129-2-5p, and miRNA-1941 were upregulated in a gender-specific manner. Since mitochondria are thought to be a source of intracellular reactive oxygen species, we additionally assessed oxidatively-induced DNA damage. Both 8-hydroxy-deoxyguanosine and malondialdehyde-dG adduct levels were significantly reduced in male offspring of exposed fathers. In conclusion, we show that paternal exposure to B[a]P can regulate mitochondrial metabolism in offspring, which may have profound implications for our understanding of health and disease risk inherited from fathers.
Collapse
Affiliation(s)
- Roger Godschalk
- Department of Pharmacology & Toxicology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, 6200MD Maastricht, The Netherlands
| | - Alex Remels
- Department of Pharmacology & Toxicology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, 6200MD Maastricht, The Netherlands
| | - Camiel Hoogendoorn
- Department of Pharmacology & Toxicology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, 6200MD Maastricht, The Netherlands
| | - Jan van Benthem
- Laboratory for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Mirjam Luijten
- Laboratory for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Nur Duale
- Department of Molecular Biology, Norwegian Institute of Public Health, Nydalen, Oslo, Norway
| | - Gunnar Brunborg
- Department of Molecular Biology, Norwegian Institute of Public Health, Nydalen, Oslo, Norway
| | - Ann-Karin Olsen
- Department of Molecular Biology, Norwegian Institute of Public Health, Nydalen, Oslo, Norway
| | - Freek G Bouwman
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Armelle Munnia
- Cancer Risk Factor Branch, Cancer Prevention Laboratory, ISPO-Cancer Prevention and Research Institute, Florence, Italy
| | - Marco Peluso
- Cancer Risk Factor Branch, Cancer Prevention Laboratory, ISPO-Cancer Prevention and Research Institute, Florence, Italy
| | - Edwin Mariman
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Frederik Jan van Schooten
- Department of Pharmacology & Toxicology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, 6200MD Maastricht, The Netherlands
| |
Collapse
|
32
|
Portha B, Grandjean V, Movassat J. Mother or Father: Who Is in the Front Line? Mechanisms Underlying the Non-Genomic Transmission of Obesity/Diabetes via the Maternal or the Paternal Line. Nutrients 2019; 11:E233. [PMID: 30678214 PMCID: PMC6413176 DOI: 10.3390/nu11020233] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/01/2019] [Accepted: 01/09/2019] [Indexed: 02/06/2023] Open
Abstract
Extensive epidemiological and experimental evidence have shown that exposure to an adverse intrauterine environment as observed in offspring of pregnancies complicated by obesity or diabetes, can program susceptibility to metabolic, endocrine and cardiovascular disorders later in life. Although most studies have concentrated on the maternal environment, it is also becoming evident that paternal exposure to obesity or diabetes can result in the later development of metabolic disorders in the offspring. Such programmed effects might not be limited to the first directly exposed generation, but could be transmitted to subsequent generations. This suggests the existence of mechanisms by which metabolic changes in parental phenotype are transmissible to offspring. The mechanisms which underpin the transmission of the programmed effects across generations are still unclear. However, epigenetic regulation of transcription has emerged as a strong candidate for mediating the heritability of metabolic diseases. Here, we review the most relevant evidence from human and animal studies showing transmission of programming effects of obesity or diabetes across generations, and the current mechanisms underlying either maternal or paternal influences on the metabolic status of offspring.
Collapse
Affiliation(s)
- Bernard Portha
- Sorbonne-Paris-Cité, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptative), Université Paris-Diderot, CNRS UMR 8251, F-75205 Paris CEDEX 13, France.
| | - Valérie Grandjean
- Inserm U1065 C3M, Team Control of Gene Expression (10), Université Côte d'Azur, 151 Route de Ginestière, 06204 Nice CEDEX 3, France.
| | - Jamileh Movassat
- Sorbonne-Paris-Cité, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptative), Université Paris-Diderot, CNRS UMR 8251, F-75205 Paris CEDEX 13, France.
| |
Collapse
|
33
|
Bozack AK, Cardenas A, Quamruzzaman Q, Rahman M, Mostofa G, Christiani DC, Kile ML. DNA methylation in cord blood as mediator of the association between prenatal arsenic exposure and gestational age. Epigenetics 2018; 13:923-940. [PMID: 30175652 PMCID: PMC6284783 DOI: 10.1080/15592294.2018.1516453] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/06/2018] [Accepted: 08/22/2018] [Indexed: 01/08/2023] Open
Abstract
Prenatal arsenic exposure is associated with adverse birth outcomes and disease risk later in life, which could be mediated through epigenetic dysregulation. We evaluated the association between arsenic and gestational age (GA) that was mediated through DNA methylation (DNAm) using data from a Bangladeshi birth cohort. Arsenic exposure was measured in maternal drinking water at ≤16 weeks GA and maternal toenails collected ≤1 month postpartum. Cord blood DNAm was measured using Infinium HumanMethylation450 arrays (n = 44, discovery phase). Top loci identified in the discovery phase were then pyrosequenced in a second group (n = 569, validation phase). Structural equation models (SEM) evaluated the direct and indirect effects of arsenic and DNAm on GA. In the discovery phase, arsenic was associated with differential DNAm of 139 loci that were associated with GA (P < 1.10X10-6; |β regression|>0.10). Each doubling in water arsenic concentration decreased GA by 2 days, which was fully mediated through the main principal component of the top-ten CpGs (P < 0.001). In the validation phase, there were direct and indirect effects of miR214-3 and MCC DNAm on GA. In an adjusted SEM model, mediation of the association between arsenic and GA by miR124-3 was borderline significant (P = 0.061). This study therefore identified DNAm at specific loci in cord blood that mediated the effect of arsenic exposure on GA. Specifically, prenatal arsenic exposure was associated with lower methylation of miR124-3 that mediated the exposure-response of arsenic on GA. Future research should evaluate if these epigenetic changes are persistent and associated with disease risk.
Collapse
Affiliation(s)
- Anne K. Bozack
- Department of Environmental Health Sciences, Columbia University, New York, NY, USA
| | - Andres Cardenas
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | | | | | | | - David C. Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Molly L. Kile
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
34
|
Liu Y. Darwin's Pangenesis and the Lamarckian Inheritance of Acquired Characters. ADVANCES IN GENETICS 2018; 101:115-144. [PMID: 30037391 DOI: 10.1016/bs.adgen.2018.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Since the earliest days of evolutionary thought, the problem of the inheritance of acquired characters has been a central debate. Darwin accepted the inheritance of acquired characters as an established fact and gave many instances. His Pangenesis was more than anything else an attempt to provide a theory for its explanation. Over the past several decades, there has been increasing evidence for the inheritance of acquired habit and immunity, and for heritable changes induced by food and fertilizer, stress, chemicals, temperature, light and other environmental factors. Many studies also suggest that parental age has certain influences on the characters of offspring. The current explanations include environmentally induced DNA changes (mainly DNA rearrangements and DNA methylation), RNA-mediated inheritance, and horizontal gene transfer. These mechanistic explanations are consistent with Darwin's Pangenesis.
Collapse
Affiliation(s)
- Yongsheng Liu
- Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, China; Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
35
|
Wu S, Guo J, Zhu L, Yang J, Chen S, Yang X. Identification and characterisation of microRNAs and Piwi-interacting RNAs in cockerels' spermatozoa by Solexa sequencing. Br Poult Sci 2018; 59:371-380. [PMID: 29667432 DOI: 10.1080/00071668.2018.1464123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
1. There has been substantial research focused on the roles of microRNAs (miRNAs) and Piwi-interacting RNAs (piRNAs) derived from mammalian spermatozoa; however, comparatively little is known about the role of spermatozoa-derived miRNAs and piRNAs within breeding cockerels' spermatozoa. 2. A small RNA library of cockerels' spermatozoa was constructed using Illumina high-throughput sequencing technology. Unique sequences with lengths of 18-26 nucleotides were mapped to miRBase 21.0 and unique sequences with lengths of 25-37 nucleotides were mapped to a piRNA database. A total of 1311 miRNAs and 2448 potential piRNAs were identified. Based on stem-loop qRT-PCR, 8 miRNAs were validated. 3. Potential target genes of the abundant miRNAs were predicted, and further Kyoto Encyclopedia of Genes and Genomes database (KEGG) and Gene Ontology (GO) analyses were performed, which revealed that some candidate miRNAs were involved in the spermatogenesis process, spermatozoa epigenetic programming and further embryonic development. 5. GO and KEGG analyses based on mapping genes of expressed piRNAs were performed, which revealed that spermatozoal piRNAs could play important regulatory roles in embryonic development of offspring. 6. The search for endogenous spermatozoa miRNAs and piRNAs will contribute to a preliminary database for functional and molecular mechanistic studies in embryonic development and spermatozoa epigenetic programming.
Collapse
Affiliation(s)
- S Wu
- a College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi , China
| | - J Guo
- a College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi , China
| | - L Zhu
- a College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi , China
| | - J Yang
- a College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi , China
| | - S Chen
- a College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi , China
| | - X Yang
- a College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi , China
| |
Collapse
|
36
|
Abstract
Background Developmental programming of the embryo is controlled by genetic information but also dictated by epigenetic information contained in spermatozoa. Lifestyle and environmental factors not only influence health in one individual but can also affect the phenotype of the following generations. This is mediated via epigenetic inheritance i.e., gametic transmission of environmentally-driven epigenetic information to the offspring. Evidence is accumulating that preconceptional exposure to certain lifestyle and environmental factors, such as diet, physical activity, and smoking, affects the phenotype of the next generation through remodeling of the epigenetic blueprint of spermatozoa. Scope of Review This review will summarize current knowledge about the different epigenetic signals in sperm that are responsive to environmental and lifestyle factors and are capable of affecting embryonic development and the phenotype of the offspring later in life. Major conclusions Like somatic cells, the epigenome of spermatozoa has proven to be dynamically reactive to a wide variety of environmental and lifestyle stressors. The functional consequence on embryogenesis and phenotype of the next generation remains largely unknown. However, strong evidence of environmentally-driven sperm-borne epigenetic factors, which are capable of altering the phenotype of the next generation, is emerging on a large scale.
Collapse
|
37
|
Review: Sperm-oocyte interactions and their implications for bull fertility, with emphasis on the ubiquitin-proteasome system. Animal 2018; 12:s121-s132. [PMID: 29477154 DOI: 10.1017/s1751731118000253] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Fertilization is an intricate cascade of events that irreversibly alter the participating male and female gamete and ultimately lead to the union of paternal and maternal genomes in the zygote. Fertilization starts with sperm capacitation within the oviductal sperm reservoir, followed by gamete recognition, sperm-zona pellucida interactions and sperm-oolemma adhesion and fusion, followed by sperm incorporation, oocyte activation, pronuclear development and embryo cleavage. At fertilization, bull spermatozoon loses its acrosome and plasma membrane components and contributes chromosomes, centriole, perinuclear theca proteins and regulatory RNAs to the zygote. While also incorporated in oocyte cytoplasm, structures of the sperm tail, including mitochondrial sheath, axoneme, fibrous sheath and outer dense fibers are degraded and recycled. The ability of some of these sperm contributed components to give rise to functional zygotic structures and properly induce embryonic development may vary between bulls, bearing on their reproductive performance, and on the fitness, health, fertility and production traits of their offspring. Proper functioning, recycling and remodeling of gamete structures at fertilization is aided by the ubiquitin-proteasome system (UPS), the universal substrate-specific protein recycling pathway present in bovine and other mammalian oocytes and spermatozoa. This review is focused on the aspects of UPS relevant to bovine fertilization and bull fertility.
Collapse
|
38
|
The "evolutionary field" hypothesis. Non-Mendelian transgenerational inheritance mediates diversification and evolution. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 134:27-37. [PMID: 29223657 DOI: 10.1016/j.pbiomolbio.2017.12.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/17/2017] [Accepted: 12/05/2017] [Indexed: 12/23/2022]
Abstract
Epigenetics is increasingly regarded as a potential contributing factor to evolution. Building on apparently unrelated results, here I propose that RNA-containing nanovesicles, predominantly small regulatory RNAs, are released from somatic tissues in the bloodstream, cross the Weismann barrier, reach the epididymis, and are eventually taken up by spermatozoa; henceforth the information is delivered to oocytes at fertilization. In the model, a LINE-1-encoded reverse transcriptase activity, present in spermatozoa and early embryos, plays a key role in amplifying and propagating these RNAs as extrachromosomal structures. It may be conceived that, over generations, the cumulative effects of sperm-delivered RNAs would cross a critical threshold and overcome the buffering capacity of embryos. As a whole, the process can promote the generation of an information-containing platform that drives the reshaping of the embryonic epigenetic landscape with the potential to generate ontogenic changes and redirect the evolutionary trajectory. Over time, evolutionary significant, stably acquired variations could be generated through the process. The interplay between these elements defines the concept of "evolutionary field", a self-consistent, comprehensive information-containing platform and a source of discontinuous evolutionary novelty.
Collapse
|
39
|
Raad G, Hazzouri M, Bottini S, Trabucchi M, Azoury J, Grandjean V. Paternal obesity: how bad is it for sperm quality and progeny health? Basic Clin Androl 2017; 27:20. [PMID: 29123667 PMCID: PMC5657098 DOI: 10.1186/s12610-017-0064-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
There is substantial evidence that paternal obesity is associated not only with an increased incidence of infertility, but also with an increased risk of metabolic disturbance in adult offspring. Apparently, several mechanisms may contribute to the sperm quality alterations associated with paternal obesity, such as physiological/hormonal alterations, oxidative stress, and epigenetic alterations. Along these lines, modifications of hormonal profiles namely reduced androgen levels and elevated estrogen levels, were found associated with lower sperm concentration and seminal volume. Additionally, oxidative stress in testis may induce an increase of the percentage of sperm with DNA fragmentation. The latter, relate to other peculiarities such as alteration of the embryonic development, increased risk of miscarriage, and development of chronic morbidity in the offspring, including childhood cancers. Undoubtedly, epigenetic alterations (ie, DNA methylation, chromatin modifications, and small RNA deregulation) of sperm related to paternal obesity and their consequences on the progeny are poorly understood determinants of paternal obesity-induced transmission. In this review, we summarize and discuss the data available in the literature regarding the biological, physiological, and molecular consequences of paternal obesity on male fertility potential and ultimately progeny health.
Collapse
Affiliation(s)
- Georges Raad
- Azoury-IVF clinic, Mount Lebanon Hospital, 5th floor, Camille Chamoun bvd, Beirut, Lebanon.,Université Cote d'Azur, Inserm, C3M (team 10), Nice, France
| | - Mira Hazzouri
- Faculty of Sciences 2, Lebanese University, Fanar, Lebanon
| | - Silvia Bottini
- Université Cote d'Azur, Inserm, C3M (team 10), Nice, France
| | | | - Joseph Azoury
- Azoury-IVF clinic, Mount Lebanon Hospital, 5th floor, Camille Chamoun bvd, Beirut, Lebanon
| | | |
Collapse
|
40
|
Gapp K, Bohacek J. Epigenetic germline inheritance in mammals: looking to the past to understand the future. GENES BRAIN AND BEHAVIOR 2017; 17:e12407. [PMID: 28782190 DOI: 10.1111/gbb.12407] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 07/15/2017] [Accepted: 08/03/2017] [Indexed: 12/25/2022]
Abstract
Life experiences can induce epigenetic changes in mammalian germ cells, which can influence the developmental trajectory of the offspring and impact health and disease across generations. While this concept of epigenetic germline inheritance has long been met with skepticism, evidence in support of this route of information transfer is now overwhelming, and some key mechanisms underlying germline transmission of acquired information are emerging. This review focuses specifically on sperm RNAs as causal vectors of inheritance. We examine how they might become altered in the germline, and how different classes of sperm RNAs might interact with other epimodifications in germ cells or in the zygote. We integrate the latest findings with earlier pioneering work in this field, point out major questions and challenges, and suggest how new experiments could address them.
Collapse
Affiliation(s)
- K Gapp
- Gurdon Institute, University of Cambridge, Cambridge, UK.,Wellcome Trust Sanger Institute, Hinxton, UK
| | - J Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Department of Health Sciences and Technology of ETH Zurich, Neuroscience Center Zurich, Switzerland
| |
Collapse
|
41
|
Capovilla M, Robichon A, Rassoulzadegan M. A new paramutation-like example at the Delta gene of Drosophila. PLoS One 2017; 12:e0172780. [PMID: 28355214 PMCID: PMC5371283 DOI: 10.1371/journal.pone.0172780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 02/09/2017] [Indexed: 11/26/2022] Open
Abstract
The hereditary transmission of a phenotype independent from DNA sequence implies epigenetic effects. Paramutation is a heritable epigenetic phenomenon observed in plants and animals. To investigate paramutation in Drosophila, we used the P{ry+t7.2= PZ}Dl05151 P-element insertion in the Drosophila melanogaster genome that causes a dominant visible phenotype: the presence of characteristic extra-veins in the fly wings. This extra-vein phenotype presents variable expressivity and incomplete penetrance. The insert is a PZ element located 680 bp upstream from the ATG of the Delta (Dl) gene, encoding the Notch ligand involved in wing vein development, and acts as a null allele. In the G2 offspring from a cross between the heterozygous transgenic stock and wild-type flies, we observed the transmission of the extra-vein phenotype to wild-type flies without the transgene, independently of gender and across many generations. This is a “paramutation-like” example in the fly: the heritable transmission of a phenotypic change not linked to a classical genetic mutation. A “paramutagenic” allele in heterozygotes transmits the phenotype of the heterozygotes to the wild-type allele (“paramutant”) in a stable manner through generations. Distinct from paramutation events so far described in Drosophila, here we deal with a dominant effect on a single gene involving variable hereditary signals.
Collapse
Affiliation(s)
- Maria Capovilla
- UMR 1355–7254 INRA/Université Côte d’Azur/CNRS, Institut Sophia Agrobiotech, 400 route des Chappes, Sophia Antipolis, France
- * E-mail:
| | - Alain Robichon
- UMR 1355–7254 INRA/Université Côte d’Azur/CNRS, Institut Sophia Agrobiotech, 400 route des Chappes, Sophia Antipolis, France
| | | |
Collapse
|
42
|
Small RNA-directed epigenetic programming of embryonic stem cell cardiac differentiation. Sci Rep 2017; 7:41799. [PMID: 28165496 PMCID: PMC5292948 DOI: 10.1038/srep41799] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 12/30/2016] [Indexed: 02/06/2023] Open
Abstract
Microinjection of small noncoding RNAs in one-cell embryos was reported in several instances to result in transcriptional activation of target genes. To determine the molecular mechanisms involved and to explore whether such epigenetic regulations could play a role in early development, we used a cell culture system as close as possible to the embryonic state. We report efficient cardiac differentiation of embryonic stem (ES) cells induced by small non-coding RNAs with sequences of Cdk9, a key player in cardiomyocyte differentiation. Transfer of oligoribonucleotides representing parts of the Cdk9 mRNA into ES and mouse embryo fibroblast cultures resulted in upregulation of transcription. Dependency on Argonaute proteins and endogenous antisense transcripts indicated that the inducer oligoribonucleotides were processed by the RNAi machinery. Upregulation of Cdk9 expression resulted in increased efficiency of cardiac differentiation suggesting a potential tool for stem cell-based regenerative medicine.
Collapse
|
43
|
Chen Q, Yan W, Duan E. Epigenetic inheritance of acquired traits through sperm RNAs and sperm RNA modifications. Nat Rev Genet 2016; 17:733-743. [PMID: 27694809 PMCID: PMC5441558 DOI: 10.1038/nrg.2016.106] [Citation(s) in RCA: 335] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Once deemed heretical, emerging evidence now supports the notion that the inheritance of acquired characteristics can occur through ancestral exposures or experiences and that certain paternally acquired traits can be 'memorized' in the sperm as epigenetic information. The search for epigenetic factors in mammalian sperm that transmit acquired phenotypes has recently focused on RNAs and, more recently, RNA modifications. Here, we review insights that have been gained from studying sperm RNAs and RNA modifications, and their roles in influencing offspring phenotypes. We discuss the possible mechanisms by which sperm become acquisitive following environmental-somatic-germline interactions, and how they transmit paternally acquired phenotypes by shaping early embryonic development.
Collapse
Affiliation(s)
- Qi Chen
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada 89512, USA
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada 89512, USA
| | - Enkui Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
44
|
Fullston T, Ohlsson-Teague EMC, Print CG, Sandeman LY, Lane M. Sperm microRNA Content Is Altered in a Mouse Model of Male Obesity, but the Same Suite of microRNAs Are Not Altered in Offspring's Sperm. PLoS One 2016; 11:e0166076. [PMID: 27814400 PMCID: PMC5096664 DOI: 10.1371/journal.pone.0166076] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/21/2016] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity is increasing worldwide and has tripled in men of reproductive age since the 1970s. Concerningly, obesity is not only comorbid with other chronic diseases, but there is mounting evidence that it increases the non-communicable disease load in their children (eg mortality, obesity, autism). Animal studies have demonstrated that paternal obesity increases the risk of metabolic (eg glucose metabolism defects, obesity) and reproductive disorders in offspring. Epigenetic changes within sperm are clear mechanistic candidates that are associated with both changes to the father’s environment and offspring phenotype. Specifically there is emerging evidence that a father’s sperm microRNA content both responds to paternal environmental cues and alters the gene expression profile and subsequent development of the early embryo. We used a mouse model of high fat diet (HFD) induced obesity to investigate whether male obesity could modulate sperm microRNA content. We also investigated whether this alteration to a father’s sperm microRNA content lead to a similar change in the sperm of male offspring. Our investigations were initially guided by a Taqman PCR array, which indicated the differential abundance of 28 sperm borne microRNAs in HFD mice. qPCR confirmation in a much larger cohort of founder males demonstrated that 13 of these microRNAs were differentially abundant (11 up-regulated; 2 down-regulated) due to HFD feeding. Despite metabolic and reproductive phenotypes also being observed in grand-offspring fathered via the male offspring lineage, there was no evidence that any of the 13 microRNAs were also dysregulated in male offspring sperm. This was presumably due to the variation seen within both groups of offspring and suggests other mechanisms might act between offspring and grand-offspring. Thus 13 sperm borne microRNAs are modulated by a father’s HFD and the presumed transfer of this altered microRNA payload to the embryo at fertilisation potentially acts to alter the embryonic molecular makeup post-fertilisation, altering its growth trajectory, ultimately affecting adult offspring phenotype and may contribute to paternal programming.
Collapse
Affiliation(s)
- Tod Fullston
- Discipline of Obstetrics & Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia 5005, Australia
- Freemason’s Foundation Centre for Men’s Health, The University of Adelaide, Adelaide, South Australia 5005, Australia
- * E-mail:
| | - E. Maria C. Ohlsson-Teague
- Discipline of Obstetrics & Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Cristin G. Print
- Department of Molecular Medicine & Pathology and New Zealand Bioinformatics Institute, University of Auckland, Auckland 1142, New Zealand
| | - Lauren Y. Sandeman
- Discipline of Obstetrics & Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Michelle Lane
- Discipline of Obstetrics & Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia 5005, Australia
- Freemason’s Foundation Centre for Men’s Health, The University of Adelaide, Adelaide, South Australia 5005, Australia
- Monash IVF Group, Melbourne, Victoria 3168, Australia
| |
Collapse
|
45
|
Transgenerational inheritance of enhanced susceptibility to radiation-induced medulloblastoma in newborn Ptch1⁺/⁻ mice after paternal irradiation. Oncotarget 2016; 6:36098-112. [PMID: 26452034 PMCID: PMC4742164 DOI: 10.18632/oncotarget.5553] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/21/2015] [Indexed: 12/21/2022] Open
Abstract
The hypothesis of transgenerational induction of increased cancer susceptibility after paternal radiation exposure has long been controversial because of inconsistent results and the lack of a mechanistic interpretation. Here, exploiting Ptch1 heterozygous knockout mice, susceptible to spontaneous and radiation-induced medulloblastoma, we show that exposure of paternal germ cells to 1 Gy X-rays, at the spermatogonial stage, increased by a considerable 1.4-fold the offspring susceptibility to medulloblastoma induced by neonatal irradiation. This effect gained further biological significance thanks to a number of supporting data on the immunohistochemical characterization of the target tissue and preneoplastic lesions (PNLs). These results altogether pointed to increased proliferation of cerebellar granule cell precursors and PNLs cells, which favoured the development of frank tumours. The LOH analysis of tumor DNA showed Ptch1 biallelic loss in all tumor samples, suggesting that mechanisms other than interstitial deletions, typical of radiation-induced medulloblastoma, did not account for the observed increased cancer risk. This data was supported by comet analysis showing no differences in DNA damage induction and repair in cerebellar cells as a function of paternal irradiation. Finally, we provide biological plausibility to our results offering evidence of a possible epigenetic mechanism of inheritance based on radiation-induced changes of the microRNA profile of paternal sperm.
Collapse
|
46
|
|
47
|
A review of fundamental principles for animal models of DOHaD research: an Australian perspective. J Dev Orig Health Dis 2016; 7:449-472. [DOI: 10.1017/s2040174416000477] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Epidemiology formed the basis of ‘the Barker hypothesis’, the concept of ‘developmental programming’ and today’s discipline of the Developmental Origins of Health and Disease (DOHaD). Animal experimentation provided proof of the underlying concepts, and continues to generate knowledge of underlying mechanisms. Interventions in humans, based on DOHaD principles, will be informed by experiments in animals. As knowledge in this discipline has accumulated, from studies of humans and other animals, the complexity of interactions between genome, environment and epigenetics, has been revealed. The vast nature of programming stimuli and breadth of effects is becoming known. As a result of our accumulating knowledge we now appreciate the impact of many variables that contribute to programmed outcomes. To guide further animal research in this field, the Australia and New Zealand DOHaD society (ANZ DOHaD) Animals Models of DOHaD Research Working Group convened at the 2nd Annual ANZ DOHaD Congress in Melbourne, Australia in April 2015. This review summarizes the contributions of animal research to the understanding of DOHaD, and makes recommendations for the design and conduct of animal experiments to maximize relevance, reproducibility and translation of knowledge into improving health and well-being.
Collapse
|
48
|
Iosef Husted C, Valencik M. Insulin-like growth factors and their potential role in cardiac epigenetics. J Cell Mol Med 2016; 20:1589-602. [PMID: 27061217 PMCID: PMC4956935 DOI: 10.1111/jcmm.12845] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/24/2016] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) constitutes a major public health threat worldwide, accounting for 17.3 million deaths annually. Heart disease and stroke account for the majority of healthcare costs in the developed world. While much has been accomplished in understanding the pathophysiology, molecular biology and genetics underlying the diagnosis and treatment of CVD, we know less about the role of epigenetics and their molecular determinants. The impact of environmental changes and epigenetics in CVD is now emerging as critically important in understanding the origin of disease and the development of new therapeutic approaches to prevention and treatment. This review focuses on the emerging role of epigenetics mediated by insulin like-growth factors-I and -II in major CVDs such as heart failure, cardiac hypertrophy and diabetes.
Collapse
Affiliation(s)
- Cristiana Iosef Husted
- Department of Pharmacology, University of Nevada, Reno School of Medicine (UNSOM), Reno, NV, USA
| | - Maria Valencik
- Department of Pharmacology, University of Nevada, Reno School of Medicine (UNSOM), Reno, NV, USA
| |
Collapse
|
49
|
Marczylo EL, Jacobs MN, Gant TW. Environmentally induced epigenetic toxicity: potential public health concerns. Crit Rev Toxicol 2016; 46:676-700. [PMID: 27278298 PMCID: PMC5030620 DOI: 10.1080/10408444.2016.1175417] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Throughout our lives, epigenetic processes shape our development and enable us to adapt to a constantly changing environment. Identifying and understanding environmentally induced epigenetic change(s) that may lead to adverse outcomes is vital for protecting public health. This review, therefore, examines the present understanding of epigenetic mechanisms involved in the mammalian life cycle, evaluates the current evidence for environmentally induced epigenetic toxicity in human cohorts and rodent models and highlights the research considerations and implications of this emerging knowledge for public health and regulatory toxicology. Many hundreds of studies have investigated such toxicity, yet relatively few have demonstrated a mechanistic association among specific environmental exposures, epigenetic changes and adverse health outcomes in human epidemiological cohorts and/or rodent models. While this small body of evidence is largely composed of exploratory in vivo high-dose range studies, it does set a precedent for the existence of environmentally induced epigenetic toxicity. Consequently, there is worldwide recognition of this phenomenon, and discussion on how to both guide further scientific research towards a greater mechanistic understanding of environmentally induced epigenetic toxicity in humans, and translate relevant research outcomes into appropriate regulatory policies for effective public health protection.
Collapse
Affiliation(s)
- Emma L Marczylo
- a Toxicology Department, CRCE, PHE, Chilton , Oxfordshire , UK
| | - Miriam N Jacobs
- a Toxicology Department, CRCE, PHE, Chilton , Oxfordshire , UK
| | - Timothy W Gant
- a Toxicology Department, CRCE, PHE, Chilton , Oxfordshire , UK
| |
Collapse
|
50
|
Ghanbarian H, Wagner N, Polo B, Baudouy D, Kiani J, Michiels JF, Cuzin F, Rassoulzadegan M, Wagner KD. Dnmt2/Trdmt1 as Mediator of RNA Polymerase II Transcriptional Activity in Cardiac Growth. PLoS One 2016; 11:e0156953. [PMID: 27270731 PMCID: PMC4894585 DOI: 10.1371/journal.pone.0156953] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/23/2016] [Indexed: 01/04/2023] Open
Abstract
Dnmt2/Trdmt1 is a methyltransferase, which has been shown to methylate tRNAs. Deficient mutants were reported to exhibit various, seemingly unrelated, defects in development and RNA-mediated epigenetic heredity. Here we report a role in a distinct developmental regulation effected by a noncoding RNA. We show that Dnmt2-deficiency in mice results in cardiac hypertrophy. Echocardiographic measurements revealed that cardiac function is preserved notwithstanding the increased dimensions of the organ due to cardiomyocyte enlargement. Mechanistically, activation of the P-TEFb complex, a critical step for cardiac growth, results from increased dissociation of the negatively regulating Rn7sk non-coding RNA component in Dnmt2-deficient cells. Our data suggest that Dnmt2 plays an unexpected role for regulation of cardiac growth by modulating activity of the P-TEFb complex.
Collapse
Affiliation(s)
- Hossein Ghanbarian
- Biotechnology Department, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nicole Wagner
- Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France
- Univ. Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100, Nice, France
| | - Beatrice Polo
- Univ. Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100, Nice, France
| | - Delphine Baudouy
- Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France
- Univ. Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100, Nice, France
| | - Jafar Kiani
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Jean-François Michiels
- Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France
- Department of Pathology, CHU Nice, Nice, France
| | - François Cuzin
- Univ. Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100, Nice, France
| | - Minoo Rassoulzadegan
- Univ. Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100, Nice, France
- * E-mail: (MR); (KDW)
| | - Kay-Dietrich Wagner
- Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France
- Univ. Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100, Nice, France
- * E-mail: (MR); (KDW)
| |
Collapse
|