Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(16): e2314885121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38588413

ABSTRACT

As a result of partial hepatectomy, the remaining liver tissue undergoes a process of renewed proliferation that leads to rapid regeneration of the liver. By following the early stages of this process, we observed dramatic programmed changes in the DNA methylation profile, characterized by both de novo and demethylation events, with a subsequent return to the original adult pattern as the liver matures. Strikingly, these transient alterations partially mimic the DNA methylation state of embryonic hepatoblasts (E16.5), indicating that hepatocytes actually undergo epigenetic dedifferentiation. Furthermore, Tet2/Tet3-deletion experiments demonstrated that these changes in methylation are necessary for carrying out basic embryonic functions, such as proliferation, a key step in liver regeneration. This implies that unlike tissue-specific regulatory regions that remain demethylated in the adult, early embryonic genes are programmed to first undergo demethylation, followed by remethylation as development proceeds. The identification of this built-in system may open targeting opportunities for regenerative medicine.


Subject(s)
DNA Methylation , Embryo, Mammalian , Embryo, Mammalian/metabolism , Hepatocytes
2.
Nat Commun ; 14(1): 3359, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37291192

ABSTRACT

Human trophoblast stem cells (hTSCs) can be derived from embryonic stem cells (hESCs) or be induced from somatic cells by OCT4, SOX2, KLF4 and MYC (OSKM). Here we explore whether the hTSC state can be induced independently of pluripotency, and what are the mechanisms underlying its acquisition. We identify GATA3, OCT4, KLF4 and MYC (GOKM) as a combination of factors that can generate functional hiTSCs from fibroblasts. Transcriptomic analysis of stable GOKM- and OSKM-hiTSCs reveals 94 hTSC-specific genes that are aberrant specifically in OSKM-derived hiTSCs. Through time-course-RNA-seq analysis, H3K4me2 deposition and chromatin accessibility, we demonstrate that GOKM exert greater chromatin opening activity than OSKM. While GOKM primarily target hTSC-specific loci, OSKM mainly induce the hTSC state via targeting hESC and hTSC shared loci. Finally, we show that GOKM efficiently generate hiTSCs from fibroblasts that harbor knockout for pluripotency genes, further emphasizing that pluripotency is dispensable for hTSC state acquisition.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells , Humans , Cellular Reprogramming/genetics , Trophoblasts , Fibroblasts , Embryonic Stem Cells , Chromatin/genetics , Octamer Transcription Factor-3/genetics
3.
Proc Natl Acad Sci U S A ; 119(52): e2212306119, 2022 12 27.
Article in English | MEDLINE | ID: mdl-36534800

ABSTRACT

Injury to muscle brings about the activation of stem cells, which then generate new myocytes to replace damaged tissue. We demonstrate that this activation is accompanied by a dramatic change in the stem-cell methylation pattern that prepares them epigenetically for terminal myocyte differentiation. These de- and de novo methylation events occur at regulatory elements associated with genes involved in myogenesis and are necessary for activation and regeneration. Local injury of one muscle elicits an almost identical epigenetic change in satellite cells from other muscles in the body, in a process mediated by circulating factors. Furthermore, this same methylation state is also generated in muscle stem cells (MuSCs) of female animals following pregnancy, even in the absence of any injury. Unlike the activation-induced expression changes, which are transient, the induced methylation profile is stably maintained in resident MuSCs and thus represents a molecular memory of previous physiological events that is probably programmed to provide a mechanism for long-term adaptation.


Subject(s)
DNA Methylation , Muscle, Skeletal , Animals , Female , Muscle, Skeletal/metabolism , Stem Cells/metabolism , Cell Differentiation/genetics , Epigenesis, Genetic , Muscle Development/genetics , Regeneration/genetics
4.
Development ; 149(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-35051273

ABSTRACT

Although it is well known that DNA methylation serves to repress gene expression, precisely how it functions during the process of development remains unclear. Here, we propose that the overall pattern of DNA methylation established in the early embryo serves as a sophisticated mechanism for maintaining a genome-wide network of gene regulatory elements in an inaccessible chromatin structure throughout the body. As development progresses, programmed demethylation in each cell type then provides the specificity for maintaining select elements in an open structure. This allows these regulatory elements to interact with a large range of transcription factors and thereby regulate the gene expression profiles that define cell identity.


Subject(s)
DNA Methylation , Gene Expression Regulation, Developmental , Animals , Chromatin Assembly and Disassembly , Humans
5.
Front Cell Dev Biol ; 9: 737681, 2021.
Article in English | MEDLINE | ID: mdl-34660595

ABSTRACT

Developmental programming is carried out by a sequence of molecular choices that epigenetically mark the genome to generate the stable cell types which make up the total organism. A number of important processes, such as genomic imprinting, selection of immune or olfactory receptors, and X-chromosome inactivation in females are dependent on the ability to stably choose one single allele in each cell. In this perspective, we propose that asynchronous replication timing (ASRT) serves as the basis for a sophisticated universal mechanism for mediating and maintaining these decisions.

6.
Nat Commun ; 12(1): 1035, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33589603

ABSTRACT

Stochastic asynchronous replication timing (AS-RT) is a phenomenon in which the time of replication of each allele is different, and the identity of the early allele varies between cells. By taking advantage of stable clonal pre-B cell populations derived from C57BL6/Castaneous mice, we have mapped the genome-wide AS-RT loci, independently of genetic differences. These regions are characterized by differential chromatin accessibility, mono-allelic expression and include new gene families involved in specifying cell identity. By combining population level mapping with single cell FISH, our data reveal the existence of a novel regulatory program that coordinates a fixed relationship between AS-RT regions on any given chromosome, with some loci set to replicate in a parallel and others set in the anti-parallel orientation. Our results show that AS-RT is a highly regulated epigenetic mark established during early embryogenesis that may be used for facilitating the programming of mono-allelic choice throughout development.


Subject(s)
Bone Marrow Cells/metabolism , Chromatin/chemistry , DNA Replication Timing , Epigenesis, Genetic , Genome , Precursor Cells, B-Lymphoid/metabolism , Alleles , Animals , Bone Marrow Cells/cytology , Chromatin/metabolism , Chromatin/ultrastructure , Clone Cells , Crosses, Genetic , Embryo, Mammalian , Embryonic Development/genetics , Female , Genetic Loci , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, B-Lymphoid/cytology
7.
Prenat Diagn ; 39(11): 1005-1010, 2019 10.
Article in English | MEDLINE | ID: mdl-31330572

ABSTRACT

Gestational age determination by traditional tools (last menstrual period, ultrasonography measurements and Ballard Maturational Assessment in newborns) has major limitations and therefore there is a need to find different approaches. In this study, we looked for a molecular marker that can be used to determine the accurate gestational age of the newborn. To this end, we performed reduced representation bisulfite sequencing (RRBS) on 41 cord blood and matching placenta samples from women between 25 and 40 weeks of gestation and generated an epigenetic clock based on the methylation level at different loci in the genome. We identified a set of 332 differentially methylated regions (DMRs) that undergo demethylation in late gestational age in cord blood cells and can predict the gestational age (r = -.7, P = 2E-05). Once the set of 411 DMRs that undergo de novo methylation in late gestational age was used in combination with the first set, it generated a more accurate clock (R = .77, P = 1.87E-05). We have compared gestational age determined by Ballard score assessment with our epigenetic clock and found high concordance. Taken together, this study demonstrates that DNA methylation can accurately predict gestational age and thus may serve as a good clinical predictor.


Subject(s)
DNA Methylation , Gestational Age , Biomarkers , Female , Genome, Human , Humans , Pregnancy
8.
Proc Natl Acad Sci U S A ; 115(41): 10387-10391, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30257947

ABSTRACT

Following erasure in the blastocyst, the entire genome undergoes de novo methylation at the time of implantation, with CpG islands being protected from this process. This bimodal pattern is then preserved throughout development and the lifetime of the organism. Using mouse embryonic stem cells as a model system, we demonstrate that the binding of an RNA polymerase complex on DNA before de novo methylation is predictive of it being protected from this modification, and tethering experiments demonstrate that the presence of this complex is, in fact, sufficient to prevent methylation at these sites. This protection is most likely mediated by the recruitment of enzyme complexes that methylate histone H3K4 over a local region and, in this way, prevent access to the de novo methylation complex. The topological pattern of H3K4me3 that is formed while the DNA is as yet unmethylated provides a strikingly accurate template for modeling the genome-wide basal methylation pattern of the organism. These results have far-reaching consequences for understanding the relationship between RNA transcription and DNA methylation.


Subject(s)
Blastocyst Inner Cell Mass/metabolism , DNA Methylation , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Transcription, Genetic , Animals , Blastocyst Inner Cell Mass/cytology , CpG Islands , DNA-Directed RNA Polymerases/metabolism , Embryo, Mammalian/cytology , Mice , Mice, Transgenic , Transcription Factors/metabolism
9.
Lancet ; 392(10149): 777-786, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30100054

ABSTRACT

DNA methylation represents an annotation system for marking the genetic text, thus providing instruction as to how and when to read the information and control transcription. Unlike sequence information, which is inherited, methylation patterns are established in a programmed process that continues throughout development, thus setting up stable gene expression profiles. This DNA methylation paradigm is a key player in medicine. Some changes in methylation closely correlate with age providing a marker for biological ageing, and these same sites could also play a part in cancer. The genome continues to undergo programmed variation in methylation after birth in response to environmental inputs, serving as a memory device that could affect ageing and predisposition to various metabolic, autoimmune, and neurological diseases. Taking advantage of tissue-specific differences, methylation can be used to detect cell death and thereby monitor many common diseases with a simple cell-free circulating-DNA blood test.


Subject(s)
Aging/genetics , Cell-Free Nucleic Acids/blood , DNA Methylation , Epigenesis, Genetic , Gene-Environment Interaction , Neoplasms/genetics , Biomarkers/blood , Diagnosis , Humans
10.
Nat Commun ; 9(1): 2040, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29795194

ABSTRACT

Development in mammals is accompanied by specific de novo and demethylation events that are thought to stabilize differentiated cell phenotypes. We demonstrate that a large percentage of the tissue-specific methylation pattern is generated postnatally. Demethylation in the liver is observed in thousands of enhancer-like sequences associated with genes that undergo activation during the first few weeks of life. Using. conditional gene ablation strategy we show that the removal of these methyl groups is stable and necessary for assuring proper hepatocyte gene expression and function through its effect on chromatin accessibility. These postnatal changes in methylation come about through exposure to hormone signaling. These results define the molecular rules of 5-methyl-cytosine regulation as an epigenetic mechanism underlying cellular responses to. changing environment.


Subject(s)
DNA Demethylation , Epigenesis, Genetic/physiology , Gene Expression Regulation, Developmental/physiology , Liver/growth & development , Signal Transduction/physiology , 5-Methylcytosine/metabolism , Animals , Animals, Newborn , Cells, Cultured , DNA-Binding Proteins/genetics , Dioxygenases , Female , Hepatocytes/metabolism , High-Throughput Nucleotide Sequencing , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Proto-Oncogene Proteins/genetics , Sequence Analysis, RNA
11.
Proc Natl Acad Sci U S A ; 114(51): 13525-13530, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29203669

ABSTRACT

DNA methylation at promoters is an important determinant of gene expression. Earlier studies suggested that the insulin gene promoter is uniquely unmethylated in insulin-expressing pancreatic ß-cells, providing a classic example of this paradigm. Here we show that islet cells expressing insulin, glucagon, or somatostatin share a lack of methylation at the promoters of the insulin and glucagon genes. This is achieved by rapid demethylation of the insulin and glucagon gene promoters during differentiation of Neurogenin3+ embryonic endocrine progenitors, regardless of the specific endocrine cell-type chosen. Similar methylation dynamics were observed in transgenic mice containing a human insulin promoter fragment, pointing to the responsible cis element. Whole-methylome comparison of human α- and ß-cells revealed generality of the findings: genes active in one cell type and silent in the other tend to share demethylated promoters, while methylation differences between α- and ß-cells are concentrated in enhancers. These findings suggest an epigenetic basis for the observed plastic identity of islet cell types, and have implications for ß-cell reprogramming in diabetes and diagnosis of ß-cell death using methylation patterns of circulating DNA.


Subject(s)
DNA Methylation , Enhancer Elements, Genetic , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Promoter Regions, Genetic , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Epigenesis, Genetic , Glucagon-Secreting Cells/cytology , Humans , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred ICR
12.
Nat Struct Mol Biol ; 24(12): 1132-1138, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29131141

ABSTRACT

Many regions of the genome replicate asynchronously and are expressed monoallelically. It is thought that asynchronous replication may be involved in choosing one allele over the other, but little is known about how these patterns are established during development. We show that, unlike somatic cells, which replicate in a clonal manner, embryonic and adult stem cells are programmed to undergo switching, such that daughter cells with an early-replicating paternal allele are derived from mother cells that have a late-replicating paternal allele. Furthermore, using ground-state embryonic stem (ES) cells, we demonstrate that in the initial transition to asynchronous replication, it is always the paternal allele that is chosen to replicate early, suggesting that primary allelic choice is directed by preset gametic DNA markers. Taken together, these studies help define a basic general strategy for establishing allelic discrimination and generating allelic diversity throughout the organism.


Subject(s)
Adult Stem Cells/cytology , Cell Proliferation/genetics , DNA Replication/genetics , Embryonic Stem Cells/cytology , Genomic Imprinting/genetics , Alleles , Animals , Cell Line , DNA Methylation/genetics , Genetic Markers/genetics , Mice
13.
Int J Dev Biol ; 61(3-4-5): 137-148, 2017.
Article in English | MEDLINE | ID: mdl-28621411

ABSTRACT

DNA methylation plays a prominent role in setting up and stabilizing the molecular design of gene regulation and by understanding this process one gains profound insight into the underlying biology of mammals. In this article, we trace the discoveries that provided the foundations of this field, starting with the mapping of methyl groups in the genome and the experiments that helped clarify how methylation patterns are maintained through cell division. We then address the basic relationship between methyl groups and gene repression, as well as the molecular rules involved in controlling this process during development in vivo. Finally, we describe ongoing work aimed at defining the role of this modification in disease and deciphering how it may serve as a mechanism for sensing the environment.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Developmental , Animals , Cell Division , Chromatin/metabolism , Colonic Neoplasms/metabolism , CpG Islands , Gene Expression Regulation, Neoplastic , Genome , Genomic Imprinting , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Testosterone/metabolism
14.
Int J Dev Biol ; 61(3-4-5): 285-292, 2017.
Article in English | MEDLINE | ID: mdl-28621425

ABSTRACT

Fragile X syndrome is the most frequent cause of inherited intellectual disability. The primary molecular defect in this disease is the expansion of a CGG repeat in the 5' region of the fragile X mental retardation1 (FMR1) gene, leading to de novo methylation of the promoter and inactivation of this otherwise normal gene, but little is known about how these epigenetic changes occur during development. In order to gain insight into the nature of this process, we have used cell fusion technology to recapitulate the events that occur during early embryogenesis. These experiments suggest that the naturally occurring Fragile XFMR1 5' region undergoes inactivation post implantation in a Dicer/Ago-dependent targeted process which involves local SUV39H-mediated tri-methylation of histone H3K9. It thus appears that Fragile X syndrome may come about through inadvertent siRNA-mediated heterochromatinization.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Gene Expression Regulation, Developmental , 5' Untranslated Regions , Animals , Cell Differentiation , Embryonic Development , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Heterochromatin/chemistry , Histones/metabolism , Humans , Mice , Nerve Tissue Proteins/genetics , Phenotype , Promoter Regions, Genetic , RNA/metabolism , RNA Interference , RNA, Small Interfering/metabolism
15.
Nat Commun ; 8: 15575, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28555639

ABSTRACT

Although much has been done to understand how rearrangement of the Igκ locus is regulated during B-cell development, little is known about the way the variable (V) segments themselves are selected. Here we show, using B6/Cast hybrid pre-B-cell clones, that a limited number of V segments on each allele is stochastically activated as characterized by the appearance of non-coding RNA and histone modifications. The activation states are clonally distinct, stable across cell division and developmentally important in directing the Ig repertoire upon differentiation. Using a new approach of allelic ATAC-seq, we demonstrate that the Igκ V alleles have differential chromatin accessibility, which may serve as the underlying basis of clonal maintenance at this locus, as well as other instances of monoallelic expression throughout the genome. These findings highlight a new level of immune system regulation that optimizes gene diversity.


Subject(s)
Alleles , Chromatin/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin kappa-Chains/metabolism , Animals , Antibodies/immunology , Female , Genetic Variation , Histones/metabolism , Immune System , Mice , Mice, Inbred C57BL , Precursor Cells, B-Lymphoid/immunology , RNA, Untranslated/genetics , Transcription, Genetic
16.
Proc Natl Acad Sci U S A ; 114(9): 2230-2234, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193856

ABSTRACT

Recently, it was suggested that tissue variation in cancer risk originates from differences in the number of stem-cell divisions underlying each tissue, leading to different mutation loads. We show that this variation is also correlated with the degree of aberrant CpG island DNA methylation in normal cells. Methylation accumulates during aging in a subset of molecules, suggesting that the epigenetic landscape within a founder-cell population may contribute to tumor formation.


Subject(s)
Aging/genetics , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Models, Statistical , Neoplasms/genetics , Stem Cells/metabolism , Aging/metabolism , Cell Differentiation , Cell Division , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , CpG Islands , DNA Methylation , Female , Genetic Predisposition to Disease , Humans , Male , Models, Genetic , Morphogenesis/genetics , Mutation , Neoplasms/classification , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity , Risk , Stem Cells/cytology
17.
Cancer Res ; 76(12): 3446-50, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27256564

ABSTRACT

DNA methylation is known to be abnormal in all forms of cancer, but it is not really understood how this occurs and what is its role in tumorigenesis. In this review, we take a wide view of this problem by analyzing the strategies involved in setting up normal DNA methylation patterns and understanding how this stable epigenetic mark works to prevent gene activation during development. Aberrant DNA methylation in cancer can be generated either prior to or following cell transformation through mutations. Increasing evidence suggests, however, that most methylation changes are generated in a programmed manner and occur in a subpopulation of tissue cells during normal aging, probably predisposing them for tumorigenesis. It is likely that this methylation contributes to the tumor state by inhibiting the plasticity of cell differentiation processes. Cancer Res; 76(12); 3446-50. ©2016 AACR.


Subject(s)
Aging , DNA Methylation , Neoplasms/genetics , Carcinogenesis , Cell Differentiation , CpG Islands , Humans , Neoplasms/etiology
18.
Article in English | MEDLINE | ID: mdl-27141050

ABSTRACT

The genome is subject to a diverse array of epigenetic modifications from DNA methylation to histone posttranslational changes. Many of these marks are somatically stable through cell division. This article focuses on our knowledge of the mechanisms governing the inheritance of epigenetic marks, particularly, repressive ones, when the DNA and chromatin template are duplicated in S phase. This involves the action of histone chaperones, nucleosome-remodeling enzymes, histone and DNA methylation binding proteins, and chromatin-modifying enzymes. Last, the timing of DNA replication is discussed, including the question of whether this constitutes an epigenetic mark that facilitates the propagation of epigenetic marks.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Models, Genetic , DNA Methylation , DNA Replication/physiology , Histone Chaperones/physiology , Histone Code , Protein Processing, Post-Translational
19.
Proc Natl Acad Sci U S A ; 113(18): 5018-23, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27091986

ABSTRACT

There is ample evidence that somatic cell differentiation during development is accompanied by extensive DNA demethylation of specific sites that vary between cell types. Although the mechanism of this process has not yet been elucidated, it is likely to involve the conversion of 5mC to 5hmC by Tet enzymes. We show that a Tet2/Tet3 conditional knockout at early stages of B-cell development largely prevents lineage-specific programmed demethylation events. This lack of demethylation affects the expression of nearby B-cell lineage genes by impairing enhancer activity, thus causing defects in B-cell differentiation and function. Thus, tissue-specific DNA demethylation appears to be necessary for proper somatic cell development in vivo.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/physiology , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Epigenesis, Genetic/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Mice , Mice, Inbred C57BL , Organ Specificity/genetics
20.
Proc Natl Acad Sci U S A ; 113(13): E1826-34, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976580

ABSTRACT

Minimally invasive detection of cell death could prove an invaluable resource in many physiologic and pathologic situations. Cell-free circulating DNA (cfDNA) released from dying cells is emerging as a diagnostic tool for monitoring cancer dynamics and graft failure. However, existing methods rely on differences in DNA sequences in source tissues, so that cell death cannot be identified in tissues with a normal genome. We developed a method of detecting tissue-specific cell death in humans based on tissue-specific methylation patterns in cfDNA. We interrogated tissue-specific methylome databases to identify cell type-specific DNA methylation signatures and developed a method to detect these signatures in mixed DNA samples. We isolated cfDNA from plasma or serum of donors, treated the cfDNA with bisulfite, PCR-amplified the cfDNA, and sequenced it to quantify cfDNA carrying the methylation markers of the cell type of interest. Pancreatic ß-cell DNA was identified in the circulation of patients with recently diagnosed type-1 diabetes and islet-graft recipients; oligodendrocyte DNA was identified in patients with relapsing multiple sclerosis; neuronal/glial DNA was identified in patients after traumatic brain injury or cardiac arrest; and exocrine pancreas DNA was identified in patients with pancreatic cancer or pancreatitis. This proof-of-concept study demonstrates that the tissue origins of cfDNA and thus the rate of death of specific cell types can be determined in humans. The approach can be adapted to identify cfDNA derived from any cell type in the body, offering a minimally invasive window for diagnosing and monitoring a broad spectrum of human pathologies as well as providing a better understanding of normal tissue dynamics.


Subject(s)
DNA Methylation , DNA/blood , Insulin-Secreting Cells/pathology , Oligodendroglia/pathology , Adolescent , Adult , Aged , Brain Ischemia/genetics , Brain Ischemia/pathology , Case-Control Studies , Cell Death , Child , Child, Preschool , DNA/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Genetic Markers , Humans , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/pathology , Organ Specificity , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/pathology , Promoter Regions, Genetic , Sensitivity and Specificity , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...