Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Adv Sci (Weinh) ; 11(32): e2400238, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38923264

ABSTRACT

The placenta links feto-maternal circulation for exchanges of nutrients, gases, and metabolic wastes between the fetus and mother, being essential for pregnancy process and maintenance. The allantois and mesodermal components of amnion, chorion, and yolk sac are derived from extraembryonic mesoderm (Ex-Mes), however, the mechanisms contributing to distinct components of the placenta and regulation the interactions between allantois and epithelium during chorioallantoic fusion and labyrinth formation remains unclear. Isl1 is expressed in progenitors of the Ex-Mes and allantois the Isl1 mut mouse line is analyzed to investigate contribution of Isl1+ Ex-Mes / allantoic progenitors to cells of the allantois and placenta. This study shows that Isl1 identifies the Ex-Mes progenitors for endothelial and vascular smooth muscle cells, and most of the mesenchymal cells of the placenta and umbilical cord. Deletion of Isl1 causes defects in allantois growth, chorioallantoic fusion, and placenta vessel morphogenesis. RNA-seq and CUT&Tag analyses revealed that Isl1 promotes allantoic endothelial, inhibits mesenchymal cell differentiation, and allantoic signals regulated by Isl1 mediating the inductive interactions between the allantois and chorion critical for chorionic epithelium differentiation, villous formation, and labyrinth angiogenesis. This study above reveals that Isl1 plays roles in regulating multiple genetic and epigenetic pathways of vascular morphogenesis, provides the insight into the mechanisms for placental formation, highlighting the necessity of Isl1 for placenta formation/pregnant maintenance.


Subject(s)
Allantois , LIM-Homeodomain Proteins , Mesoderm , Placenta , Transcription Factors , Animals , Pregnancy , Mice , Female , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Allantois/metabolism , Placenta/metabolism , Placenta/blood supply , Transcription Factors/genetics , Transcription Factors/metabolism , Mesoderm/metabolism , Mesoderm/embryology , Morphogenesis/genetics , Morphogenesis/physiology , Placentation/genetics , Placentation/physiology
3.
Cell Res ; 32(9): 843-854, 2022 09.
Article in English | MEDLINE | ID: mdl-35840807

ABSTRACT

The sinoatrial node (SAN) is the origin of the electrical signals for rhythmic heartbeats in mammals. The spontaneous firing of SAN pacemaker cells (SANPCs) triggers cardiac contraction. 'Local Ca2+ release' (LCR), a unique cellular activity, acts as the 'engine' of the spontaneous firing of SANPCs. However, the mechanism of LCR initiation remains unclear. Here, we report that endogenous glutamate drives LCRs in SANPCs. Using a glutamate sensor, we unraveled a tight correlation between glutamate accumulation and LCR occurrence, indicating a potential relationship between glutamate and LCRs. Intracellular application of glutamate significantly enhanced the LCRs in both intact and permeabilized SANPCs. Mechanistically, we revealed that mitochondrial excitatory amino acid transporter 1 (EAAT1)-dependent mitochondrial glutamate import promoted ROS generation, which in turn led to the oxidation of Ca2+-handling proteins, ultimately resulting in enhanced LCRs. Importantly, EAAT1 depletion reduced both the spontaneous firing rates of isolated SANPCs and the heart rate in vitro and in vivo, suggesting the central role of EAAT1 as a glutamate transporter in the regulation of cardiac autonomic rhythm. In conclusion, our results indicate that glutamate serves as an LCR igniter in SANPCs, adding a potentially important element to the coupled-clock theory that explains the origin of spontaneous firing. These findings shed new light on the future prevention and treatment of cardiac pacemaker cell-related arrhythmias.


Subject(s)
Glutamic Acid , Sinoatrial Node , Animals , Calcium/metabolism , Glutamic Acid/metabolism , Mammals , Myocytes, Cardiac/metabolism , Sinoatrial Node/metabolism
4.
Front Public Health ; 9: 649170, 2021.
Article in English | MEDLINE | ID: mdl-33937177

ABSTRACT

Increased population movement has increased the risk of reintroducing parasites to elimination areas and also dispersing drug-resistant parasites to new regions. Therefore, reliable and repeatable methods to trace back to the source of imported infections are essential. The recently developed 23-single-nucleotide polymorphism (SNP) barcode from organellar genomes of mitochondrion (mt) and apicoplast (apico) provides a valuable tool to locate the geographic origin of Plasmodium falciparum. This study aims to explore the feasibility of using the 23-SNP barcode for tracking P. falciparum by polymerase chain reaction and sequencing, while providing geographical haplotypes of isolates that originated from Central Africa. Based on 23-SNP barcode analysis, SNPs were found at seven loci; 27 isolates were confirmed to have originated in West Africa, and this study also showed four isolates from Central Africa (Equatorial Guinea, 3; Republic of Congo, 1) that originated in East Africa. This study provides the sequence data from Central Africa and fills 23-SNP barcode data gaps of sample origins.


Subject(s)
Plasmodium falciparum , Africa, Eastern , Africa, Western , Congo , Equatorial Guinea , Plasmodium falciparum/genetics , Polymerase Chain Reaction
5.
Cell Res ; 31(9): 951-964, 2021 09.
Article in English | MEDLINE | ID: mdl-33824424

ABSTRACT

As an excitatory transmitter system, the glutamatergic transmitter system controls excitability and conductivity of neurons. Since both cardiomyocytes and neurons are excitable cells, we hypothesized that cardiomyocytes may also be regulated by a similar system. Here, we have demonstrated that atrial cardiomyocytes have an intrinsic glutamatergic transmitter system, which regulates the generation and propagation of action potentials. First, there are abundant vesicles containing glutamate beneath the plasma membrane of rat atrial cardiomyocytes. Second, rat atrial cardiomyocytes express key elements of the glutamatergic transmitter system, such as the glutamate metabolic enzyme, ionotropic glutamate receptors (iGluRs), and glutamate transporters. Third, iGluR agonists evoke iGluR-gated currents and decrease the threshold of electrical excitability in rat atrial cardiomyocytes. Fourth, iGluR antagonists strikingly attenuate the conduction velocity of electrical impulses in rat atrial myocardium both in vitro and in vivo. Knockdown of GRIA3 or GRIN1, two highly expressed iGluR subtypes in atria, drastically decreased the excitatory firing rate and slowed down the electrical conduction velocity in cultured human induced pluripotent stem cell (iPSC)-derived atrial cardiomyocyte monolayers. Finally, iGluR antagonists effectively prevent and terminate atrial fibrillation in a rat isolated heart model. In addition, the key elements of the glutamatergic transmitter system are also present and show electrophysiological functions in human atrial cardiomyocytes. In conclusion, our data reveal an intrinsic glutamatergic transmitter system directly modulating excitability and conductivity of atrial cardiomyocytes through controlling iGluR-gated currents. Manipulation of this system may open potential new avenues for therapeutic intervention of cardiac arrhythmias.


Subject(s)
Atrial Fibrillation , Induced Pluripotent Stem Cells , Action Potentials , Animals , Heart Atria , Humans , Myocytes, Cardiac , Rats
6.
Protein Cell ; 12(7): 545-556, 2021 07.
Article in English | MEDLINE | ID: mdl-33548033

ABSTRACT

Activation of the heart normally begins in the sinoatrial node (SAN). Electrical impulses spontaneously released by SAN pacemaker cells (SANPCs) trigger the contraction of the heart. However, the cellular nature of SANPCs remains controversial. Here, we report that SANPCs exhibit glutamatergic neuron-like properties. By comparing the single-cell transcriptome of SANPCs with that of cells from primary visual cortex in mouse, we found that SANPCs co-clustered with cortical neurons. Tissue and cellular imaging confirmed that SANPCs contained key elements of glutamatergic neurotransmitter system, expressing genes encoding glutamate synthesis pathway (Gls), ionotropic and metabotropic glutamate receptors (Grina, Gria3, Grm1 and Grm5), and glutamate transporters (Slc17a7). SANPCs highly expressed cell markers of glutamatergic neurons (Snap25 and Slc17a7), whereas Gad1, a marker of GABAergic neurons, was negative. Functional studies revealed that inhibition of glutamate receptors or transporters reduced spontaneous pacing frequency of isolated SAN tissues and spontaneous Ca2+ transients frequency in single SANPC. Collectively, our work suggests that SANPCs share dominant biological properties with glutamatergic neurons, and the glutamatergic neurotransmitter system may act as an intrinsic regulation module of heart rhythm, which provides a potential intervention target for pacemaker cell-associated arrhythmias.


Subject(s)
Biological Clocks/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Primary Visual Cortex/metabolism , Sinoatrial Node/metabolism , Transcriptome , Action Potentials/physiology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Calcium/metabolism , Carrier Proteins/classification , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/classification , Nerve Tissue Proteins/metabolism , Neurons/cytology , Primary Visual Cortex/cytology , Receptors, Ionotropic Glutamate/classification , Receptors, Ionotropic Glutamate/genetics , Receptors, Ionotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/classification , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Single-Cell Analysis , Sinoatrial Node/cytology , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL