Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nat Commun ; 15(1): 3937, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38729924

RESUMO

Human natural killer (NK) cell-based therapies are under assessment for treating various cancers, but cryopreservation reduces both the recovery and function of NK cells, thereby limiting their therapeutic feasibility. Using cryopreservation protocols optimized for T cells, here we find that ~75% of NK cells die within 24 h post-thaw, with the remaining cells displaying reduced cytotoxicity. Using CRISPR-Cas9 gene editing and confocal microscopy, we find that cryopreserved NK cells largely die via apoptosis initiated by leakage of granzyme B from cytotoxic vesicles. Pretreatment of NK cells with a combination of Interleukins-15 (IL-15) and IL-18 prior to cryopreservation improves NK cell recovery to ~90-100% and enables equal tumour control in a xenograft model of disseminated Raji cell lymphoma compared to non-cryopreserved NK cells. The mechanism of IL-15 and IL-18-induced protection incorporates two mechanisms: a transient reduction in intracellular granzyme B levels via degranulation, and the induction of antiapoptotic genes.


Assuntos
Apoptose , Criopreservação , Granzimas , Interleucina-15 , Interleucina-18 , Células Matadoras Naturais , Granzimas/metabolismo , Interleucina-15/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Humanos , Interleucina-18/metabolismo , Animais , Criopreservação/métodos , Camundongos , Linhagem Celular Tumoral , Sistemas CRISPR-Cas
2.
Cell ; 186(15): 3166-3181.e18, 2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37413989

RESUMO

Proper preimplantation development is essential to assemble a blastocyst capable of implantation. Live imaging has uncovered major events driving early development in mouse embryos; yet, studies in humans have been limited by restrictions on genetic manipulation and lack of imaging approaches. We have overcome this barrier by combining fluorescent dyes with live imaging to reveal the dynamics of chromosome segregation, compaction, polarization, blastocyst formation, and hatching in the human embryo. We also show that blastocyst expansion mechanically constrains trophectoderm cells, causing nuclear budding and DNA shedding into the cytoplasm. Furthermore, cells with lower perinuclear keratin levels are more prone to undergo DNA loss. Moreover, applying trophectoderm biopsy, a mechanical procedure performed clinically for genetic testing, increases DNA shedding. Thus, our work reveals distinct processes underlying human development compared with mouse and suggests that aneuploidies in human embryos may not only originate from chromosome segregation errors during mitosis but also from nuclear DNA shedding.


Assuntos
Diagnóstico Pré-Implantação , Gravidez , Feminino , Humanos , Animais , Camundongos , Diagnóstico Pré-Implantação/métodos , Blastocisto , Implantação do Embrião , Testes Genéticos/métodos , Aneuploidia , Biópsia/métodos
3.
Cell ; 186(18): 3776-3792.e16, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37478861

RESUMO

In vitro stem cell models that replicate human gastrulation have been generated, but they lack the essential extraembryonic cells needed for embryonic development, morphogenesis, and patterning. Here, we describe a robust and efficient method that prompts human extended pluripotent stem cells to self-organize into embryo-like structures, termed peri-gastruloids, which encompass both embryonic (epiblast) and extraembryonic (hypoblast) tissues. Although peri-gastruloids are not viable due to the exclusion of trophoblasts, they recapitulate critical stages of human peri-gastrulation development, such as forming amniotic and yolk sac cavities, developing bilaminar and trilaminar embryonic discs, specifying primordial germ cells, initiating gastrulation, and undergoing early neurulation and organogenesis. Single-cell RNA-sequencing unveiled transcriptomic similarities between advanced human peri-gastruloids and primary peri-gastrulation cell types found in humans and non-human primates. This peri-gastruloid platform allows for further exploration beyond gastrulation and may potentially aid in the development of human fetal tissues for use in regenerative medicine.


Assuntos
Implantação do Embrião , Gastrulação , Células-Tronco Pluripotentes , Animais , Feminino , Humanos , Gravidez , Diferenciação Celular , Embrião de Mamíferos , Desenvolvimento Embrionário , Organogênese , Células-Tronco Pluripotentes/metabolismo , Primatas
4.
Dev Cell ; 58(12): 1005-1006, 2023 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-37339602

RESUMO

In this issue of Developmental Cell, Arekatla et al. use optogenetic technologies to dissect the roles of ERK and AKT dynamics in pluripotency. They show how mouse embryonic stem cells can retain memory of signaling events controlling their fate.


Assuntos
Optogenética , Transdução de Sinais , Animais , Camundongos , Diferenciação Celular , Células-Tronco Embrionárias Murinas
5.
Fertil Steril ; 120(3 Pt 1): 467-472, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37150393

RESUMO

Preimplantation development is the only stage of human development that can be studied outside the body in real time, as human embryos can be produced by in vitro fertilization and cultured in the laboratory as self-contained structures until the blastocyst stage. Here, we focus some of the key cellular and morphogenetic processes by which the 1-cell embryo is transformed gradually into a blastocyst ready for implantation. Although most of our knowledge about the dynamic series of events patterning preimplantation human development derives from work in mouse embryos, we discuss key differences that could exist with humans. Furthermore, we highlight how new approaches may enable to reveal many of the unknown processes driving human preimplantation development, particularly using noninvasive imaging and genetic technologies.


Assuntos
Blastocisto , Implantação do Embrião , Humanos , Animais , Camundongos , Fertilização in vitro , Desenvolvimento Embrionário , Embrião de Mamíferos
6.
Cell ; 186(10): 2078-2091.e18, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37172562

RESUMO

Neural tube (NT) defects arise from abnormal neurulation and result in the most common birth defects worldwide. Yet, mechanisms of primate neurulation remain largely unknown due to prohibitions on human embryo research and limitations of available model systems. Here, we establish a three-dimensional (3D) prolonged in vitro culture (pIVC) system supporting cynomolgus monkey embryo development from 7 to 25 days post-fertilization. Through single-cell multi-omics analyses, we demonstrate that pIVC embryos form three germ layers, including primordial germ cells, and establish proper DNA methylation and chromatin accessibility through advanced gastrulation stages. In addition, pIVC embryo immunofluorescence confirms neural crest formation, NT closure, and neural progenitor regionalization. Finally, we demonstrate that the transcriptional profiles and morphogenetics of pIVC embryos resemble key features of similarly staged in vivo cynomolgus and human embryos. This work therefore describes a system to study non-human primate embryogenesis through advanced gastrulation and early neurulation.


Assuntos
Defeitos do Tubo Neural , Neurulação , Técnicas de Cultura de Tecidos , Animais , Humanos , Blastocisto , Embrião de Mamíferos , Desenvolvimento Embrionário , Macaca fascicularis , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/patologia , Técnicas de Cultura de Tecidos/métodos
7.
Nat Commun ; 14(1): 3101, 2023 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-37248263

RESUMO

During preimplantation development, contractile forces generated at the apical cortex segregate cells into inner and outer positions of the embryo, establishing the inner cell mass (ICM) and trophectoderm. To which extent these forces influence ICM-trophectoderm fate remains unresolved. Here, we found that the nuclear lamina is coupled to the cortex via an F-actin meshwork in mouse and human embryos. Actomyosin contractility increases during development, upregulating Lamin-A levels, but upon internalization cells lose their apical cortex and downregulate Lamin-A. Low Lamin-A shifts the localization of actin nucleators from nucleus to cytoplasm increasing cytoplasmic F-actin abundance. This results in stabilization of Amot, Yap phosphorylation and acquisition of ICM over trophectoderm fate. By contrast, in outer cells, Lamin-A levels increase with contractility. This prevents Yap phosphorylation enabling Cdx2 to specify the trophectoderm. Thus, forces transmitted to the nuclear lamina control actin organization to differentially regulate the factors specifying lineage identity.


Assuntos
Actinas , Proteínas Adaptadoras de Transdução de Sinal , Humanos , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Lâmina Nuclear/metabolismo , Proteínas de Ciclo Celular , Proteínas de Sinalização YAP , Blastocisto/metabolismo , Laminas
8.
J Anat ; 242(3): 417-435, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36423208

RESUMO

Somites are transient structures derived from the pre-somitic mesoderm (PSM), involving mesenchyme-to-epithelial transition (MET) where the cells change their shape and polarize. Using Scanning electron microscopy (SEM), immunocytochemistry and confocal microscopy, we study the progression of these events along the tail-to-head axis of the embryo, which mirrors the progression of somitogenesis (younger cells located more caudally). SEM revealed that PSM epithelialization is a gradual process, which begins much earlier than previously thought, starting with the dorsalmost cells, then the medial ones, and then, simultaneously, the ventral and lateral cells, before a somite fully separates from the PSM. The core (internal) cells of the PSM and somites never epithelialize, which suggests that the core cells could be 'trapped' within the somitocoele after cells at the surfaces of the PSM undergo MET. Three-dimensional imaging of the distribution of the cell polarity markers PKCζ, PAR3, ZO1, the Golgi marker GM130 and the apical marker N-cadherin reveal that the pattern of polarization is distinctive for each marker and for each surface of the PSM, but the order of these events is not the same as the progression of cell elongation. These observations challenge some assumptions underlying existing models of somite formation.


Assuntos
Mesoderma , Somitos , Morfogênese , Caderinas/metabolismo , Desenvolvimento Embrionário
9.
Stem Cell Reports ; 17(9): 1991-2004, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-35961310

RESUMO

IL-6 has been shown to be required for somatic cell reprogramming into induced pluripotent stem cells (iPSCs). However, how Il6 expression is regulated and whether it plays a role during embryo development remains unknown. Here, we describe that IL-6 is necessary for C/EBPα-enhanced reprogramming of B cells into iPSCs but not for B cell to macrophage transdifferentiation. C/EBPα overexpression activates both Il6 and Il6ra genes in B cells and in PSCs. In embryo development, Cebpa is enriched in the trophectoderm of blastocysts together with Il6, while Il6ra is mostly expressed in the inner cell mass (ICM). In addition, Il6 expression in blastocysts requires Cebpa. Blastocysts secrete IL-6 and neutralization of the cytokine delays the morula to blastocyst transition. The observed requirement of C/EBPα-regulated IL-6 signaling for pluripotency during somatic cell reprogramming thus recapitulates a physiologic mechanism in which the trophectoderm acts as niche for the ICM through the secretion of IL-6.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT , Interleucina-6 , Blastocisto , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Desenvolvimento Embrionário , Interleucina-6/metabolismo , Mórula/metabolismo
10.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35101917

RESUMO

In warm-blooded vertebrate embryos (mammals and birds), the axial tissues of the body form from a growth zone at the tail end, Hensen's node, which generates neural, mesodermal, and endodermal structures along the midline. While most cells only pass through this region, the node has been suggested to contain a small population of resident stem cells. However, it is unknown whether the rest of the node constitutes an instructive niche that specifies this self-renewal behavior. Here, we use heterotopic transplantation of groups and single cells and show that cells not destined to enter the node can become resident and self-renew. Long-term resident cells are restricted to the posterior part of the node and single-cell RNA-sequencing reveals that the majority of these resident cells preferentially express G2/M phase cell-cycle-related genes. These results provide strong evidence that the node functions as a niche to maintain self-renewal of axial progenitors.


Assuntos
Padronização Corporal/fisiologia , Organizadores Embrionários/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Embrião de Galinha , Endoderma/embriologia , Gástrula/embriologia , Mesoderma/embriologia , Sistema Nervoso , Notocorda/embriologia , Organizadores Embrionários/metabolismo , Nicho de Células-Tronco/genética , Células-Tronco/metabolismo , Células-Tronco/fisiologia
11.
Nat Cell Biol ; 24(2): 155-167, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35102267

RESUMO

During mammalian development, the first asymmetric cell divisions segregate cells into inner and outer positions of the embryo to establish the pluripotent and trophectoderm lineages. Typically, polarity components differentially regulate the mitotic spindle via astral microtubule arrays to trigger asymmetric division patterns. However, early mouse embryos lack centrosomes, the microtubule-organizing centres (MTOCs) that usually generate microtubule asters. Thus, it remains unknown whether spindle organization regulates lineage segregation. Here we find that heterogeneities in cell polarity in the early 8-cell-stage mouse embryo trigger the assembly of a highly asymmetric spindle organization. This spindle arises in an unusual modular manner, forming a single microtubule aster from an apically localized, non-centrosomal MTOC, before joining it to the rest of the spindle apparatus. When fully assembled, this 'monoastral' spindle triggers spatially asymmetric division patterns to segregate cells into inner and outer positions. Moreover, the asymmetric inheritance of spindle components causes differential cell polarization to determine pluripotent versus trophectoderm lineage fate.


Assuntos
Diferenciação Celular , Divisão Celular , Linhagem da Célula , Polaridade Celular , Embrião de Mamíferos/fisiologia , Fuso Acromático/fisiologia , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Fuso Acromático/genética , Fuso Acromático/metabolismo
12.
Nat Rev Mol Cell Biol ; 22(8): 548-562, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33927361

RESUMO

The cytoskeleton - comprising actin filaments, microtubules and intermediate filaments - serves instructive roles in regulating cell function and behaviour during development. However, a key challenge in cell and developmental biology is to dissect how these different structures function and interact in vivo to build complex tissues, with the ultimate aim to understand these processes in a mammalian organism. The preimplantation mouse embryo has emerged as a primary model system for tackling this challenge. Not only does the mouse embryo share many morphological similarities with the human embryo during its initial stages of life, it also permits the combination of genetic manipulations with live-imaging approaches to study cytoskeletal dynamics directly within an intact embryonic system. These advantages have led to the discovery of novel cytoskeletal structures and mechanisms controlling lineage specification, cell-cell communication and the establishment of the first forms of tissue architecture during development. Here we highlight the diverse organization and functions of each of the three cytoskeletal filaments during the key events that shape the early mammalian embryo, and discuss how they work together to perform key developmental tasks, including cell fate specification and morphogenesis of the blastocyst. Collectively, these findings are unveiling a new picture of how cells in the early embryo dynamically remodel their cytoskeleton with unique spatial and temporal precision to drive developmental processes in the rapidly changing in vivo environment.


Assuntos
Citoesqueleto/fisiologia , Desenvolvimento Embrionário/fisiologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Comunicação Celular , Divisão Celular , Linhagem da Célula , Embrião de Mamíferos , Humanos , Morfogênese
15.
Nature ; 585(7825): 404-409, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32848249

RESUMO

To implant in the uterus, the mammalian embryo first specifies two cell lineages: the pluripotent inner cell mass that forms the fetus, and the outer trophectoderm layer that forms the placenta1. In many organisms, asymmetrically inherited fate determinants drive lineage specification2, but this is not thought to be the case during early mammalian development. Here we show that intermediate filaments assembled by keratins function as asymmetrically inherited fate determinants in the mammalian embryo. Unlike F-actin or microtubules, keratins are the first major components of the cytoskeleton that display prominent cell-to-cell variability, triggered by heterogeneities in the BAF chromatin-remodelling complex. Live-embryo imaging shows that keratins become asymmetrically inherited by outer daughter cells during cell division, where they stabilize the cortex to promote apical polarization and YAP-dependent expression of CDX2, thereby specifying the first trophectoderm cells of the embryo. Together, our data reveal a mechanism by which cell-to-cell heterogeneities that appear before the segregation of the trophectoderm and the inner cell mass influence lineage fate, via differential keratin regulation, and identify an early function for intermediate filaments in development.


Assuntos
Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Queratinas/metabolismo , Actinas/metabolismo , Animais , Divisão Celular , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/metabolismo , Ectoderma/citologia , Embrião de Mamíferos/embriologia , Feminino , Humanos , Filamentos Intermediários/metabolismo , Camundongos , Microtúbulos/metabolismo , Complexos Multiproteicos/metabolismo , Trofoblastos/citologia
16.
Curr Opin Cell Biol ; 62: 78-85, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31731147

RESUMO

Understanding the development of apicobasal polarity (ABP) is a long-standing problem in biology. The molecular components involved in the development and maintenance of APB have been largely identified and are known to have ubiquitous roles across organisms. Our knowledge of the functional consequences of ABP establishment and maintenance is far less comprehensive. Recent studies using novel experimental approaches and cellular models have revealed a growing link between ABP and the genetic program of cell lineage. This mini-review describes some of the most recent advances in this new field, highlighting examples from Caenorhabditis elegans and mouse embryos, human pluripotent stem cells, and epithelial cells. We also speculate on the most interesting and challenging avenues that can be explored.


Assuntos
Polaridade Celular/fisiologia , Células Epiteliais/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos
17.
Artigo em Inglês | MEDLINE | ID: mdl-31615786

RESUMO

Our understanding of how the first mammalian cell lineages arise has been shaped largely by studies of the preimplantation mouse embryo. Painstaking work over many decades has begun to reveal how a single totipotent cell is transformed into a multilayered structure representing the foundations of the body plan. Here, we review how the first lineage decision is initiated by epigenetic regulation but consolidated by the integration of morphological features and transcription factor activity. The establishment of pluripotent and multipotent stem cell lines has enabled deeper analysis of molecular and epigenetic regulation of cell fate decisions. The capability to assemble these stem cells into artificial embryos is an exciting new avenue of research that offers a long-awaited window into cell fate specification in the human embryo. Together, these approaches are poised to profoundly increase our understanding of how the first lineage decisions are made during mammalian embryonic development.


Assuntos
Linhagem da Célula , Epigênese Genética , Células-Tronco Multipotentes/citologia , Animais , Blastocisto/citologia , Diferenciação Celular , Linhagem Celular , Cromatina/metabolismo , Metilação de DNA , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Histonas/química , Humanos , Técnicas In Vitro , Camundongos , Gravidez , Retroelementos , Células-Tronco/citologia
18.
Dev Cell ; 45(6): 667-679, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29920273

RESUMO

The preimplantation mouse embryo is a simple self-contained system, making it an excellent model to discover how mammalian cells function in real time and in vivo. Work over the last decade has revealed some key morphogenetic mechanisms that drive early development, yielding rudimentary instructions for the generation of a mammalian embryo. Here, we review the instructions revealed thus far, and then discuss remaining challenges to discover upstream factors controlling cell fate determination, test the role of mechanisms based on biological noise, and take advantage of recent technological developments to advance the spatial and temporal resolution of our current understanding.


Assuntos
Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/fisiologia , Camundongos/embriologia , Animais , Blastocisto/fisiologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Polaridade Celular/fisiologia , Embrião de Mamíferos/fisiologia , Imageamento Tridimensional/métodos , Modelos Biológicos , Morfogênese/fisiologia
19.
Curr Biol ; 28(10): 1651-1660.e4, 2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29754899

RESUMO

The Hippo pathway is an evolutionarily conserved signaling network that integrates diverse cues to control organ size and cell fate. The central downstream pathway protein in Drosophila is the transcriptional co-activator Yorkie (YAP and TAZ in humans), which regulates gene expression with the Scalloped/TEA domain family member (TEAD) transcription factors [1-8]. A central regulatory step in the Hippo pathway is phosphorylation of Yorkie by the NDR family kinase Warts, which promotes Yorkie cytoplasmic localization by stimulating association with 14-3-3 proteins [9-12]. Numerous reports have purported a static model of Hippo signaling whereby, upon Hippo activation, Yorkie/YAP/TAZ become cytoplasmic and therefore inactive, and upon Hippo repression, Yorkie/YAP/TAZ transit to the nucleus and are active. However, we have little appreciation for the dynamics of Yorkie/YAP/TAZ subcellular localization because most studies have been performed in fixed cells and tissues. To address this, we used live multiphoton microscopy to investigate the dynamics of an endogenously tagged Yorkie-Venus protein in growing epithelial organs. We found that the majority of Yorkie rapidly traffics between the cytoplasm and nucleus, rather than being statically localized in either compartment. In addition, discrete cell populations within the same organ display different rates of Yorkie nucleo-cytoplasmic shuttling. By assessing Yorkie dynamics in warts mutant tissue, we found that the Hippo pathway regulates Yorkie subcellular distribution by regulating its rate of nuclear import. Furthermore, Yorkie's localization fluctuates dramatically throughout the cell cycle, being predominantly cytoplasmic during interphase and, unexpectedly, chromatin enriched during mitosis. Yorkie's association with mitotic chromatin is Scalloped dependent, suggesting a potential role in mitotic bookmarking.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Animais , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Microscopia de Fluorescência por Excitação Multifotônica , Transporte Proteico , Proteínas de Sinalização YAP
20.
Cell ; 173(3): 776-791.e17, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29576449

RESUMO

Transformation from morula to blastocyst is a defining event of preimplantation embryo development. During this transition, the embryo must establish a paracellular permeability barrier to enable expansion of the blastocyst cavity. Here, using live imaging of mouse embryos, we reveal an actin-zippering mechanism driving this embryo sealing. Preceding blastocyst stage, a cortical F-actin ring assembles at the apical pole of the embryo's outer cells. The ring structure forms when cortical actin flows encounter a network of polar microtubules that exclude F-actin. Unlike stereotypical actin rings, the actin rings of the mouse embryo are not contractile, but instead, they expand to the cell-cell junctions. Here, they couple to the junctions by recruiting and stabilizing adherens and tight junction components. Coupling of the actin rings triggers localized myosin II accumulation, and it initiates a tension-dependent zippering mechanism along the junctions that is required to seal the embryo for blastocyst formation.


Assuntos
Actinas/química , Blastocisto/metabolismo , Microtúbulos/metabolismo , Miosina Tipo II/química , Animais , Comunicação Celular , Proteínas do Citoesqueleto/química , Embrião de Mamíferos , Desenvolvimento Embrionário , Feminino , Proteínas de Fluorescência Verde , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos C57BL , Mórula , RNA Interferente Pequeno/metabolismo , Junções Íntimas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA