Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44.391
Filtrar
1.
Anim Biotechnol ; 35(1): 2391520, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39222080

RESUMEN

MicroRNAs (miRNAs) were identified to be involved in various biological functions by regulating the degradation or suppressing the translation of their downstream target genes. Recent studies have identified miR-29a play a key role in functions of mammal cell differentiation, proliferation, apoptosis, and signal transduction. However, the underlying functions for miR-29a in jejunal epithelial cells biological function still to be investigated. In order to explore the yak jejunal epithelial cells proliferation and barrier dysfunction with over expression of miR-29a gene, three 0-day-old Pamir male yaks were randomly selected and slaughtered in present study, and the jejunal epithelial cells were isolated and cultured to determine yak jejunal epithelial cells proliferation and protein composition on differential expression of miR-29a gene in Pamir plateau. Here, we demonstrated that the overexpression of miR-29a gene could inhibit the proliferation of Pamir yaks jejunum epithelial cells, and contribute to the apoptosis of Pamir yaks jejunal epithelial cells with some extent. A total of 133 differentially expressed proteins were identified in different expression of miR-29a groups by label-free Mass Spectrometry (MS), which could be concluded to two predominant themes: cell proliferation and inflammatory response. Interestingly, GPR41, as a bridge protein, was contacted two predominant themes to involved in Pamir Yaks jejunal mechanical barrier PPI network, and the target proteins displayed strong mutual interactions in the complex PPI network. Overall, our study suggested that the over-expression miR-29a inhibited the jejunal epithelial cells proliferation and the expressions of specific proteins, which damaged jejunal barrier function to slow down the intestine structure and function advanced mature development during young livestock period for influence the enhanced performance of production efficiency.


Asunto(s)
Apoptosis , Proliferación Celular , Células Epiteliales , Yeyuno , MicroARNs , Animales , Bovinos/genética , MicroARNs/genética , MicroARNs/metabolismo , Células Epiteliales/fisiología , Células Epiteliales/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Yeyuno/citología , Yeyuno/metabolismo , Proliferación Celular/genética , Masculino
2.
CNS Neurosci Ther ; 30(9): e70026, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39223758

RESUMEN

AIMS: Incidence of acute mountain sickness (AMS) ranges from 40%-90%, with high-altitude cerebral edema (HACE) representing a life-threatening end stage of severe AMS. However, practical and convenient preventive strategies for HACE are lacking. Remote ischemic preconditioning (RIPC) has demonstrated preventive effects on ischemia- or hypoxia-induced cardiovascular and cerebrovascular diseases. This study aimed to investigate the potential molecular mechanism of HACE and the application of RIPC in preventing HACE onset. METHODS: A hypobaric hypoxia chamber was used to simulate a high-altitude environment of 7000 meters. Metabolomics and metabolic flux analysis were employed to assay metabolite levels. Transcriptomics and quantitative real-time PCR (q-PCR) were used to investigate gene expression levels. Immunofluorescence staining was performed on neurons to label cellular proteins. The fluorescent probes Mito-Dendra2, iATPSnFR1.0, and CMTMRos were used to observe mitochondria, ATP, and membrane potential in cultured neurons, respectively. TUNEL staining was performed to detect and quantify apoptotic cell death. Hematoxylin and eosin (H&E) staining was utilized to analyze pathological changes, such as tissue swelling in cerebral cortex samples. The Rotarod test was performed to assess motor coordination and balance in rats. Oxygen-glucose deprivation (OGD) of cultured cells was employed as an in vitro model to simulate the hypoxia and hypoglycemia induced by RIPC in animal experiments. RESULTS: We revealed a causative perturbation of glucose metabolism in the brain preceding cerebral edema. Ischemic preconditioning treatment significantly reprograms glucose metabolism, ameliorating cell apoptosis and hypoxia-induced energy deprivation. Notably, ischemic preconditioning improves mitochondrial membrane potential and ATP production through enhanced glucose-coupled mitochondrial metabolism. In vivo studies confirm that RIPC alleviates cerebral edema, reduces cell apoptosis induced by high-altitude hypoxia, and improves motor dysfunction resulting from cerebral edema. CONCLUSIONS: Our study elucidates the metabolic basis of HACE pathogenesis. This study provides a new strategy for preventing HACE that RIPC reduces brain edema through reprogramming metabolism, highlighting the potential of targeting metabolic reprogramming for neuroprotective interventions in neurological diseases caused by ischemia or hypoxia.


Asunto(s)
Mal de Altura , Edema Encefálico , Glucosa , Precondicionamiento Isquémico , Ratas Sprague-Dawley , Animales , Edema Encefálico/prevención & control , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Glucosa/metabolismo , Precondicionamiento Isquémico/métodos , Masculino , Mal de Altura/prevención & control , Mal de Altura/metabolismo , Ratas , Células Cultivadas , Neuronas/metabolismo , Neuronas/patología , Apoptosis/fisiología , Reprogramación Metabólica
3.
J Orthop Surg Res ; 19(1): 550, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39252111

RESUMEN

Osteoarthritis (OA) is a chronic joint disease characterized by the degeneration, destruction, and excessive ossification of articular cartilage. The prevalence of OA is rising annually, concomitant with the aging global population and increasing rates of obesity. This condition imposes a substantial and escalating burden on individual health, healthcare systems, and broader social and economic frameworks. The etiology of OA is multifaceted and not fully understood. Current research suggests that the death of chondrocytes, encompassing mechanisms such as cellular apoptosis, pyroptosis, autophagy, ferroptosis and cuproptosis, contributes to both the initiation and progression of the disease. These cell death pathways not only diminish the population of chondrocytes but also exacerbate joint damage through the induction of inflammation and other deleterious processes. This paper delineates the morphological characteristics associated with various modes of cell death and summarizes current research results on the molecular mechanisms of different cell death patterns in OA. The objective is to review the advancements in understanding chondrocyte cell death in OA, thereby offering novel insights for potential clinical interventions.


Asunto(s)
Muerte Celular , Condrocitos , Progresión de la Enfermedad , Osteoartritis , Condrocitos/patología , Humanos , Osteoartritis/patología , Osteoartritis/terapia , Muerte Celular/fisiología , Apoptosis/fisiología , Cartílago Articular/patología , Autofagia/fisiología , Animales , Piroptosis/fisiología , Ferroptosis/fisiología
4.
Physiol Res ; 73(4): 577-591, 2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39264079

RESUMEN

Xi-Kun Yuan Pin-Shi Ni Zhen-Hao Yan Zhi Yu Zhuang-Zhi Wang Chen-Kai Zhang Fang-Hui Li Xiao-Ming Yu 1Sports Department, Nanjing University of Science and Technology ZiJin College, Nanjing, China, 2School of Sport Sciences, Nanjing Normal University, Nanjing, China, 3Shanghai Seventh People's Hospital, Shanghai, China To investigate the effects of life-long exercise (LLE) on age-related inflammatory cytokines, apoptosis, oxidative stress, ferroptosis markers, and the NRF2/KAEP 1/Klotho pathway in rats. Eight-month-old female Sprague-Dawley rats were divided into four groups: 1) LLE: 18-month LLE training starting at 8 months of age, 2) Old moderate-intensity continuous training (OMICT): 8 months of moderate-intensity continuous training starting at 18 months of age, 3) Adult sedentary (ASED): 8 month-old adult sedentary control group, and 4) Old sedentary (OSED): a 26-month-old sedentary control group. Hematoxylin eosin staining was performed to observe the pathological changes of kidney tissue injury in rats; Masson's staining to observe the deposition of collagen fibers in rat kidney tissues; and western blotting to detect the expression levels of IL-6, IL 1beta, p53, p21, TNF-alpha, GPX4, KAEP 1, NRF2, SLC7A11, and other proteins in kidney tissues. Results: Compared with the ASED group, the OSED group showed significant morphological changes in renal tubules and glomeruli, which were swollen and deformed, with a small number of inflammatory cells infiltrated in the tubules. Compared with the OSED group, the expression levels of inflammation-related proteins such as IL-1beta, IL-6, TNF alpha, and MMP3 were significantly lower in the LLE group. Quantitative immunofluorescence analysis and western blotting revealed that compared with the ASED group, KAEP 1 protein fluorescence intensity and protein expression levels were significantly enhanced, while Klotho and NRF2 protein fluorescence intensity and protein expression levels were reduced in the OSED group. Compared with the OSED group, KAEP 1 protein fluorescence intensity and protein expression levels were reduced in the LLE and OMICT groups. Klotho and KAEP 1 protein expression levels and immunofluorescence intensity were higher in the LLE group than in the OSED group. The expression levels of GPX4 and SLC7A11, two negative marker proteins associated with ferroptosis, were significantly higher in the LLE group than in the OSED group, while the expression of p53 a cellular senescence-associated protein that negatively regulates SLC7A11, and the downstream protein p21 were significantly decreased. LLE may ameliorated aging-induced oxidative stress, inflammatory response, apoptosis, and ferroptosis by regulating Klotho and synergistically activating the NRF2/KAEP 1 pathway. Keywords: Life-long exercise, Moderate intensity continuous training, Aging, Kidney tissue, Ferroptosis.


Asunto(s)
Apoptosis , Ferroptosis , Riñón , Proteínas Klotho , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Condicionamiento Físico Animal , Ratas Sprague-Dawley , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/fisiología , Femenino , Apoptosis/fisiología , Ratas , Ferroptosis/fisiología , Riñón/metabolismo , Riñón/patología , Condicionamiento Físico Animal/fisiología , Envejecimiento/metabolismo , Envejecimiento/patología , Inflamación/metabolismo , Inflamación/patología , Transducción de Señal/fisiología , Glucuronidasa/metabolismo , Biomarcadores/metabolismo
5.
Pathol Res Pract ; 262: 155564, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39216322

RESUMEN

Colorectal cancer (CRC) remains a significant global health challenge, marked by increasing incidence and mortality rates in recent years. The pathogenesis of CRC is complex, involving chronic inflammation of the intestinal mucosa, heightened immunoinflammatory responses, and resistance to apoptosis. The suppressor of cytokine signaling (SOCS) family, comprised of key negative regulators within cytokine signaling pathways, plays a crucial role in cell proliferation, growth, and metabolic regulation. Deficiencies in various SOCS proteins can trigger the activation of the Janus kinase (JAK) and signal transducers and activators of transcription (STAT) pathways, following the binding of cytokines and growth factors to their receptors. Mounting evidence indicates that SOCS proteins are integral to the development and progression of CRC, positioning them as promising targets for novel anticancer therapies. This review delves into the structure, function, and molecular mechanisms of SOCS family members, examining their roles in cell proliferation, apoptosis, migration, epithelial-mesenchymal transition (EMT), and immune modulation. Additionally, it explores their potential impact on the regulation of CRC immunotherapy, offering new insights and perspectives that may inform the development of innovative therapeutic strategies for CRC.


Asunto(s)
Neoplasias Colorrectales , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/inmunología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Transducción de Señal/fisiología , Transición Epitelial-Mesenquimal/fisiología , Proliferación Celular/fisiología , Apoptosis/fisiología , Animales
6.
Brain Res Bull ; 216: 111049, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39142444

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder distinguished by gradual depletion of motor neurons. RNA binding motif protein 5 (RBM5), an abundantly expressed RNA-binding protein, plays a critical role in the process of cellular death. However, little is known about the role of RBM5 in the pathogenesis of ALS. Here, we found that RBM5 was upregulated in ALS hSOD1G93A-NSC34 cell models and hSOD1G93A mice due to a reduction of miR-141-5p. The upregulation of RBM5 increased the apoptosis of motor neurons by inhibiting Rac1-mediated neuroprotection. In contrast, genetic knockdown of RBM5 rescued motor neurons from hSOD1G93A-induced degeneration by activating Rac1 signaling. The neuroprotective effect of RBM5-knockdown was significantly inhibited by the Rac1 inhibitor, NSC23766. These findings suggest that RBM5 could potentially serve as a therapeutic target in ALS by activating the Rac1 signalling.


Asunto(s)
Esclerosis Amiotrófica Lateral , Apoptosis , Neuronas Motoras , Proteínas Proto-Oncogénicas c-akt , Proteínas de Unión al ARN , Transducción de Señal , Proteína de Unión al GTP rac1 , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Apoptosis/fisiología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Ratones , Humanos , Transducción de Señal/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones Transgénicos , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/genética , Masculino , Proteínas de Unión al ADN , Proteínas de Ciclo Celular , Proteínas Supresoras de Tumor
7.
J Dent Res ; 103(9): 937-947, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-39104161

RESUMEN

Alveolar bone (AB) remodeling, including formation and absorption, is the foundation of orthodontic tooth movement (OTM). However, the sources and mechanisms underlying new bone formation remain unclear. Therefore, we aimed to understand the potential mechanism of bone formation during OTM, focusing on the leptin receptor+ (Lepr+) osteogenitors and periodontal ligament cells (PDLCs). We demonstrated that Lepr+ cells activated by force-induced PDLC apoptosis served as distinct osteoprogenitors during orthodontic bone regeneration. We investigated bone formation both in vivo and in vitro. Single-cell RNA sequencing analysis and lineage tracing demonstrated that Lepr represents a subcluster of stem cells that are activated and differentiate into osteoblasts during OTM. Targeted ablation of Lepr+ cells in a mouse model disrupted orthodontic force-guided bone regeneration. Furthermore, apoptosis and sequential fluorescent labeling assays revealed that the apoptosis of PDLCs preceded new bone deposition. We found that PDL stem cell-derived apoptotic vesicles activated Lepr+ cells in vitro. Following apoptosis inhibition, orthodontic force-activated osteoprogenitors and osteogenesis were significantly downregulated. Notably, we found that bone formation occurred on the compression side during OTM; this has been first reported here. To conclude, we found a potential mechanism of bone formation during OTM that may provide new insights into AB regeneration.


Asunto(s)
Apoptosis , Osteogénesis , Ligamento Periodontal , Receptores de Leptina , Técnicas de Movimiento Dental , Ligamento Periodontal/citología , Animales , Apoptosis/fisiología , Ratones , Técnicas de Movimiento Dental/métodos , Osteogénesis/fisiología , Células Madre/fisiología , Regeneración Ósea/fisiología , Osteoblastos , Diferenciación Celular , Proceso Alveolar/citología
8.
Balkan Med J ; 41(5): 348-356, 2024 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-39129512

RESUMEN

Background: Zinc (Zn), an essential micronutrient, regulates and maintains neurological functions. However, both Zn deficiency and excess can cause oxidative stress and neurodegenerative diseases. As previously reported, immunoglobulin G (IgG) can modulate oxidative stress in various disorders. Aims: To investigate whether IgG treatment can alleviate oxidative stress caused by Zn0 on microglia in vitro. Study Design: In vitro study. Methods: The feasibility of Zn0 treatment was evaluated using the MTS assay. Oxidative stress following treatment with Zn0, either alone or with IgG supplementation, was determined with dihydrorhodamine 123 staining. Flow cytometry was employed to ascertain the intracellular protein levels of TRIM21, PINK, PARKIN, MFN2, Beclin-1, and active LC3B. Methods: The feasibility of Zn0 treatment was evaluated using the MTS assay. Oxidative stress following treatment with Zn0, either alone or with IgG supplementation, was determined with dihydrorhodamine 123 staining. Flow cytometry was employed to ascertain the intracellular protein levels of TRIM21, PINK, PARKIN, MFN2, Beclin-1, and active LC3B. Results: In silico screening confirmed the association between Zn0 cytotoxicity and apoptosis. Furthermore, oxidative stress was identified as a critical mechanism that underlies Zn0 neurotoxicity. The in silico analysis revealed that Zn can interact with the constant region of the Ig heavy chain, suggesting a potential role for IgG in alleviating Zn0-induced cytotoxicity. Experimental findings supported this hypothesis, as IgG administration significantly reduced Zn0-induced mitochondrial stress in a dose-dependent manner. The upregulation of PINK1 levels by Zn0 exposure suggests that mitochondrial injury promotes mitophagy. Interestingly, Zn0 decreased TRIM21 levels, which is reversed by IgG administration. Conclusion: These findings elucidate the cellular responses to Zn0 and highlight the potential use of intravenous immunoglobulin in mitigating the adverse effects of acute Zn0 exposure.


Asunto(s)
Inmunoglobulina G , Microglía , Estrés Oxidativo , Inmunoglobulina G/farmacología , Microglía/efectos de los fármacos , Microglía/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Zinc/farmacología , Zinc/análisis , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Humanos , Técnicas In Vitro , Animales , Citometría de Flujo/métodos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología
9.
J Orthop Surg Res ; 19(1): 467, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39118123

RESUMEN

BACKGROUND: Osteosarcoma is a soft tissue neoplasm with elevated recurrence risk and highly metastatic potential. Metal response element binding transcriptional factor 2 (MTF2) has been revealed to exert multiple activities in human tissues. The present research was conducted to explore the functions and related response mechanism of MTF2 in osteosarcoma which have not been introduced yet. METHODS: Bioinformatics tools identified the differential MTF2 expression in osteosarcoma tissues. MTF2 expression in osteosarcoma cells was examined with Western blot. Cell Counting Kit-8 (CCK-8) assay, 5-Ethynyl-2'-deoxyuridine (EDU) staining, wound healing as well as transwell assays measured cell proliferation, migration and invasion, respectively. Flow cytometry assay detected the cellular apoptotic level. Western blot also measured the expressions of proteins associated with epithelial mesenchymal transition (EMT), apoptosis and enhancer of zeste homolog 2 (EZH2)/secreted frizzled-related protein 1 (SFRP1)/Wnt signaling. Co-immunoprecipitation (Co-IP) assay confirmed MTF2-EZH2 interaction. RESULTS: MTF2 expression was increased in osteosarcoma tissues and cells. MTF2 interference effectively inhibited the proliferation, migration and invasion of osteosarcoma cells and promoted the cellular apoptotic rate. MTF2 directly bound to EZH2 and MTF2 silence reduced EZH2 expression, activated SFRP1 expression and blocked Wnt signaling in osteosarcoma cells. EZH2 upregulation or SFRP1 antagonist WAY-316606 partly counteracted the impacts of MTF2 down-regulation on the SFRP1/Wnt signaling and the biological phenotypes of osteosarcoma cells. CONCLUSIONS: MTF2 might down-regulate SFRP1 to activate Wnt signaling and drive the progression of osteosarcoma via interaction with EZH2 protein.


Asunto(s)
Neoplasias Óseas , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2 , Osteosarcoma , Vía de Señalización Wnt , Humanos , Apoptosis/fisiología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Progresión de la Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Vía de Señalización Wnt/fisiología , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo
10.
Placenta ; 155: 32-41, 2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39121585

RESUMEN

INTRODUCTION: Preeclampsia is a pregnancy-specific disorder characterized by de novo development of hypertension and proteinuria over 20 weeks gestation that has been associated with the dysfunction of trophoblasts. Current evidence suggests that syncytin-1 plays an important role in the non-fusogenic biological activity of trophoblasts, except for specific fusogenic function. However, the underlying mechanism remains unclear. METHODS: The expression and location of syncytin-1 in normal and the late-onset preeclampsia placentas were detected by quantitative real-time PCR, western blotting and immunofluorescence. Morphological and apoptosis analysis were processed in placentas. The ex vivo extravillous explant culture model was used to explore the effect of syncytin-1 on EVT outgrowths. Real-time quantitative PCR and immunoblotting were used to calculate syncytin-1 levels in the trophoblast cells before and after syncytin-1 knockdown or overexpression. CCK-8 assay was used to detect the cell viability. TUNEL staining and immunoblotting were processed in trophoblast cells. Transwell assays and wound healing assays were utilize to assess the invasion and migration of trophoblastic cells. Conditional knockout of syncytin-a mouse model was conducted to present the change of placentas in vivo. The ex vivo extravillous explant culture model was used to explore the effect of syncytin-1 on EVT outgrowths. Western blotting was used to identify the key proteins of PI3K/Akt pathways and invasion-related proteins in trophoblast cells. RESULTS AND DISCUSSION: Here, reduced syncytin-1 was identified in the late-onset preeclampsia placentas. Reduced syncytin-1 may attenuates the EMT process by promoting apoptosis, inhibiting proliferation and invasion by suppressed PI3K/Akt pathway in trophoblast cells. Our findings provide novel insights into the non-fusogenic biological function of reduced syncytin-1 that may be involves in the pathogenesis of preeclampsia.


Asunto(s)
Apoptosis , Productos del Gen env , Preeclampsia , Proteínas Gestacionales , Trofoblastos , Femenino , Preeclampsia/metabolismo , Preeclampsia/patología , Preeclampsia/genética , Embarazo , Trofoblastos/metabolismo , Trofoblastos/patología , Apoptosis/fisiología , Proteínas Gestacionales/metabolismo , Proteínas Gestacionales/genética , Humanos , Animales , Productos del Gen env/metabolismo , Productos del Gen env/genética , Ratones , Placenta/metabolismo , Placenta/patología , Adulto , Ratones Noqueados , Movimiento Celular/fisiología , Transducción de Señal/fisiología
12.
Placenta ; 155: 42-51, 2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39121586

RESUMEN

INTRODUCTION: Trophoblast homeostasis and differentiation require a proper endoplasmic reticulum (ER) function. The Krüppel-like factor-6 (KLF6) transcription factor modulates trophoblast migration, differentiation, and reactive oxygen species (ROS) production. Since ROS may impact on ER homeostasis, we assessed whether downregulation of KLF6 altered the unfolded protein response (UPR) and cellular process associated with ER homeostasis. MATERIALS AND METHODS: Protein and RNA expression were analyzed by Western blot and qRT-PCR, respectively, in extravillous trophoblast HTR-8/SVneo cells silenced for KLF6. Apoptosis was detected by flow cell cytometry using Annexin V Apoptosis Detection Kit. Protein trafficking was assessed by confocal microscopy of a reporter fluorescent protein whose release from the ER was synchronized. RESULTS: KLF6 downregulation reduced the expression of BiP, the master regulator of the UPR, at protein, mRNA, and pre-mRNA levels. Ire1α protein, XBP1 splicing, and DNAJB9 mRNA levels were also reduced in KLF6-silenced cells. Instead, PDI, Ero1α, and the p-eIF2α/eIF2α ratio as well as autophagy and proteasome dependent protein degradation remained unchanged while intracellular trafficking was increased. Under thapsigargin-induced stress, KLF6 silencing impaired BiP protein and mRNA expression increase, as well as the activation of the Ire1α pathway, but it raised the p-eIF2α/eIF2α ratio and CHOP protein levels. Nevertheless, apoptosis was not increased. DISCUSSION: Results provide the first evidence of KLF6 as a modulator of the UPR components. The increase in protein trafficking and protection from apoptosis, observed in KLF6-silenced cells, are consistent with its role in extravillous trophoblast migration and differentiation.


Asunto(s)
Apoptosis , Retículo Endoplásmico , Factor 6 Similar a Kruppel , Trofoblastos , Respuesta de Proteína Desplegada , Humanos , Trofoblastos/metabolismo , Trofoblastos/fisiología , Factor 6 Similar a Kruppel/metabolismo , Factor 6 Similar a Kruppel/genética , Retículo Endoplásmico/metabolismo , Respuesta de Proteína Desplegada/fisiología , Apoptosis/fisiología , Homeostasis , Línea Celular , Femenino , Estrés del Retículo Endoplásmico/fisiología , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Trofoblastos Extravellosos
13.
Exp Eye Res ; 247: 110027, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39127238

RESUMEN

Corneal injuries often lead to epithelial damage, apoptosis, and inflammation which impact visual function. Effective epithelial healing is critical for optimal vision and functioning of the cornea. Mesenchymal stem/stromal cells (MSCs)-derived extracellular vesicles (EVs) present promising avenues for cell-free therapy, however, evaluation of their specific roles in corneal epithelial injury requires further investigations with due consideration to the endogenous human corneal epithelial cell-derived EVs (HCEC-EVs). This study aims to isolate and characterize the EVs from a commonly available human corneal epithelial cell line (HCE-2 [50. B1], ATCC) and evaluate their corneal epithelial repair, anti-apoptotic, and immunomodulatory potential in comparison with human bone marrow mesenchymal stem cell-derived EVs (BM-MSC-EVs) in vitro. Both the BM-MSC- and HCEC-EVs exhibited similar morphology with a diameter <150 nm. However, the yield of EVs from HCECs was higher than that of BM-MSCs. Nanoparticle tracking analysis revealed an average EV size of ∼120 nm, while western blotting confirmed the presence of CD63, CD81, and TSG101, whereas Calnexin could not be detected in the BM-MSC- and HCEC-EVs. The corneal epithelial repair was monitored through in vitro wound healing assay, whereas apoptosis was studied through flow cytometry-based Propidium iodide staining in H2O2-treated cells. IL-1ß-stimulated HCECs were treated with BM-MSC- and HCEC-EVs for 24 h and expression of pro- (IL-6 and TNF-α) and anti-inflammatory (IL-10 and TGF-ß) cytokines was evaluated through ELISA. Our results, limited to in vitro investigations, suggest that compared with HCEC-EVs, BM-MSC-EVs showed: i) accelerated corneal epithelial healing, ii) enhanced anti-apoptotic potential, and iii) improved anti-inflammatory properties, in cultured HCECs.


Asunto(s)
Apoptosis , Epitelio Corneal , Vesículas Extracelulares , Inmunomodulación , Células Madre Mesenquimatosas , Cicatrización de Heridas , Humanos , Vesículas Extracelulares/metabolismo , Epitelio Corneal/metabolismo , Apoptosis/fisiología , Células Madre Mesenquimatosas/metabolismo , Inmunomodulación/fisiología , Cicatrización de Heridas/fisiología , Western Blotting , Células Cultivadas , Citocinas/metabolismo , Citometría de Flujo
14.
Brain Res Bull ; 216: 111055, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39173779

RESUMEN

BACKGROUND: Cerebral ischemia is characterized by its rapid onset and high rates of recurrence, morbidity, and mortality, with blood-brain barrier (BBB) permeability playing a vital role in brain injury. Therefore, it is important to understand the molecular mechanism which regulates the BBB during cerebral ischemia. MATERIALS AND METHODS: An in vitro model of oxygen-glucose deprivation (OGD) and an in vivo model of cerebral ischemia/reperfusion (I/R) were constructed. PD-1 overexpression vectors and vectors containing si-RNA were transfected and injected into in vitro and in vivo models. Western blotting, real-time quantitative PCR (qPCR), immunofluorescence (IF) analysis, and immunohistochemical staining were employed to evaluate the expression levels of programmed cell death-1 (PD-1), microglia M1 and M2 biomarkers, and tight junction proteins. Flow cytometry and ELISA were used to measure the levels of pro-inflammatory cytokines. The BBB permeability of brain tissues was evaluated by Evans blue dye (EBD) extravasation and transendothelial electrical resistance (TEER). Brain water content was measured to assess the extent of inflammatory exudation. The infarct volume and neurological severity score (NSS) were used to assess the severity of brain injury. Brain cell apoptosis was assessed by the TUNEL assay and hematoxylin-eosin (H&E) staining. RESULTS: PD-1 helped to convert the microglia M1 phenotype to the M2 phenotype and to reduce BBB permeability both in vitro and in vivo. Overexpression of PD-1 promoted a shift of the M1 phenotype to the M2 phenotype and reduced BBB permeability via the ERK and p38 MAPK signaling pathways. PD-1 reduced inflammatory exudation, BBB permeability, cell apoptosis, and brain injury in vivo. CONCLUSION: Our present study verified that PD-1 exerts an anti-inflammatory effect by converting the microglia M1 phenotype to the M2 phenotype, reducing BBB permeability, and thereby relieves brain injury caused by cerebral ischemia. PD-1 is potential therapeutic target for brain injury caused by cerebral ischemia.


Asunto(s)
Barrera Hematoencefálica , Isquemia Encefálica , Sistema de Señalización de MAP Quinasas , Microglía , Receptor de Muerte Celular Programada 1 , Daño por Reperfusión , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Animales , Microglía/metabolismo , Daño por Reperfusión/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Isquemia Encefálica/metabolismo , Masculino , Ratones , Sistema de Señalización de MAP Quinasas/fisiología , Apoptosis/fisiología , Ratones Endogámicos C57BL , Polaridad Celular/fisiología
15.
Mil Med ; 189(Supplement_3): 416-422, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39160884

RESUMEN

INTRODUCTION: Many trauma patients die from hemorrhagic shock in the military and civilian settings. Although two-thirds of hemorrhagic shock victims die of reasons other than exsanguination, such as the consequent cytokine storm, anti-inflammatory therapies failed to be utilized. Apoptotic cell-based treatments enhance innate ability to exert systemic immunomodulation as demonstrated in several clinical applications and hence might present a novel approach in hemorrhagic shock treatment. MATERIALS AND METHODS: Twenty-two rats underwent a pressure-controlled hemorrhagic shock model and followed up for 24 hours. An infusion of apoptotic cells (Allocetra-OTS, Enlivex Therapeutics Ltd, Nes Ziona, Israel) was administered to the treatment group. Hemodynamics, blood counts, biochemistry findings, and cytokine profile were compared to a saline-resuscitated control group. RESULTS: The treatment group's mean arterial pressure decreased from 94.8 mmHg to 28.2 mmHg, resulting in an 8.13 mg/dL increase in lactate and a 1.9 g/L decrease in hemoglobin, similar to the control group. White blood cells and platelets decreased more profoundly in the treatment group. A similar cytokine profile after 24 hours was markedly attenuated in the treatment group 2 hours after bleeding. Levels of pro-inflammatory cytokines such as interleukin (IL)-1a (28.4 pg/mL vs. 179.1 pg/mL), IL-1b (47.4 pg/mL vs. 103.9 pg/mL), IL-6 (526.2 pg/mL vs. 3492 pg/mL), interferon γ (11.4 pg/mL vs. 427.9 pg/mL), and tumor necrosis factor α (19.0 pg/mL vs. 31.7 pg/mL) were profoundly lower in the treatment group. CONCLUSION: In a pressure-control hemorrhagic shock model in rats, apoptotic cell infusion showed preliminary signs of a uniform attenuated cytokine response. Apoptotic cell-based therapies might serve as a novel immunomodulatory therapy for hemorrhagic shock.


Asunto(s)
Apoptosis , Choque Hemorrágico , Choque Hemorrágico/terapia , Choque Hemorrágico/complicaciones , Choque Hemorrágico/inmunología , Animales , Ratas , Masculino , Apoptosis/fisiología , Ratas Sprague-Dawley , Modelos Animales de Enfermedad , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Citocinas/sangre , Citocinas/análisis , Inflamación/terapia
16.
J Orthop Surg Res ; 19(1): 483, 2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39152465

RESUMEN

BACKGROUND: Effective bone formation relies on osteoblast differentiation, a process subject to intricate post-translational regulation. Ubiquitin-specific proteases (USPs) repress protein degradation mediated by the ubiquitin-proteasome pathway. Several USPs have been documented to regulate osteoblast differentiation, but whether other USPs are involved in this process remains elusive. METHODS: In this study, we conducted a comparative analysis of 48 USPs in differentiated and undifferentiated hFOB1.19 osteoblasts, identifying significantly upregulated USPs. Subsequently, we generated USP knockdown hFOB1.19 cells and evaluated their osteogenic differentiation using Alizarin red staining. We also assessed cell viability, cell cycle progression, and apoptosis through MTT, 7-aminoactinomycin D staining, and Annexin V/PI staining assays, respectively. Quantitative PCR and Western blotting were employed to measure the expression levels of osteogenic differentiation markers. Additionally, we investigated the interaction between the USP and its target protein using co-immunoprecipitation (co-IP). Furthermore, we depleted the USP in hFOB1.19 cells to examine its effect on the ubiquitination and stability of the target protein using immunoprecipitation (IP) and Western blotting. Finally, we overexpressed the target protein in USP-deficient hFOB1.19 cells and evaluated its impact on their osteogenic differentiation using Alizarin red staining. RESULTS: USP36 is the most markedly upregulated USP in differentiated hFOB1.19 osteoblasts. Knockdown of USP36 leads to reduced viability, cell cycle arrest, heightened apoptosis, and impaired osteogenic differentiation in hFOB1.19 cells. USP36 interacts with WD repeat-containing protein 5 (WDR5), and the knockdown of USP36 causes an increased level of WDR5 ubiquitination and accelerated degradation of WDR5. Excessive WDR5 improved the impaired osteogenic differentiation of USP36-deficient hFOB1.19 cells. CONCLUSIONS: These observations suggested that USP36 may function as a key regulator of osteoblast differentiation, and its regulatory mechanism may be related to the stabilization of WDR5.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Osteoblastos , Osteogénesis , Osteoblastos/metabolismo , Osteoblastos/citología , Diferenciación Celular/fisiología , Diferenciación Celular/genética , Humanos , Supervivencia Celular/fisiología , Supervivencia Celular/genética , Proliferación Celular/fisiología , Proliferación Celular/genética , Osteogénesis/fisiología , Osteogénesis/genética , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Línea Celular , Apoptosis/genética , Apoptosis/fisiología , Ubiquitinación , Técnicas de Silenciamiento del Gen
17.
Neurosurg Rev ; 47(1): 448, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39164434

RESUMEN

In this review, the author highlights the role of IL4 in mitigating all the "hallmarks" of cancer growth and resistance to current immunotherapy, providing a framework for its role in GBM as well as guideline for future treatment regimens. This review is organized around six strategies by which IL4 contributes to the immune resistance seen in GBM: (i) apoptosis evasion, (ii) self-sufficiency in growth signals, (iii) insensitivity to anti-growth signals, (iv) invasion and metastasis, (v) limitless replicative potential, (vi) sustained angiogenesis.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Interleucina-4 , Humanos , Glioblastoma/patología , Glioblastoma/inmunología , Neoplasias Encefálicas/patología , Inmunoterapia/métodos , Resistencia a Antineoplásicos , Neovascularización Patológica , Apoptosis/fisiología
18.
CNS Neurosci Ther ; 30(8): e14836, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39097918

RESUMEN

INTRODUCTION: Cerebral ischemia-reperfusion injury (CIRI) is a common and debilitating complication of cerebrovascular diseases such as stroke, characterized by mitochondrial dysfunction and cell apoptosis. Unraveling the molecular mechanisms behind these processes is essential for developing effective CIRI treatments. This study investigates the role of RACK1 (receptor for activated C kinase 1) in CIRI and its impact on mitochondrial autophagy. METHODS: We utilized high-throughput transcriptome sequencing and weighted gene co-expression network analysis (WGCNA) to identify core genes associated with CIRI. In vitro experiments used human neuroblastoma SK-N-SH cells subjected to oxygen and glucose deprivation (OGD) to simulate ischemia, followed by reperfusion (OGD/R). RACK1 knockout cells were created using CRISPR/Cas9 technology, and cell viability, apoptosis, and mitochondrial function were assessed. In vivo experiments involved middle cerebral artery occlusion/reperfusion (MCAO/R) surgery in rats, evaluating neurological function and cell apoptosis. RESULTS: Our findings revealed that RACK1 expression increases during CIRI and is protective by regulating mitochondrial autophagy through the PINK1/Parkin pathway. In vitro, RACK1 knockout exacerbated cell apoptosis, while overexpression of RACK1 reversed this process, enhancing mitochondrial function. In vivo, RACK1 overexpression reduced cerebral infarct volume and improved neurological deficits. The regulatory role of RACK1 depended on the PINK1/Parkin pathway, with RACK1 knockout inhibiting PINK1 and Parkin expression, while RACK1 overexpression restored them. CONCLUSION: This study demonstrates that RACK1 safeguards against neural damage in CIRI by promoting mitochondrial autophagy through the PINK1/Parkin pathway. These findings offer crucial insights into the regulation of mitochondrial autophagy and cell apoptosis by RACK1, providing a promising foundation for future CIRI treatments.


Asunto(s)
Autofagia , Mitocondrias , Proteínas Quinasas , Receptores de Cinasa C Activada , Daño por Reperfusión , Ubiquitina-Proteína Ligasas , Animales , Humanos , Ratas , Apoptosis/fisiología , Autofagia/fisiología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Línea Celular Tumoral , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/metabolismo , Mitocondrias/metabolismo , Proteínas de Neoplasias , Neuroprotección/fisiología , Proteínas Quinasas/metabolismo , Proteínas Quinasas/genética , Ratas Sprague-Dawley , Receptores de Cinasa C Activada/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética
19.
Physiol Rep ; 12(15): e16181, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39138135

RESUMEN

This study aimed to evaluate the influence of combined intermittent fasting (IF) and high-intensity interval training (HIIT) on morphology, caspase-independent apoptosis signaling pathway, and myostatin expression in soleus and gastrocnemius (white portion) muscles from healthy rats. Sixty-day-old male Wistar rats (n = 60) were divided into four groups: control (C), IF, high-intensity-interval training (T), and high-intensity-interval training and intermittent fasting (T-IF). The C and T groups received ad libitum chow daily; IF and T-IF received the same standard chow every other day. Animals from T and T-IF underwent a HIIT protocol five times a week for 12 weeks. IF reduced gastrocnemius mass and increased pro-apoptotic proteins apoptosis-inducing factor (AIF) and endonuclease G (EndoG) in soleus and cleaved-to-non-cleaved PARP-1 ratio and myostatin expression in gastrocnemius white portion. HIIT increased AIF and apoptosis repressor with caspase recruitment domain expression in soleus and cleaved-to-total PARP-1 ratio in gastrocnemius muscle white portion. The combination of IF and HIIT reduced fiber cross-sectional area in both muscles, increased EndoG and AIF expression, and decreased cleaved-to-non-cleaved PARP-1 ratio in gastrocnemius muscle white portion. Muscle responses to IF and HIIT are directly impacted by the muscle fiber type composition and are modulated, at least in part, by myostatin and caspase-independent apoptosis signaling.


Asunto(s)
Factor Inductor de la Apoptosis , Apoptosis , Ayuno , Entrenamiento de Intervalos de Alta Intensidad , Fibras Musculares de Contracción Lenta , Atrofia Muscular , Miostatina , Ratas Wistar , Transducción de Señal , Animales , Masculino , Apoptosis/fisiología , Ayuno/metabolismo , Ayuno/fisiología , Miostatina/metabolismo , Entrenamiento de Intervalos de Alta Intensidad/métodos , Ratas , Transducción de Señal/fisiología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Factor Inductor de la Apoptosis/metabolismo , Fibras Musculares de Contracción Lenta/metabolismo , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Rápida/patología , Endodesoxirribonucleasas/metabolismo , Condicionamiento Físico Animal/métodos , Condicionamiento Físico Animal/fisiología , Músculo Esquelético/metabolismo , Ayuno Intermitente , Poli(ADP-Ribosa) Polimerasa-1
20.
J Sex Med ; 21(8): 652-662, 2024 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-38972660

RESUMEN

BACKGROUND: Diabetes mellitus commonly causes endothelial cell and smooth muscle cell death in penile cavernous tissue. AIM: The study sought to study the mode of cell death in the penile cavernous tissue in type 1 diabetic rats. METHODS: A total of 36 Sprague Dawley rats 10 weeks of age were randomly divided into 2 groups: a normoglycemic group and type 1 diabetic group (intraperitoneal injection of Streptozotocin (STZ), 60 mg/kg). We randomly selected 6 rats from each group for tests at the end of 11, 14, and 18 weeks of age, respectively. All rats were able to eat and drink freely. The ratio of maximum intracavernous pressure to mean arterial pressure, concentration of serum testosterone, level of nitric oxide in the penile cavernosum, and expression of active caspase-1 (pyroptosis) and active caspase-3 (apoptosis) were determined. OUTCOMES: At the end of weeks 4 and 8 of type 1 diabetes, the proportions of endothelial cells and smooth muscle cells undergoing apoptosis and pyroptosis in penile cavernous tissue are different. RESULTS: The ratio of maximum intracavernous pressure to mean arterial pressure and nitric oxide levels were significantly lower in the 4- and 8-week diabetic groups than in the normoglycemic group (P < .01). Penile endothelial cell pyroptosis (5.67 ± 0.81%), smooth muscle cell apoptosis (23.72 ± 0.48%), total cell pyroptosis (9.67 ± 0.73%), and total apoptosis (10.52 ± 1.45%) were significantly greater in the 4-week diabetic group than in the normoglycemic group (P < .01). The proportion of endothelial cell pyroptosis (24.4 ± 3.69%), endothelial cell apoptosis (22.13 ± 2.43%), total cell pyroptosis (14.75 ± 0.93%), and total apoptosis (14.82 ± 1.08%) in the penile tissues of the 8-week diabetic group were significantly greater than those in the normoglycemic group (P < .01).The 8-week survival proportions of diabetic endothelial cells (38.86 ± 8.85%) and smooth muscle cells (44.46 ± 2.94%) was significantly lower than the 4-week survival proportions of endothelial cells (93.17 ± 8.07%) and smooth muscle cells (75.12 ± 4.76%) (P < .05). CLINICAL TRANSLATION: Inhibition of cell death by different methods at different stages may be the key to the treatment of type 1 diabetes-induced erectile dysfunction. STRENGTHS AND LIMITATIONS: The effect of type 1 diabetes on other types of cell death in penile cavernous tissue needs further study. CONCLUSION: The mode of death of endothelial cells in the cavernous tissue of the penis in the early stage in diabetic rats is dominated by pyroptosis, and the death of smooth muscle cells is dominated by apoptosis. Endothelial cell and smooth muscle cell death are not consistent at different stages of diabetes progression.


Asunto(s)
Apoptosis , Caspasa 3 , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Óxido Nítrico , Pene , Ratas Sprague-Dawley , Masculino , Animales , Pene/patología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/complicaciones , Ratas , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/patología , Caspasa 3/metabolismo , Apoptosis/fisiología , Óxido Nítrico/metabolismo , Piroptosis/fisiología , Testosterona/sangre , Caspasa 1/metabolismo , Células Endoteliales/patología , Muerte Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA