Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Nature ; 579(7798): 291-296, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103174

RESUMEN

The DNA-dependent protein kinase (DNA-PK), which comprises the KU heterodimer and a catalytic subunit (DNA-PKcs), is a classical non-homologous end-joining (cNHEJ) factor1. KU binds to DNA ends, initiates cNHEJ, and recruits and activates DNA-PKcs. KU also binds to RNA, but the relevance of this interaction in mammals is unclear. Here we use mouse models to show that DNA-PK has an unexpected role in the biogenesis of ribosomal RNA (rRNA) and in haematopoiesis. The expression of kinase-dead DNA-PKcs abrogates cNHEJ2. However, most mice that both expressed kinase-dead DNA-PKcs and lacked the tumour suppressor TP53 developed myeloid disease, whereas all other previously characterized mice deficient in both cNHEJ and TP53 expression succumbed to pro-B cell lymphoma3. DNA-PK autophosphorylates DNA-PKcs, which is its best characterized substrate. Blocking the phosphorylation of DNA-PKcs at the T2609 cluster, but not the S2056 cluster, led to KU-dependent defects in 18S rRNA processing, compromised global protein synthesis in haematopoietic cells and caused bone marrow failure in mice. KU drives the assembly of DNA-PKcs on a wide range of cellular RNAs, including the U3 small nucleolar RNA, which is essential for processing of 18S rRNA4. U3 activates purified DNA-PK and triggers phosphorylation of DNA-PKcs at T2609. DNA-PK, but not other cNHEJ factors, resides in nucleoli in an rRNA-dependent manner and is co-purified with the small subunit processome. Together our data show that DNA-PK has RNA-dependent, cNHEJ-independent functions during ribosome biogenesis that require the kinase activity of DNA-PKcs and its phosphorylation at the T2609 cluster.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Hematopoyesis/genética , Autoantígeno Ku/metabolismo , Linfoma/enzimología , Linfoma/fisiopatología , ARN Ribosómico 18S/metabolismo , Proteínas de Unión al Calcio/genética , Dominio Catalítico/fisiología , Reparación del ADN/genética , Activación Enzimática/genética , Células HeLa , Humanos , Linfoma/genética , Modelos Animales , Mutación , Fosforilación , Unión Proteica , Biosíntesis de Proteínas/genética , ARN Ribosómico 18S/genética , ARN Nucleolar Pequeño/metabolismo
2.
Inorg Chem ; 63(36): 16733-16739, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39185620

RESUMEN

Sulfur hexafluoride (SF6) is widely used as an insulating gas, being an etchant and contrast agent in the electrical, semiconductor, and medical industries. However, due to its long lifetime and high global warming potential in the atmosphere, SF6 must be carefully handled to prevent leakage during production and usage. Herein, we report a sod-net metal-azolate framework (MAF) named MAF-stu-111, which decorates methyl and aldehyde groups in the porous windows, showing high adsorption affinity for SF6 at low pressure. Stability tests, gas adsorption, and breakthrough experiments demonstrated that MAF-stu-111 possesses excellent water and chemical stability, fully reversible SF6 uptake, high SF6/N2 separation selectivity (10:90, 285.2), good reusability, and high SF6 recovery purity (99.03%). Theoretical calculations revealed that hydrogen atoms of methyl and aldehyde groups can form multiple hydrogen bonds with SF6 molecules, which ensure that SF6 molecules are firmly held within the MAF-stu-111 framework, playing a key role in the selective separation of SF6/N2.

3.
Nature ; 554(7690): 112-117, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29364875

RESUMEN

Many craniofacial disorders are caused by heterozygous mutations in general regulators of housekeeping cellular functions such as transcription or ribosome biogenesis. Although it is understood that many of these malformations are a consequence of defects in cranial neural crest cells, a cell type that gives rise to most of the facial structures during embryogenesis, the mechanism underlying cell-type selectivity of these defects remains largely unknown. By exploring molecular functions of DDX21, a DEAD-box RNA helicase involved in control of both RNA polymerase (Pol) I- and II-dependent transcriptional arms of ribosome biogenesis, we uncovered a previously unappreciated mechanism linking nucleolar dysfunction, ribosomal DNA (rDNA) damage, and craniofacial malformations. Here we demonstrate that genetic perturbations associated with Treacher Collins syndrome, a craniofacial disorder caused by heterozygous mutations in components of the Pol I transcriptional machinery or its cofactor TCOF1 (ref. 1), lead to relocalization of DDX21 from the nucleolus to the nucleoplasm, its loss from the chromatin targets, as well as inhibition of rRNA processing and downregulation of ribosomal protein gene transcription. These effects are cell-type-selective, cell-autonomous, and involve activation of p53 tumour-suppressor protein. We further show that cranial neural crest cells are sensitized to p53-mediated apoptosis, but blocking DDX21 loss from the nucleolus and chromatin rescues both the susceptibility to apoptosis and the craniofacial phenotypes associated with Treacher Collins syndrome. This mechanism is not restricted to cranial neural crest cells, as blood formation is also hypersensitive to loss of DDX21 functions. Accordingly, ribosomal gene perturbations associated with Diamond-Blackfan anaemia disrupt DDX21 localization. At the molecular level, we demonstrate that impaired rRNA synthesis elicits a DNA damage response, and that rDNA damage results in tissue-selective and dosage-dependent effects on craniofacial development. Taken together, our findings illustrate how disruption in general regulators that compromise nucleolar homeostasis can result in tissue-selective malformations.


Asunto(s)
Nucléolo Celular/metabolismo , Nucléolo Celular/patología , Daño del ADN , ADN Ribosómico/metabolismo , Disostosis Mandibulofacial/genética , Disostosis Mandibulofacial/patología , Estrés Fisiológico , Animales , Apoptosis , Benzotiazoles/farmacología , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/patología , Cromatina/metabolismo , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , ADN Ribosómico/genética , ARN Polimerasas Dirigidas por ADN/deficiencia , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Disostosis Mandibulofacial/embriología , Ratones , Naftiridinas/farmacología , Cresta Neural/enzimología , Cresta Neural/patología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Fenotipo , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transporte de Proteínas/efectos de los fármacos , ARN Helicasas/metabolismo , ARN Polimerasa I/antagonistas & inhibidores , ARN Ribosómico/biosíntesis , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Proteínas Ribosómicas/biosíntesis , Proteínas Ribosómicas/genética , Ribosomas/genética , Ribosomas/metabolismo , Cráneo/patología , Estrés Fisiológico/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Xenopus , Pez Cebra/embriología , Proteínas de Pez Cebra/deficiencia
4.
Can J Physiol Pharmacol ; 102(1): 1-13, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37903419

RESUMEN

Cardiovascular diseases remain a leading cause of hospitalization affecting approximately 38 million people worldwide. While pharmacological and revascularization techniques can improve the patient's survival and quality of life, they cannot help reversing myocardial infarction injury and heart failure. Direct reprogramming of somatic cells to cardiomyocyte and cardiac progenitor cells offers a new approach to cellular reprogramming and paves the way for translational regenerative medicine. Direct reprogramming can bypass the pluripotent stage with the potential advantage of non-immunogenic cell products, reduced carcinogenic risk, and no requirement for embryonic tissue. The process of directly reprogramming cardiac cells was first achieved through the overexpression of transcription factors such as GATA4, MEF2C, and TBX5. However, over the past decade, significant work has been focused on enhancing direct reprogramming using a mixture of transcription factors, microRNAs, and small molecules to achieve cardiac cell fate. This review discusses the evolution of direct reprogramming, recent progress in achieving efficient cardiac cell fate conversion, and describes the reprogramming mechanisms at a molecular level. We also explore various viral and non-viral delivery methods currently being used to aid in the delivery of reprogramming factors to improve efficiency. However, further studies will be needed to overcome molecular and epigenetic barriers to successfully achieve translational cardiac regenerative therapeutics.


Asunto(s)
Técnicas de Reprogramación Celular , Calidad de Vida , Humanos , Técnicas de Reprogramación Celular/métodos , Miocitos Cardíacos , Reprogramación Celular , Factores de Transcripción/genética , Medicina Regenerativa/métodos , Fibroblastos
5.
Environ Sci Technol ; 57(40): 15203-15212, 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37729390

RESUMEN

Cr(VI) rebound is the primary risk associated with the reduction remediation of Cr(VI)-contaminated soil. The potential impact of sulfites, which can be produced by microbial activities or originate from sulfur-containing remediation agents, on the Cr(VI) rebound in the vadose zone has been overlooked. When sulfites are present, the stability of CrxFe1-x(OH)3 is compromised and significantly inferior to that of Cr(OH)3, as demonstrated in this paper. First, Fe acts as a catalyst for the conversion of adsorbed sulfite to SO4·-, which subsequently triggers the oxidation of Cr(III) and results in the rebound of Cr(VI). The heterogeneous catalysis by Fe on the surface of CrxFe1-x(OH)3 plays a predominant role, contributing to 78% of the actual oxidation of Cr(III) among all employed catalytic processes. The presence of ambient Cl- can exacerbate the rebound effect of Cr(VI) by promoting the generation of HOCl. Furthermore, a portion of released Cr(VI) was reduced to Cr(III) by dissolved sulfite in the presence of dissolved Fe as a catalyst, thereby increasing the dissolution and migration risk associated with CrxFe1-x(OH)3. Hence, the presence of sulfites results in a significant increase in the Cr(VI) rebound and Cr(III) release from CrxFe1-x(OH)3. This challenges the conventional understanding of the stability of CrxFe1-x(OH)3.

6.
Mol Ther ; 30(1): 54-74, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34678511

RESUMEN

Fibroblasts can be reprogrammed into cardiovascular progenitor cells (CPCs) using transgenic approaches, although the underlying mechanism remains unclear. We determined whether activation of endogenous genes such as Gata4, Nkx2.5, and Tbx5 can rapidly establish autoregulatory loops and initiate CPC generation in adult extracardiac fibroblasts using a CRISPR activation system. The induced fibroblasts (>80%) showed phenotypic changes as indicated by an Nkx2.5 cardiac enhancer reporter. The progenitor characteristics were confirmed by colony formation and expression of cardiovascular genes. Cardiac sphere induction segregated the early and late reprogrammed cells that can generate functional cardiomyocytes and vascular cells in vitro. Therefore, they were termed CRISPR-induced CPCs (ciCPCs). Transcriptomic analysis showed that cell cycle and heart development pathways were important to accelerate CPC formation during the early reprogramming stage. The CRISPR system opened the silenced chromatin locus, thereby allowing transcriptional factors to access their own promoters and eventually forming a positive feedback loop. The regenerative potential of ciCPCs was assessed after implantation in mouse myocardial infarction models. The engrafted ciCPCs differentiated into cardiovascular cells in vivo but also significantly improved contractile function and scar formation. In conclusion, multiplex gene activation was sufficient to drive CPC reprogramming, providing a new cell source for regenerative therapeutics.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Infarto del Miocardio , Animales , Diferenciación Celular/genética , Reprogramación Celular/genética , Fibroblastos/metabolismo , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Células Madre/metabolismo
7.
Stem Cells ; 37(7): 844-854, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30913336

RESUMEN

Pluripotent stem cells (PSCs) are an attractive, reliable source for generating functional cardiomyocytes for regeneration of infarcted heart. However, inefficient cell engraftment into host tissue remains a notable challenge to therapeutic success due to mechanical damage or relatively inhospitable microenvironment. Evidence has shown that excessively formed scar tissues around cell delivery sites present as mechanical and biological barriers that inhibit migration and engraftment of implanted cells. In this review, we focus on the functional responses of stem cells and cardiomyocytes during the process of cardiac fibrosis and scar formation. Survival, migration, contraction, and coupling function of implanted cells may be affected by matrix remodeling, inflammatory factors, altered tissue stiffness, and presence of electroactive myofibroblasts in the fibrotic microenvironment. Although paracrine factors from implanted cells can improve cardiac fibrosis, the transient effect is insufficient for complete repair of an infarcted heart. Furthermore, investigation of interactions between implanted cells and fibroblasts including myofibroblasts helps the identification of new targets to optimize the host substrate environment for facilitating cell engraftment and functional integration. Several antifibrotic approaches, including the use of pharmacological agents, gene therapies, microRNAs, and modified biomaterials, can prevent progression of heart failure and have been developed as adjunct therapies for stem cell-based regeneration. Investigation and optimization of new biomaterials is also required to enhance cell engraftment of engineered cardiac tissue and move PSCs from a laboratory setting into translational medicine.


Asunto(s)
Cicatriz/prevención & control , Terapia Genética/métodos , Infarto del Miocardio/terapia , Células Madre Pluripotentes/citología , Trasplante de Células Madre/métodos , Ingeniería de Tejidos/métodos , Animales , Cicatriz/genética , Cicatriz/inmunología , Cicatriz/patología , Modelos Animales de Enfermedad , Fibrosis , Supervivencia de Injerto/efectos de los fármacos , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Mecanotransducción Celular/genética , Mecanotransducción Celular/inmunología , MicroARNs/genética , MicroARNs/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Comunicación Paracrina/genética , Comunicación Paracrina/inmunología , Células Madre Pluripotentes/metabolismo , Regeneración/genética , Regeneración/inmunología
8.
Int J Mol Sci ; 21(20)2020 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-33081233

RESUMEN

Limited adult cardiac cell proliferation after cardiovascular disease, such as heart failure, hampers regeneration, resulting in a major loss of cardiomyocytes (CMs) at the site of injury. Recent studies in cellular reprogramming approaches have provided the opportunity to improve upon previous techniques used to regenerate damaged heart. Using these approaches, new CMs can be regenerated from differentiation of iPSCs (similar to embryonic stem cells), the direct reprogramming of fibroblasts [induced cardiomyocytes (iCMs)], or induced cardiac progenitors. Although these CMs have been shown to functionally repair infarcted heart, advancements in technology are still in the early stages of development in research laboratories. In this review, reprogramming-based approaches for generating CMs are briefly introduced and reviewed, and the challenges (including low efficiency, functional maturity, and safety issues) that hinder further translation of these approaches into a clinical setting are discussed. The creative and combined optimal methods to address these challenges are also summarized, with optimism that further investigation into tissue engineering, cardiac development signaling, and epigenetic mechanisms will help to establish methods that improve cell-reprogramming approaches for heart regeneration.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Cardiopatías/terapia , Corazón/fisiología , Trasplante de Células Madre/métodos , Ingeniería de Tejidos/métodos , Animales , Humanos , Regeneración
9.
Exp Cell Res ; 371(1): 20-30, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29842877

RESUMEN

Although resident cardiac stem cells have been reported, regeneration of functional cardiomyocytes (CMs) remains a challenge. The present study identifies an alternative progenitor source for CM regeneration without the need for genetic manipulation or invasive heart biopsy procedures. Unlike limb skeletal muscles, masseter muscles (MM) in the mouse head are developed from Nkx2-5 mesodermal progenitors. Adult masseter muscle satellite cells (MMSCs) display heterogeneity in developmental origin and cell phenotypes. The heterogeneous MMSCs that can be characterized by cell sorting based on stem cell antigen-1 (Sca1) show different lineage potential. While cardiogenic potential is preserved in Sca1+ MMSCs as shown by expression of cardiac progenitor genes (including Nkx2-5), skeletal myogenic capacity is maintained in Sca1- MMSCs with Pax7 expression. Sca1+ MMSC-derived beating cells express cardiac genes and exhibit CM-like morphology. Electrophysiological properties of MMSC-derived CMs are demonstrated by calcium transients and action potentials. These findings show that MMSCs could serve as a novel cell source for cardiomyocyte replacement.


Asunto(s)
Diferenciación Celular , Músculo Masetero/citología , Desarrollo de Músculos/genética , Miocitos Cardíacos/citología , Células Satélite del Músculo Esquelético/citología , Potenciales de Acción/fisiología , Animales , Ataxina-1/genética , Ataxina-1/metabolismo , Biomarcadores/metabolismo , Calcio/metabolismo , Linaje de la Célula/genética , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Músculo Masetero/metabolismo , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Fenotipo , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Proteína Fluorescente Roja
10.
Stem Cells ; 35(2): 337-350, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27538588

RESUMEN

Therapeutic angiogenesis has emerged as a promising strategy to regenerate the damaged blood vessels resulting from ischemic diseases such as myocardial infarction (MI). However, the functional integration of implanted endothelial cells (ECs) in infarcted heart remains challenging. We herein develop an EC generation approach by inhibiting microRNA-495 (miR-495) in human induced pluripotent stem cells (hiPSCs) and assess the angiogenic potential for MI treatment. The anti-angiogenic miR-495 belonging to Dlk1-Dio3 miR cluster was identified through expression profiling and computational analysis. Loss-of-function experiments for miR-495 were performed using a lentiviral transfer of antisense sequence in hiPSCs. The pluripotency of hiPSCs was not impacted by the genetic modification. Induced with differentiation medium, miR-495 inhibition enhanced the expression of EC genes of hiPSCs, as well as the yield of ECs. Newly derived ECs displayed prominent angiogenic characteristics including tube formation, cell migration, and proliferation. Mechanistically, miR-495 mediated the expression of endothelial or angiogenic genes by directly targeting vascular endothelial zinc finger 1. After transplantation in immunodeficient MI mice, the derived ECs significantly increased neovascularization in the infarcted heart, prevented functional worsening, and attenuated expansion of infarct size. The functional integration of the implanted ECs into coronary networks was also enhanced by inhibiting miR-495. miR-495 represents a new target not only for promoting EC generation from hiPSCs but also for enhancing angiogenesis and engraftment of hiPSC-derived ECs in ischemic heart. Stem Cells 2017;35:337-350.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/antagonistas & inhibidores , Neovascularización Fisiológica , Animales , Secuencia de Bases , Cardiotónicos/metabolismo , Diferenciación Celular/genética , Vasos Coronarios/patología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Familia de Multigenes , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Trasplante de Células Madre , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Cell Biochem Funct ; 35(2): 113-123, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28233339

RESUMEN

It has been reported that CXCR4-overexpressing mesenchymal stem cells (MSCCX4 ) can repair heart tissue post myocardial infarction. This study aims to investigate the MSCCX4-derived paracrine cardio-protective signaling in the presence of myocardial infarction. Mesenchymal stem cells (MSCs) were divided into 3 groups: MSC only, MSCCX4 , and CXCR4 gene-specific siRNA-transduced MSC. Mesenchymal stem cells were exposed to hypoxia, and then MSCs-conditioned culture medium was incubated with neonatal and adult cardiomyocytes, respectively. Cell proliferation-regulating genes were assessed by real-time polymerase chain reaction (RT-PCR). In vitro: The number of cardiomyocytes undergoing DNA synthesis, cytokinesis, and mitosis was increased to a greater extent in MSCCX4 medium-treated group than control group, while this proproliferative effect was reduced in CXCR4 gene-specific siRNA-transduced MSC-treated cells. Accordingly, the maximal enhancement of vascular endothelial growth factor, cyclin 2, and transforming growth factor-ß2 was observed in hypoxia-exposed MSCCX4 . In vivo: MSCs were labeled with enhanced green fluorescent protein (EGFP) and engrafted into injured myocardium in rats. The number of EGFP and CD31 positive cells in the MSCCX4 group was significantly increased than other 2 groups, associated with the reduced left ventricular (LV) fibrosis, the increased LV free wall thickness, the enhanced angiogenesis, and the improved contractile function. CXCR4 overexpression can mobilize MSCs into ischemic area, whereby these cells can promoted angiogenesis and alleviate LV remodeling via paracrine signaling mechanism.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Comunicación Paracrina/genética , Receptores CXCR4/genética , Animales , Animales Recién Nacidos , Hipoxia de la Célula , Medios de Cultivo Condicionados/farmacología , Ciclina A2/genética , Ciclina A2/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Neovascularización Fisiológica , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Transfección , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Remodelación Ventricular
12.
J Cell Mol Med ; 19(8): 1825-35, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25824297

RESUMEN

The chemokine (C-X-C motif) receptor 4 (CXCR4) is expressed on native cardiomyocytes and can modulate isolated cardiomyocyte contractility. This study examines the role of CXCR4 in cardiomyocyte response to ischaemia-reperfusion (I/R) injury. Isolated adult rat ventricular cardiomyocytes were subjected to hypoxia/reoxygenation (H/R) to simulate I/R injury. In response to H/R injury, the decrease in CXCR4 expression was associated with dysfunctional energy metabolism indicated by an increased adenosine diphosphate/adenosine triphosphate (ADP/ATP) ratio. CXCR4-overexpressing cardiomyocytes were used to determine whether such overexpression (OE) can prevent bio-energetic disruption-associated cell death. CXCR4 OE was performed with adenoviral infection with CXCR4 encoding-gene or non-translated nucleotide sequence (Control). The increased CXCR4 expression was observed in cardiomyocytes post CXCR4-adenovirus transduction and this OE significantly reduced the cardiomyocyte contractility under basal conditions. Although the same extent of H/R-provoked cytosolic calcium overload was measured, the hydrogen peroxide-induced decay of mitochondrial membrane potential was suppressed in CXCR4 OE group compared with control group, and the mitochondrial swelling was significantly attenuated in CXCR4 group, implicating that CXCR4 OE prevents permeability transition pore opening exposure to overload calcium. Interestingly, this CXCR4-induced mitochondrial protective effect is associated with the enhanced signal transducer and activator of transcription 3 (expression in mitochondria. Consequently, in the presence of H/R, mitochondrial dysfunction was mitigated and cardiomyocyte death was decreased to 65% in the CXCR4 OE group as compared with the control group. I/R injury leads to the reduction in CXCR4 in cardiomyocytes associated with the dysfunctional energy metabolism, and CXCR4 OE can alleviate mitochondrial dysfunction to improve cardiomyocyte survival.


Asunto(s)
Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Receptores CXCR4/metabolismo , Adenoviridae/metabolismo , Animales , Calcio/farmacología , Cardiotónicos/farmacología , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/metabolismo , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/patología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo
13.
Pediatr Discov ; 2(3)2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39308981

RESUMEN

Despite advances in prenatal screening and a notable decrease in mortality rates, congenital heart disease (CHD) remains the most prevalent congenital disorder in newborns globally. Current therapeutic surgical approaches face challenges due to the significant rise in complications and disabilities. Emerging cardiac regenerative therapies offer promising adjuncts for CHD treatment. One novel avenue involves investigating methods to stimulate cardiomyocyte proliferation. However, the mechanism of altered cardiomyocyte proliferation in CHD is not fully understood, and there are few feasible approaches to stimulate cardiomyocyte cell cycling for optimal healing in CHD patients. In this review, we explore recent progress in understanding genetic and epigenetic mechanisms underlying defective cardiomyocyte proliferation in CHD from development through birth. Targeting cell cycle pathways shows promise for enhancing cardiomyocyte cytokinesis, division, and regeneration to repair heart defects. Advancements in human disease modeling techniques, CRISPR-based genome and epigenome editing, and next-generation sequencing technologies will expedite the exploration of abnormal machinery governing cardiomyocyte differentiation, proliferation, and maturation across diverse genetic backgrounds of CHD. Ongoing studies on screening drugs that regulate cell cycling are poised to translate this nascent technology of enhancing cardiomyocyte proliferation into a new therapeutic paradigm for CHD surgical interventions.

14.
J Hazard Mater ; 465: 133112, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38043420

RESUMEN

Hexavalent chromium (Cr(VI)) contamination in soil and groundwater is usually remediated via reduction techniques. The formation of crystalline chromium phosphate (CrPO4·6 H2O) occurs as a byproduct during Cr(VI) remediation processes in the presence of phosphate, yet its stability in the environment has received limited attention. In this study, the formation conditions, structure, properties, and risks associated with the dissolution and oxidation of CrPO4·6 H2O were comprehensively assessed. Results showed that crystalline CrPO4·6 H2O was formed under pH 5 - 7 at room temperature. CrPO4·6 H2O exhibits higher dissolution risk compared to Cr(OH)3·3 H2O due to a long Cr-P bond (4.2 Å). H+ and OH- increased the risk of dissolution at pH 5 and 11, respectively, owing to the formation of CrH2PO42+ and Cr(OH)4-. In addition, under faintly acidic conditions, the high solubility of CrPO4·6 H2O increases the risk of oxidation; under neutral and weakly alkaline conditions, the presence of positively charged Cr(H2O)63+ structures on the surface elevates its susceptibility to contact and oxidation by δ-MnO2 compared to Cr(OH)3·3 H2O. Specifically, at pH 11, the conversion of CrPO4·6 H2O to Cr(OH)3·3 H2O results in similar oxidation risks for both Cr(III) precipitates.

15.
Water Res ; 265: 122221, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39128334

RESUMEN

Low molecular weight organic acids (LMWOA) are commonly present in natural water and play a pivotal role in the reduction of Cr(VI). In frozen solutions, the efficiency of Cr(VI) reduction is significantly enhanced due to the freezing concentration effect. However, this facilitation is found to be contingent upon the functional groups of LMWOA in this study. To be specific, LMWOA and Cr(VI) can form five-membered ring complexes, which greatly enhance electron transfer efficiency through Ligand-to-Metal Charge Transfer (LMCT). DFT calculations indicate that oxygen-containing groups located on carbon atoms at α positions play a crucial role in forming these complexes, ultimately determining the kinetics of Cr(VI) reduction. Moreover, freezing not only increases proton concentrations but also reduces free water molecule content in the liquid-like layer (LLL), thereby affecting LMWOA species through regulation of protonation and hydrolysis, and subsequently impacting reaction mechanisms. The stoichiometric ratios between LMWOA and Cr(VI) exceed theoretical values due to complexation with Cr(III). The reduction of Cr(VI) by LMWOA in frozen solutions is inhibited by soil solution, while the degree of inhibition varies among different types of LMWOA.


Asunto(s)
Cromo , Cromo/química , Cinética , Congelación , Peso Molecular , Oxidación-Reducción , Teoría Funcional de la Densidad , Soluciones
16.
J Agric Food Chem ; 72(31): 17176-17190, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39067070

RESUMEN

Water scarcity and land infertility pose significant challenges to agricultural development, particularly in arid and semiarid regions. Improving soil-water-retention capacity and fertilizer utilization efficiency through the application of soil additives has become a pivotal approach in agricultural practices. Hydrogels exhibit exceptional water absorption and fertilizer retention capabilities, making them extensively utilized in the fields of agriculture, forestry, and desert control. Currently, most reviews primarily focus on the raw materials, classification, synthesis methods, and application prospects of hydrogels, with limited attention given to strategies for enhancing water-retention performance, mechanisms underlying fertilizer absorption, and environmental risks. This review covers the commonly used cross-linking methods in hydrogel synthesis and the structure-activity relationship between hydrogels and water as well as fertilizer. Additionally, a thorough analysis of the ecological benefits and risks associated with hydrogels is presented. Finally, future prospects and challenges are delineated from the perspectives of material design and engineering applications.


Asunto(s)
Fertilizantes , Hidrogeles , Suelo , Agua , Fertilizantes/análisis , Hidrogeles/química , Agua/química , Suelo/química , Agricultura/métodos
17.
Nat Commun ; 15(1): 7350, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39187567

RESUMEN

Owing to their capability to produce reactive oxygen species (ROS) under solar irradiation, covalent organic frameworks (COFs) with pre-designable structure and unique architectures show great potentials for water purification. However, the sluggish charge separation, inefficient oxygen activation and poor structure stability in COFs restrict their practical applications to decontaminate water. Herein, via a facile one-pot synthetic strategy, we show the direct conversion of reversible imine linkage into rigid thiazole linkage can adjust the π-conjugation and local charge polarization of skeleton to boost the exciton dissociation on COFs. The rigid linkage can also improve the robustness of skeleton and the stability of COFs during the consecutive utilization process. More importantly, the thiazole linkage in COFs with optimal C 2p states (COF-S) effectively increases the activities of neighboring benzene unit to directly modulate the O2-adsorption energy barrier and improve the ROS production efficiency, resulting in the excellent photocatalytic degradation efficiency of seven toxic emerging contaminants (e.g. degrading ~99% of 5 mg L-1 paracetamol in only 7 min) and effective bacterial/algal inactivation performance. Besides, COF-S can be immobilized in continuous-flow reactor and in enlarged reactor to efficiently eliminate pollutants under natural sunlight irradiation, demonstrating the feasibility for practical application.

18.
Environ Pollut ; 341: 123000, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38000728

RESUMEN

Polyethylene (PE) microplastics are emerging pollutants that pose a significant threat to the environment and human health. However, little is known about the effects of PEs on soil‒plant interactions, especially in heavy metal (HM)-contaminated soil. In this study, the effects of PE on rhizosphere soil enzyme activities, microbial interactions and nutrient cycling processes were analyzed from ecological network and functional gene perspectives for the first time. The results indicated that PE-MP addition significantly reduced the biomass of Bidens pilosa L. In addition, the partial increase in carbon, nitrogen, and phosphorus enzyme activities suggested that the effects of PE as a carbon source on microbial functions in HM-contaminated soil should not be ignored. The average path length of bacterial network nodes was found to be higher than that of fungal network nodes, demonstrating that the bacterial ecological network in PE-MP and HM cocontaminated environments has good buffering capacity against changes in external environmental conditions. Furthermore, structural equation modeling demonstrated that particle size and dosage affect soil nutrient cycling processes and that cycling processes are acutely aware of changes in any factor, such as soil moisture, soil pH and soil nitrogen nutrients. Hence, PE-MP addition in HM-contaminated soil has the potential to alter soil ecological functions and nutrient cycles.


Asunto(s)
Metales Pesados , Contaminantes del Suelo , Humanos , Suelo/química , Microplásticos , Plásticos , Polietileno , Metales Pesados/toxicidad , Nitrógeno , Carbono , Contaminantes del Suelo/toxicidad , Contaminantes del Suelo/análisis , Microbiología del Suelo
19.
Int J Biol Macromol ; 274(Pt 2): 133385, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38914402

RESUMEN

This study synthesized a carboxymethyl chitosan-modified bimetallic Co/Zn-ZIF (CZ@CMC) with strong hydrophilicity and adsorption performance via the one-pot method. Tetracycline hydrochloride (TCH) was used as the model contaminant to evaluate the adsorption and peroxymonosulfate (PMS) activation properties of CZ@CMC. Mechanism showed that the adsorption behavior occurred through pore filling, electrostatic attraction, surface complexation, hydrogen bonding, and π-π stacking. In addition, a CZ@CMC/PMS system was constructed, which had excellent catalytic performance. The hydrophilicity and selective adsorption properties of CMC conferred a greatly accelerated CZ@CMC in catalyzing the PMS process with kobs of 0.095 min-1, in which OH, 1O2, SO4-, O2-, and Co(III) were the main ROS which quenching tests, EPR, and chemical probe experiments verified. In addition, the degradation pathways of TCH were obtained utilizing DFT and HPLC-MS and analyzed to show that the system possessed a good detoxification capacity. This work is expected to provide a green, efficient, and stable strategy to enhance the adsorption properties of catalytic materials and subsequently their co-catalytic properties.


Asunto(s)
Quitosano , Cobalto , Estructuras Metalorgánicas , Tetraciclina , Zinc , Quitosano/química , Quitosano/análogos & derivados , Cobalto/química , Tetraciclina/química , Adsorción , Estructuras Metalorgánicas/química , Zinc/química , Catálisis , Contaminantes Químicos del Agua/química , Contaminantes Químicos del Agua/aislamiento & purificación , Purificación del Agua/métodos , Interacciones Hidrofóbicas e Hidrofílicas , Peróxidos
20.
Stem Cell Rev Rep ; 20(6): 1569-1586, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38713406

RESUMEN

Although stem/progenitor cell therapy shows potential for myocardial infarction repair, enhancing the therapeutic efficacy could be achieved through additional genetic modifications. HCLS1-associated protein X-1 (HAX1) has been identified as a versatile modulator responsible for cardio-protective signaling, while its role in regulating stem cell survival and functionality remains unknown. In this study, we investigated whether HAX1 can augment the protective potential of Sca1+ cardiac stromal cells (CSCs) for myocardial injury. The overexpression of HAX1 significantly increased cell proliferation and conferred enhanced resistance to hypoxia-induced cell death in CSCs. Mechanistically, HAX1 can interact with Mst1 (a prominent conductor of Hippo signal transduction) and inhibit its kinase activity for protein phosphorylation. This inhibition led to enhanced nuclear translocation of Yes-associated protein (YAP) and activation of downstream therapeutic-related genes. Notably, HAX1 overexpression significantly increased the pro-angiogenic potential of CSCs, as demonstrated by elevated expression of vascular endothelial growth factors. Importantly, implantation of HAX1-overexpressing CSCs promoted neovascularization, protected against functional deterioration, and ameliorated cardiac fibrosis in ischemic mouse hearts. In conclusion, HAX1 emerges as a valuable and efficient inducer for enhancing the effectiveness of cardiac stem or progenitor cell therapeutics.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proliferación Celular , Vía de Señalización Hippo , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Proteínas Señalizadoras YAP , Animales , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ratones , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Proliferación Celular/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Humanos , Infarto del Miocardio/terapia , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/genética , Neovascularización Fisiológica , Células Madre/metabolismo , Células Madre/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Trasplante de Células Madre , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/genética , Proteínas Proto-Oncogénicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA