Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 380
Filtrer
1.
EMBO Rep ; 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39256596

RÉSUMÉ

The embryonic cell surface is rich in glycosphingolipids (GSLs), which change during differentiation. The reasons for GSL subgroup variation during early embryogenesis remain elusive. By combining genomic approaches, flow cytometry, confocal imaging, and transcriptomic data analysis, we discovered that α1,2-fucosylated GSLs control the differentiation of human pluripotent cells (hPCs) into germ layer tissues. Overexpression of α1,2-fucosylated GSLs disrupts hPC differentiation into mesodermal lineage and reduces differentiation into cardiomyocytes. Conversely, reducing α1,2-fucosylated groups promotes hPC differentiation and mesoderm commitment in response to external signals. We find that bone morphogenetic protein 4 (BMP4), a mesodermal gene inducer, suppresses α1,2-fucosylated GSL expression. Overexpression of α1,2-fucosylated GSLs impairs SMAD activation despite BMP4 presence, suggesting α-fucosyl end groups as BMP pathway regulators. Additionally, the absence of α1,2-fucosylated GSLs in early/late mesoderm and primitive streak stages in mouse embryos aligns with the hPC results. Thus, α1,2-fucosylated GSLs may regulate early cell-fate decisions and embryo development by modulating cell signaling.

2.
eNeuro ; 11(8)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39137988

RÉSUMÉ

Sensory axons projecting to the central nervous system are organized into topographic maps that represent the locations of sensory stimuli. In some sensory systems, even adjacent sensory axons are arranged topographically, forming "fine-scale" topographic maps. Although several broad molecular gradients are known to instruct coarse topography, we know little about the molecular signaling that regulates fine-scale topography at the level of two adjacent axons. Here, we provide evidence that transsynaptic bone morphogenetic protein (BMP) signaling mediates local interneuronal communication to regulate fine-scale topography in the nociceptive system of Drosophila larvae. We first show that the topographic separation of the axon terminals of adjacent nociceptors requires their common postsynaptic target, the A08n neurons. This phenotype is recapitulated by knockdown of the BMP ligand, Decapentaplegic (Dpp), in these neurons. In addition, removing the Type 2 BMP receptors or their effector (Mad transcription factor) in single nociceptors impairs the fine-scale topography, suggesting the contribution of BMP signaling originated from A08n. This signaling is likely mediated by phospho-Mad in the presynaptic terminals of nociceptors to ensure local interneuronal communication. Finally, reducing Dpp levels in A08n reduces the nociceptor-A08n synaptic contacts. Our data support that transsynaptic BMP signaling establishes the fine-scale topography by facilitating the formation of topographically correct synapses. Local BMP signaling for synapse formation may be a developmental strategy that independently regulates neighboring axon terminals for fine-scale topography.


Sujet(s)
Protéines morphogénétiques osseuses , Protéines de Drosophila , Cellules réceptrices sensorielles , Transduction du signal , Animaux , Protéines de Drosophila/métabolisme , Protéines morphogénétiques osseuses/métabolisme , Transduction du signal/physiologie , Cellules réceptrices sensorielles/métabolisme , Cellules réceptrices sensorielles/physiologie , Drosophila , Larve , Nocicepteurs/métabolisme , Nocicepteurs/physiologie , Animal génétiquement modifié , Synapses/métabolisme , Synapses/physiologie , Terminaisons présynaptiques/métabolisme , Terminaisons présynaptiques/physiologie , Protéines de liaison à l'ADN , Facteurs de transcription
3.
Life Sci ; 355: 122969, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-39142506

RÉSUMÉ

Bone is a connective tissue that is metabolically active and serves multiple functions, including movement, structural support, and organ protection. It is comprised primarily of three types of bone cells, namely osteoblasts, osteocytes, and osteoclasts. Osteoblasts are bone-forming cells, and the differentiation of mesenchymal stem cells towards osteoblasts is regulated by several growth factors, cytokines, and hormones via various signaling pathways, including TGF-ß/BMP (transforming growth factor-beta/bone morphogenetic protein) signaling as a primary one. Non-coding RNAs (ncRNAs), such as microRNAs and long ncRNAs, play crucial roles in regulating osteoblast differentiation via the TGF-ß/BMP signaling cascade. Dysregulation of these ncRNAs leads to bone-pathological conditions such as osteoporosis, skeletal dysplasia, and osteosclerosis. This review provides a concise overview of the latest advancements in understanding the involvement of ncRNAs/TGF-ß/BMP axis in osteoblast differentiation. These findings have the potential to identify new molecular targets for early detection of bone metabolism disorders and the development of innovative therapy strategies.


Sujet(s)
Protéines morphogénétiques osseuses , Différenciation cellulaire , Ostéoblastes , ARN non traduit , Transduction du signal , Facteur de croissance transformant bêta , Ostéoblastes/métabolisme , Humains , Facteur de croissance transformant bêta/métabolisme , Animaux , Protéines morphogénétiques osseuses/métabolisme , ARN non traduit/génétique , ARN non traduit/métabolisme , Ostéogenèse
4.
Biomolecules ; 14(8)2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39199400

RÉSUMÉ

Bone morphogenetic proteins (BMPs) are a group of growth factors belonging to the transforming growth factor ß(TGF-ß) family. While initially recognized for their role in bone formation, BMPs have emerged as significant players in liver diseases. Among BMPs with various physiological activities, this comprehensive review aims to delve into the involvement of BMP9 specifically in liver diseases and provide insights into the complex BMP signaling pathway. Through an enhanced understanding of BMP9, we anticipate the discovery of new therapeutic options and potential strategies for managing liver diseases.


Sujet(s)
Facteur-2 de croissance et de différenciation , Maladies du foie , Transduction du signal , Humains , Facteur-2 de croissance et de différenciation/métabolisme , Maladies du foie/métabolisme , Animaux
5.
Biol Open ; 13(9)2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39207258

RÉSUMÉ

In developing tissues, morphogen gradients are thought to initialize gene expression patterns. However, the relationship between the dynamics of morphogen-encoded signals and gene expression decisions is largely unknown. Here we examine the dynamics of the Bone Morphogenetic Protein (BMP) pathway in Drosophila blastoderm-stage embryos. In this tissue, the BMP pathway is highly dynamic: it begins as a broad and weak signal on the dorsal half of the embryo, then 20-30 min later refines into a narrow, intense peak centered on the dorsal midline. This dynamical progression of the BMP signal raises questions of how it stably activates target genes. Therefore, we performed live imaging of the BMP signal and found that dorsal-lateral cells experience only a short transient in BMP signaling, after which the signal is lost completely. Moreover, we measured the transcriptional response of the BMP target gene pannier in live embryos and found it to remain activated in dorsal-lateral cells, even after the BMP signal is lost. Our findings may suggest that the BMP pathway activates a memory, or 'ratchet' mechanism that may sustain gene expression.


Sujet(s)
Protéines morphogénétiques osseuses , Régulation de l'expression des gènes au cours du développement , Transduction du signal , Animaux , Protéines morphogénétiques osseuses/métabolisme , Protéines morphogénétiques osseuses/génétique , Drosophila/embryologie , Drosophila/génétique , Embryon non mammalien/métabolisme , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme
6.
Sci Total Environ ; 948: 174772, 2024 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-39019263

RÉSUMÉ

Mounting evidence in animal experiments proves that early life stage exposure to organophosphate flame retardants (OPFRs) affects the locomotor behavior and changes the transcriptions of central nervous system genes. Unfortunately, their effect on human motor neuron (MN) development, which is necessary for body locomotion and survival, has not yet characterized. Here, we utilized a spinal cord MN differentiation model from human embryonic stem cells (ESCs) and adopted this model to test the effects of two typical OPFRs tris (2-butoxyethyl) phosphate (TBEP) and tris (2-chloroethyl) phosphate (TCEP), on MN development and the possible mechanisms underlying. Our findings revealed TBEP exerted a much more inhibitory effect on MN survival, while TCEP exhibited a stronger stimulatory effect on ESCs differentiation into MN, and thus TBEP exhibited a stronger inhibition on MN development than TCEP. RNA sequencing analysis identified TBEP and TCEP inhibited MN survival mainly by disrupting extracellular matrix (ECM)-receptor interaction. Focusing on the pathway guided MN differentiation, we found both TBEP and TCEP activated BMP signaling, whereas TCEP simultaneously downregulated Wnt signaling. Collectively, this is the first study demonstrated TBEP and TCEP disrupted human MN development by affecting their survival and differentiation, thereby raising concern about their potential harm in causing MN disorders.


Sujet(s)
Différenciation cellulaire , Ignifuges , Motoneurones , Organophosphates , Ignifuges/toxicité , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Organophosphates/toxicité , Motoneurones/effets des médicaments et des substances chimiques , Composés organiques du phosphore/toxicité , Survie cellulaire/effets des médicaments et des substances chimiques
7.
Angiogenesis ; 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38955953

RÉSUMÉ

The proliferation of the endothelium is a highly coordinated process to ensure the emergence, expansion, and homeostasis of the vasculature. While Bone Morphogenetic Protein (BMP) signaling fine-tunes the behaviors of endothelium in health and disease, how BMP signaling influences the proliferation of endothelium and therefore, modulates angiogenesis remains largely unknown. Here, we evaluated the role of Activin A Type I Receptor (ACVR1/ALK2), a key BMP receptor in the endothelium, in modulating the proliferation of endothelial cells. We show that ACVR1/ALK2 is a key modulator for the proliferation of endothelium in the retinal vessels. Loss of endothelial ALK2 leads to a significant reduction in endothelial proliferation and results in fewer branches/endothelial cells in the retinal vessels. Interestingly, venous endothelium appears to be more susceptible to ALK2 deletion. Mechanistically, ACVR1/ALK2 inhibits the expression of CDKN1A/p21, a critical negative regulator of cell cycle progression, in a SMAD1/5-dependent manner, thereby enabling the venous endothelium to undergo active proliferation by suppressing CDKN1A/p21. Taken together, our findings show that BMP signaling mediated by ACVR1/ALK2 provides a critical yet previously underappreciated input to modulate the proliferation of venous endothelium, thereby fine-tuning the context of angiogenesis in health and disease.

8.
Sci China Life Sci ; 67(9): 1915-1927, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38913236

RÉSUMÉ

The BMP signaling pathway plays a crucial role in regulating early embryonic development and tissue homeostasis. SMAD6 encodes a negative regulator of BMP, and rare variants of SMAD6 are recurrently found in individuals with birth defects. However, we observed that a subset of rare pathogenic variants of SMAD6 consistently exhibited positive regulatory effects instead of the initial negative effects on the BMP signaling pathway. We sought to determine whether these SMAD6 variants have common pathogenic mechanisms. Here, we showed that pathogenic SMAD6 variants accompanying this functional reversal exhibit similar increases in deamidation. Mechanistically, increased deamidation of SMAD6 variants promotes the accumulation of the BMP receptor BMPR1A and the formation of new complexes, both of which lead to BMP signaling pathway activation. Specifically, two residues, N262 and N404, in SMAD6 were identified as the crucial sites of deamidation, which was catalyzed primarily by glutamine-fructose-6-phosphate transaminase 2 (GFPT2). Additionally, treatment of cells harboring SMAD6 variants with a deamidase inhibitor restored the inhibitory effect of SMAD6 on the BMP signaling pathway. Conversely, when wild-type SMAD6 was manually simulated to mimic the deamidated state, the reversed function of activating BMP signaling was reproduced. Taken together, these findings show that deamidation of SMAD6 plays a crucial role in the functional reversal of BMP signaling activity, which can be induced by a subset of various SMAD6 variants. Our study reveals a common pathogenic mechanism shared by these variants and provides a potential strategy for preventing birth defects through deamidation regulation, which might prevent the off-target effects of gene editing.


Sujet(s)
Transduction du signal , Protéine Smad6 , Humains , Protéine Smad6/métabolisme , Protéine Smad6/génétique , Cellules HEK293 , Récepteurs de la protéine morphogénique osseuse de type I/métabolisme , Récepteurs de la protéine morphogénique osseuse de type I/génétique , Protéines morphogénétiques osseuses/métabolisme , Protéines morphogénétiques osseuses/génétique , Mutation
9.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891830

RÉSUMÉ

The dynamic process of Drosophila spermatogenesis involves asymmetric division, mitosis, and meiosis, which ultimately results in the production of mature spermatozoa. Disorders of spermatogenesis can lead to infertility in males. ADAR (adenosine deaminase acting on RNA) mutations in Drosophila cause male infertility, yet the causative factors remain unclear. In this study, immunofluorescence staining was employed to visualize endogenous ADAR proteins and assess protein levels via fluorescence-intensity analysis. In addition, the early differentiation disorders and homeostatic alterations during early spermatogenesis in the testes were examined through quantification of transit-amplifying region length, counting the number of GSCs (germline stem cells), and fertility experiments. Our findings suggest that deletion of ADAR causes testicular tip transit-amplifying cells to accumulate and become infertile in older male Drosophila. By overexpressing ADAR in early germline cells, male infertility can be partially rescued. Transcriptome analysis showed that ADAR maintained early spermatogenesis homeostasis through the bone-morphogenetic-protein (BMP) signaling pathway. Taken together, these findings have the potential to help explore the role of ADAR in early spermatogenesis.


Sujet(s)
Adenosine deaminase , Protéines morphogénétiques osseuses , Protéines de Drosophila , Drosophila melanogaster , Transduction du signal , Spermatogenèse , Animaux , Mâle , Spermatogenèse/génétique , Drosophila melanogaster/génétique , Drosophila melanogaster/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Adenosine deaminase/métabolisme , Adenosine deaminase/génétique , Protéines morphogénétiques osseuses/métabolisme , Protéines morphogénétiques osseuses/génétique , Infertilité masculine/génétique , Infertilité masculine/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Testicule/métabolisme
10.
Cell Rep ; 43(6): 114342, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38865240

RÉSUMÉ

The nucleus pulposus (NP) in the intervertebral disc (IVD) arises from embryonic notochord. Loss of notochordal-like cells in humans correlates with onset of IVD degeneration, suggesting that they are critical for healthy NP homeostasis and function. Comparative transcriptomic analyses identified expression of progenitor-associated genes (GREM1, KRT18, and TAGLN) in the young mouse and non-degenerated human NP, with TAGLN expression reducing with aging. Lineage tracing using Tagln-CreERt2 mice identified peripherally located proliferative NP (PeriNP) cells in developing and postnatal NP that provide a continuous supply of cells to the entire NP. PeriNP cells were diminished in aged mice and absent in puncture-induced degenerated discs. Single-cell transcriptomes of postnatal Tagln-CreERt2 IVD cells indicate enrichment for TGF-ß signaling in Tagln descendant NP sub-populations. Notochord-specific removal of TGF-ß/BMP mediator Smad4 results in loss of Tagln+ cells and abnormal NP morphologies. We propose Tagln+ PeriNP cells are potential progenitors crucial for NP homeostasis.


Sujet(s)
Dégénérescence de disque intervertébral , Nucleus pulposus , Cellules souches , Nucleus pulposus/métabolisme , Nucleus pulposus/anatomopathologie , Dégénérescence de disque intervertébral/anatomopathologie , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/génétique , Animaux , Humains , Souris , Cellules souches/métabolisme , Disque intervertébral/métabolisme , Disque intervertébral/anatomopathologie , Facteur de croissance transformant bêta/métabolisme
11.
Elife ; 122024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38856718

RÉSUMÉ

Abnormal lung development can cause congenital pulmonary cysts, the mechanisms of which remain largely unknown. Although the cystic lesions are believed to result directly from disrupted airway epithelial cell growth, the extent to which developmental defects in lung mesenchymal cells contribute to abnormal airway epithelial cell growth and subsequent cystic lesions has not been thoroughly examined. In the present study using genetic mouse models, we dissected the roles of bone morphogenetic protein (BMP) receptor 1a (Bmpr1a)-mediated BMP signaling in lung mesenchyme during prenatal lung development and discovered that abrogation of mesenchymal Bmpr1a disrupted normal lung branching morphogenesis, leading to the formation of prenatal pulmonary cystic lesions. Severe deficiency of airway smooth muscle cells and subepithelial elastin fibers were found in the cystic airways of the mesenchymal Bmpr1a knockout lungs. In addition, ectopic mesenchymal expression of BMP ligands and airway epithelial perturbation of the Sox2-Sox9 proximal-distal axis were detected in the mesenchymal Bmpr1a knockout lungs. However, deletion of Smad1/5, two major BMP signaling downstream effectors, from the lung mesenchyme did not phenocopy the cystic abnormalities observed in the mesenchymal Bmpr1a knockout lungs, suggesting that a Smad-independent mechanism contributes to prenatal pulmonary cystic lesions. These findings reveal for the first time the role of mesenchymal BMP signaling in lung development and a potential pathogenic mechanism underlying congenital pulmonary cysts.


Congenital disorders are medical conditions that are present from birth. Although many congenital disorders are rare, they can have a severe impact on the quality of life of those affected. For example, congenital pulmonary airway malformation (CPAM) is a rare congenital disorder that occurs in around 1 out of every 25,000 pregnancies. In CPAM, abnormal, fluid-filled sac-like pockets of tissue, known as cysts, form within the lungs of unborn babies. After birth, these cysts become air-filled and do not behave like normal lung tissue and stop a baby's lungs from working properly. In severe cases, babies with CPAM need surgery immediately after birth. We still do not understand exactly what the underlying causes of CPAM might be. CPAM is not considered to be hereditary ­ that is, it does not appear to be passed down in families ­ nor is it obviously linked to any environmental factors. CPAM is also very difficult to study, because researchers cannot access tissue samples during the critical early stages of the disease. To overcome these difficulties, Luo et al. wanted to find a way to study CPAM in the laboratory. First, they developed a non-human animal 'model' that naturally forms CPAM-like lung cysts, using genetically modified mice where the gene for the signaling molecule Bmpr1a had been deleted in lung cells. Normally, Bmpr1a is part of a set of the molecular instructions, collectively termed BMP signaling, which guide healthy lung development early in life. However, mouse embryos lacking Bmpr1a developed abnormal lung cysts that were similar to those found in CPAM patients, suggesting that problems with BMP signalling might also trigger CPAM in humans. Luo et al. also identified several other genes in the Bmpr1a-deficient mouse lungs that had abnormal patterns of activity. All these genes were known to be controlled by BMP signaling, and to play a role in the development and organisation of lung tissue. This suggests that when these genes are not controlled properly, they could drive formation of CPAM cysts when BMP signaling is compromised. This work is a significant advance in the tools available to study CPAM. Luo et al.'s results also shed new light on the molecular mechanisms underpinning this rare disorder. In the future, Luo et al. hope this knowledge will help us develop better treatments for CPAM, or even help to prevent it altogether.


Sujet(s)
Récepteurs de la protéine morphogénique osseuse de type I , Poumon , Mésoderme , Souris knockout , Transduction du signal , Animaux , Récepteurs de la protéine morphogénique osseuse de type I/génétique , Récepteurs de la protéine morphogénique osseuse de type I/métabolisme , Récepteurs de la protéine morphogénique osseuse de type I/déficit , Souris , Poumon/embryologie , Poumon/métabolisme , Poumon/anatomopathologie , Mésoderme/embryologie , Mésoderme/métabolisme , Kystes/métabolisme , Kystes/anatomopathologie , Kystes/génétique , Protéines morphogénétiques osseuses/métabolisme , Protéines morphogénétiques osseuses/génétique , Maladies pulmonaires/métabolisme , Maladies pulmonaires/anatomopathologie , Maladies pulmonaires/génétique , Modèles animaux de maladie humaine
12.
Adv Sci (Weinh) ; 11(26): e2307452, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38708713

RÉSUMÉ

Tumor heterogeneity, the presence of multiple distinct subpopulations of cancer cells between patients or among the same tumors, poses a major challenge to current targeted therapies. The way these different subpopulations interact among themselves and the stromal niche environment, and how such interactions affect cancer stem cell behavior has remained largely unknown. Here, it is shown that an FGF-BMP7-INHBA signaling positive feedback loop integrates interactions among different cell populations, including mammary gland stem cells, luminal epithelial and stromal fibroblast niche components not only in organ regeneration but also, with certain modifications, in cancer progression. The reciprocal dependence of basal stem cells and luminal epithelium is based on basal-derived BMP7 and luminal-derived INHBA, which promote their respective expansion, and is regulated by stromal-epithelial FGF signaling. Targeting this interaction loop, for example, by reducing the function of one or more of its components, inhibits organ regeneration and breast cancer progression. The results have profound implications for overcoming drug resistance because of tumor heterogeneity in future targeted therapies.


Sujet(s)
Tumeurs du sein , Niche de cellules souches , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Animaux , Femelle , Niche de cellules souches/physiologie , Cellules souches tumorales/métabolisme , Transduction du signal , Souris , Cellules épithéliales/métabolisme , Protéine morphogénétique osseuse de type 7/métabolisme , Protéine morphogénétique osseuse de type 7/génétique , Microenvironnement tumoral
13.
Environ Pollut ; 351: 124101, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38710361

RÉSUMÉ

Both nanoplastics (NPs) and 3-tert-butyl-4-hydroxyanisole (3-BHA) are environmental contaminants that can bio-accumulate through the food chain. However, the combined effects of which on mammalian female reproductive system remain unclear. Here, the female ICR-CD1 mice were used to evaluate the damage effects of ovaries and uterus after NPs and 3-BHA co-treatment for 35 days. Firstly, co-exposure significantly reduced the body weight and organ index of ovaries and uterus in mice. Secondly, combined effects of NPs and 3-BHA exacerbated the histopathological abnormalities to the ovaries and uterus and decreased female sex hormones such as FSH and LH while increased antioxidant activities including CAT and GSH-Px. Moreover, the apoptotic genes, inflammatory cytokines and the key reproductive development genes such as FSTL1 were significantly up-regulated under co-exposure conditions. Thirdly, through transcriptional and bioinformatics analysis, immunofluorescence and western blotting assays, together with molecular docking simulation, we determined that co-exposure up-regulated the FSTL1, TGF-ß and p-Smad1/5/9 but down-regulated the expression of BMP4. Finally, the pharmacological rescue experiments further demonstrated that co-exposure of NPs and 3-BHA mainly exacerbated the female reproductive toxicity through FSTL1-mediated BMP4/TGF-ß/SMAD signaling pathway. Taken together, our studies provided the theoretical basis of new environmental pollutants on the reproductive health in female mammals.


Sujet(s)
Souris de lignée ICR , Ovaire , Polystyrènes , Utérus , Animaux , Femelle , Souris , Utérus/effets des médicaments et des substances chimiques , Utérus/métabolisme , Ovaire/effets des médicaments et des substances chimiques , Ovaire/métabolisme , Polystyrènes/toxicité , Reproduction/effets des médicaments et des substances chimiques , Microplastiques/toxicité , Protéine morphogénétique osseuse de type 4/génétique , Protéine morphogénétique osseuse de type 4/métabolisme , Nanoparticules/toxicité , Simulation de docking moléculaire , Polluants environnementaux/toxicité , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique
14.
Elife ; 122024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38690987

RÉSUMÉ

Elastic cartilage constitutes a major component of the external ear, which functions to guide sound to the middle and inner ears. Defects in auricle development cause congenital microtia, which affects hearing and appearance in patients. Mutations in several genes have been implicated in microtia development, yet, the pathogenesis of this disorder remains incompletely understood. Here, we show that Prrx1 genetically marks auricular chondrocytes in adult mice. Interestingly, BMP-Smad1/5/9 signaling in chondrocytes is increasingly activated from the proximal to distal segments of the ear, which is associated with a decrease in chondrocyte regenerative activity. Ablation of Bmpr1a in auricular chondrocytes led to chondrocyte atrophy and microtia development at the distal part. Transcriptome analysis revealed that Bmpr1a deficiency caused a switch from the chondrogenic program to the osteogenic program, accompanied by enhanced protein kinase A activation, likely through increased expression of Adcy5/8. Inhibition of PKA blocked chondrocyte-to-osteoblast transformation and microtia development. Moreover, analysis of single-cell RNA-seq of human microtia samples uncovered enriched gene expression in the PKA pathway and chondrocyte-to-osteoblast transformation process. These findings suggest that auricle cartilage is actively maintained by BMP signaling, which maintains chondrocyte identity by suppressing osteogenic differentiation.


Sujet(s)
Chondrocytes , Microtie congénitale , Cyclic AMP-Dependent Protein Kinases , Transduction du signal , Animaux , Chondrocytes/métabolisme , Microtie congénitale/génétique , Microtie congénitale/métabolisme , Souris , Cyclic AMP-Dependent Protein Kinases/métabolisme , Cyclic AMP-Dependent Protein Kinases/génétique , Protéines morphogénétiques osseuses/métabolisme , Protéines morphogénétiques osseuses/génétique , Humains , Récepteurs de la protéine morphogénique osseuse de type I/métabolisme , Récepteurs de la protéine morphogénique osseuse de type I/génétique , Chondrogenèse/génétique , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique
15.
Aging Biol ; 22024.
Article de Anglais | MEDLINE | ID: mdl-38736850

RÉSUMÉ

The gut microbiome plays important roles in host function and health. Core microbiomes have been described for different species, and imbalances in their composition, known as dysbiosis, are associated with pathology. Changes in the gut microbiome and dysbiosis are common in aging, possibly due to multi-tissue deterioration, which includes metabolic shifts, dysregulated immunity, and disrupted epithelial barriers. However, the characteristics of these changes, as reported in different studies, are varied and sometimes conflicting. Using clonal populations of Caenorhabditis elegans to highlight trends shared among individuals, we employed 16s rRNA gene sequencing, CFU counts and fluorescent imaging, identifying an Enterobacteriaceae bloom as a common denominator in aging animals. Experiments using Enterobacter hormaechei, a representative commensal, suggested that the Enterobacteriaceae bloom was facilitated by a decline in Sma/BMP immune signaling in aging animals and demonstrated its potential for exacerbating infection susceptibility. However, such detrimental effects were context-dependent, mitigated by competition with commensal communities, highlighting the latter as determinants of healthy versus unhealthy aging, depending on their ability to restrain opportunistic pathobionts.

16.
Zhongguo Zhong Yao Za Zhi ; 49(7): 1762-1773, 2024 Apr.
Article de Chinois | MEDLINE | ID: mdl-38812188

RÉSUMÉ

The study aimed to investigate the therapeutic effects of the n-butanol extract of Pulsatilla Decoction(BEPD) on ulcerative colitis(UC) via the bone morphogenetic protein(BMP) signaling pathway. C57BL/6 mice were divided into six groups: control, model, mesalazine, and BEPD low-, medium-, and high-dose groups. Except for the control group, the rest groups were treated with 3% dextran sulfate sodium(DSS) freely for seven consecutive days to establish the UC mouse model, followed by treatment with different concentrations of BEPD and mesalazine by gavage. The murine body weight and disease activity index(DAI) were recorded. After the mice were sacrificed, their colon tissues were collected for histological analysis. Alcian blue/periodic acid-Schiff(AB/PAS) staining was used to detect the number and mucus secretion status of goblet cells; immunohistochemistry was performed to measure the expression of ki67, cleaved caspase-3, mucin 2(Muc2), and matrix metalloproteinase-9(MMP9) in colon tissues; and immunofluorescence was used to analyze the expression of tight junction proteins in colon tissues, and enzyme linked immunosorbent assay(ELISA) was employed to quantify the levels of tumor necrosis factor-α(TNF-α), interleukin(IL)-1ß, and IL-6. Western blot was conducted to evaluate the expression of BMP pathway-related proteins in mouse colon tissues. Quantitative real-time PCR(qRT-PCR) was performed to measure the expression of genes related to goblet cell differentiation in mouse colon tissues. In addition, this study also examined the protective effect and underlying mechanism of BEPD-containing serum on lipopolysaccharide(LPS)-induced barrier damages in LS174T goblet cells in vitro. The results showed that BEPD significantly alleviated UC symptoms in mice, restored goblet cell diffe-rentiation function, promoted Muc2 secretion and tight junction protein expression, and suppressed inflammatory factor secretion while activating the BMP signaling pathway. Therefore, BEPD may exert its therapeutic effects on UC by activating the BMP signaling pathway, providing a new strategy for drug intervention in UC.


Sujet(s)
Rectocolite hémorragique , Médicaments issus de plantes chinoises , Souris de lignée C57BL , Pulsatilla , Transduction du signal , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/administration et posologie , Mâle , Pulsatilla/composition chimique , Humains , Protéines morphogénétiques osseuses/métabolisme , Protéines morphogénétiques osseuses/génétique
17.
Theriogenology ; 225: 67-80, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38795512

RÉSUMÉ

Here, we examined the effects of the BMP signaling pathway inhibitor LDN-193189 on the pluripotency of porcine embryonic stem cells (ESCs) in the absence of feeder cells using molecular and transcriptomic techniques. Additionally, the effects of some extracellular matrix components on porcine ESC pluripotency were evaluated to develop an optimized and sustainable feeder-free culture system for porcine ESCs. Feeder cells were found to play an important role in supporting the pluripotency of porcine ESCs by blocking trophoblast and mesodermal differentiation through the inhibition of the BMP pathway. Additionally, treatment with LDN-193189, an inhibitor of the BMP pathway, maintained the pluripotency and homogeneity of porcine ESCs for an extended period in the absence of feeder cells by stimulating the secretion of chemokines and suppressing differentiation, based on transcriptome analysis. Conclusively, these results suggest that LDN-193189 could be a suitable replacement for feeder cells in the maintenance of porcine ESC pluripotency during culture. Additionally, these findings contribute to the understanding of pluripotency gene networks and comparative embryogenesis.


Sujet(s)
Cellules souches embryonnaires , Pyrazoles , Transduction du signal , Animaux , Suidae , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Pyrazoles/pharmacologie , Pyrimidines/pharmacologie , Protéines morphogénétiques osseuses/métabolisme , Cellules souches pluripotentes/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Protéines Smad/métabolisme , Protéines Smad/génétique , Cellules nourricières , Techniques de culture cellulaire
18.
Front Mol Neurosci ; 17: 1356544, 2024.
Article de Anglais | MEDLINE | ID: mdl-38742226

RÉSUMÉ

In the early cerebellar primordium, there are two progenitor zones, the ventricular zone (VZ) residing atop the IVth ventricle and the rhombic lip (RL) at the lateral edges of the developing cerebellum. These zones give rise to the several cell types that form the GABAergic and glutamatergic populations of the adult cerebellum, respectively. Recently, an understanding of the molecular compartmentation of these zones has emerged. To add to this knowledge base, we report on the Msx genes, a family of three transcription factors, that are expressed downstream of Bone Morphogenetic Protein (BMP) signaling in these zones. Using fluorescent RNA in situ hybridization, we have characterized the Msx (Msh Homeobox) genes and demonstrated that their spatiotemporal pattern segregates specific regions within the progenitor zones. Msx1 and Msx2 are compartmentalized within the rhombic lip (RL), while Msx3 is localized within the ventricular zone (VZ). The relationship of the Msx genes with an early marker of the glutamatergic lineage, Atoh1, was examined in Atoh1-null mice and it was found that the expression of Msx genes persisted. Importantly, the spatial expression of Msx1 and Msx3 altered in response to the elimination of Atoh1. These results point to the Msx genes as novel early markers of cerebellar progenitor zones and more importantly to an updated view of the molecular parcellation of the RL with respect to the canonical marker of the RL, Atoh1.

19.
Dev Cell ; 59(9): 1210-1230.e9, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38569548

RÉSUMÉ

The Drosophila larval ventral nerve cord (VNC) shares many similarities with the spinal cord of vertebrates and has emerged as a major model for understanding the development and function of motor systems. Here, we use high-quality scRNA-seq, validated by anatomical identification, to create a comprehensive census of larval VNC cell types. We show that the neural lineages that comprise the adult VNC are already defined, but quiescent, at the larval stage. Using fluorescence-activated cell sorting (FACS)-enriched populations, we separate all motor neuron bundles and link individual neuron clusters to morphologically characterized known subtypes. We discovered a glutamate receptor subunit required for basal neurotransmission and homeostasis at the larval neuromuscular junction. We describe larval glia and endorse the general view that glia perform consistent activities throughout development. This census represents an extensive resource and a powerful platform for future discoveries of cellular and molecular mechanisms in repair, regeneration, plasticity, homeostasis, and behavioral coordination.


Sujet(s)
Drosophila melanogaster , Larve , Motoneurones , Animaux , Larve/génétique , Larve/métabolisme , Motoneurones/métabolisme , Motoneurones/cytologie , Drosophila melanogaster/génétique , Drosophila melanogaster/métabolisme , Névroglie/métabolisme , Névroglie/cytologie , Jonction neuromusculaire/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , RNA-Seq/méthodes , Analyse de l'expression du gène de la cellule unique
20.
Methods Mol Biol ; 2803: 13-33, 2024.
Article de Anglais | MEDLINE | ID: mdl-38676882

RÉSUMÉ

The adept and systematic differentiation of embryonic stem cells (ESCs) and human-induced pluripotent stem cells (hiPSCs) to diverse lineage-prone cell types involves crucial step-by-step process that mimics the vital strategic commitment phase that is usually observed during the process of embryo development. The development of precise tissue-specific cell types from these stem cells indeed plays an important role in the advancement of imminent stem cell-based therapeutic strategies. Therefore, the usage of hiPSC-derived cell types for subsequent cardiovascular disease modeling, drug screening, and therapeutic drug development undeniably entails an in-depth understanding of each and every step to proficiently stimulate these stem cells into desired cardiomyogenic lineage. Thus, to accomplish this definitive and decisive fate, it is essential to efficiently induce the mesoderm or pre-cardiac mesoderm, succeeded by the division of cells into cardiovascular and ultimately ensuing with the cardiomyogenic lineage outcome. This usually commences from the earliest phases of pluripotent cell induction. In this chapter, we discuss our robust and reproducible step-wise protocol that will describe the subtype controlled, precise lineage targeted standardization of activin/nodal, and BMP signaling molecules/cytokines, for the efficient differentiation of ventricular cardiomyocytes from hiPSCs via the embryoid body method. In addition, we also describe techniques to dissociate hiPSCs, hiPSC-derived early cardiomyocytes for mesoderm and pre-cardiac mesoderm assessment, and hiPSC-derived cardiomyocytes for early and mature markers assessment.


Sujet(s)
Différenciation cellulaire , Cellules souches pluripotentes induites , Myocytes cardiaques , Humains , Activines/pharmacologie , Activines/métabolisme , Protéines morphogénétiques osseuses/métabolisme , Techniques de culture cellulaire/méthodes , Lignage cellulaire , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Protéine Nodal/métabolisme , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE