Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Nanobiotechnology ; 22(1): 318, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38849914

RESUMEN

Mitochondria occupy a central role in the biology of most eukaryotic cells, functioning as the hub of oxidative metabolism where sugars, fats, and amino acids are ultimately oxidized to release energy. This crucial function fuels a variety of cellular activities. Disruption in mitochondrial metabolism is a common feature in many diseases, including cancer, neurodegenerative conditions and cardiovascular diseases. Targeting tumor cell mitochondrial metabolism with multifunctional nanosystems emerges as a promising strategy for enhancing therapeutic efficacy against cancer. This review comprehensively outlines the pathways of mitochondrial metabolism, emphasizing their critical roles in cellular energy production and metabolic regulation. The associations between aberrant mitochondrial metabolism and the initiation and progression of cancer are highlighted, illustrating how these metabolic disruptions contribute to oncogenesis and tumor sustainability. More importantly, innovative strategies employing nanomedicines to precisely target mitochondrial metabolic pathways in cancer therapy are fully explored. Furthermore, key challenges and future directions in this field are identified and discussed. Collectively, this review provides a comprehensive understanding of the current state and future potential of nanomedicine in targeting mitochondrial metabolism, offering insights for developing more effective cancer therapies.


Asunto(s)
Mitocondrias , Nanomedicina , Neoplasias , Humanos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Nanomedicina/métodos , Animales , Metabolismo Energético/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Nanopartículas/química , Sistemas de Liberación de Medicamentos/métodos
2.
J Nanobiotechnology ; 22(1): 171, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38610017

RESUMEN

Salivary extracellular vesicles (EVs) have emerged as key tools for non-invasive diagnostics, playing a crucial role in the early detection and monitoring of diseases. These EVs surpass whole saliva in biomarker detection due to their enhanced stability, which minimizes contamination and enzymatic degradation. The review comprehensively discusses methods for isolating, enriching, quantifying, and characterizing salivary EVs. It highlights their importance as biomarkers in oral diseases like periodontitis and oral cancer, and underscores their potential in monitoring systemic conditions. Furthermore, the review explores the therapeutic possibilities of salivary EVs, particularly in personalized medicine through engineered EVs for targeted drug delivery. The discussion also covers the current challenges and future prospects in the field, emphasizing the potential of salivary EVs in advancing clinical practice and disease management.


Asunto(s)
Vesículas Extracelulares , Neoplasias de la Boca , Humanos , Medicina de Precisión , Sistemas de Liberación de Medicamentos , Saliva
3.
J Transl Med ; 21(1): 211, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36949458

RESUMEN

The human body is colonized by abundant and diverse microorganisms, collectively known as the microbiome. The oral cavity has more than 700 species of bacteria and consists of unique microbiome niches on mucosal surfaces, on tooth hard tissue, and in saliva. The homeostatic balance between the oral microbiota and the immune system plays an indispensable role in maintaining the well-being and health status of the human host. Growing evidence has demonstrated that oral microbiota dysbiosis is actively involved in regulating the initiation and progression of an array of autoimmune diseases.Oral microbiota dysbiosis is driven by multiple factors, such as host genetic factors, dietary habits, stress, smoking, administration of antibiotics, tissue injury and infection. The dysregulation in the oral microbiome plays a crucial role in triggering and promoting autoimmune diseases via several mechanisms, including microbial translocation, molecular mimicry, autoantigen overproduction, and amplification of autoimmune responses by cytokines. Good oral hygiene behaviors, low carbohydrate diets, healthy lifestyles, usage of prebiotics, probiotics or synbiotics, oral microbiota transplantation and nanomedicine-based therapeutics are promising avenues for maintaining a balanced oral microbiome and treating oral microbiota-mediated autoimmune diseases. Thus, a comprehensive understanding of the relationship between oral microbiota dysbiosis and autoimmune diseases is critical for providing novel insights into the development of oral microbiota-based therapeutic approaches for combating these refractory diseases.


Asunto(s)
Enfermedades Autoinmunes , Microbioma Gastrointestinal , Microbiota , Probióticos , Humanos , Disbiosis/microbiología , Boca/microbiología
4.
Cell Death Differ ; 31(1): 9-27, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37985811

RESUMEN

RNA modifications, known as the "epitranscriptome", represent a key layer of regulation that influences a wide array of biological processes in mesenchymal stem cells (MSCs). These modifications, catalyzed by specific enzymes, often termed "writers", "readers", and "erasers", can dynamically alter the MSCs' transcriptomic landscape, thereby modulating cell differentiation, proliferation, and responses to environmental cues. These enzymes include members of the classes METTL, IGF2BP, WTAP, YTHD, FTO, NAT, and others. Many of these RNA-modifying agents are active during MSC lineage differentiation. This review provides a comprehensive overview of the current understanding of different RNA modifications in MSCs, their roles in regulating stem cell behavior, and their implications in MSC-based therapies. It delves into how RNA modifications impact MSC biology, the functional significance of individual modifications, and the complex interplay among these modifications. We further discuss how these intricate regulatory mechanisms contribute to the functional diversity of MSCs, and how they might be harnessed for therapeutic applications. The review also highlights current challenges and potential future directions in the study of RNA modifications in MSCs, emphasizing the need for innovative tools to precisely map these modifications and decipher their context-specific effects. Collectively, this work paves the way for a deeper understanding of the role of the epitranscriptome in MSC biology, potentially advancing therapeutic strategies in regenerative medicine and MSC-based therapies.


Asunto(s)
Células Madre Mesenquimatosas , ARN , Diferenciación Celular/fisiología
5.
Clin Transl Med ; 14(5): e1705, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38797935

RESUMEN

Ribosomal RNA (rRNA) modifications, essential components of ribosome structure and function, significantly impact cellular proteomics and cancer biology. These chemical modifications transcend structural roles, critically shaping ribosome functionality and influencing cellular protein profiles. In this review, the mechanisms by which rRNA modifications regulate both rRNA functions and broader cellular physiological processes are critically discussed. Importantly, by altering the translational output, rRNA modifications can shift the cellular equilibrium towards oncogenesis, thus playing a key role in cancer development and progression. Moreover, a special focus is placed on the functions of mitochondrial rRNA modifications and their aberrant expression in cancer, an area with profound implications yet largely uncharted. Dysregulation in these modifications can lead to metabolic dysfunction and apoptosis resistance, hallmark traits of cancer cells. Furthermore, the current challenges and future perspectives in targeting rRNA modifications are highlighted as a therapeutic approach for cancer treatment. In conclusion, rRNA modifications represent a frontier in cancer research, offering novel insights and therapeutic possibilities. Understanding and harnessing these modifications can pave the way for breakthroughs in cancer treatment, potentially transforming the approach to combating this complex disease.


Asunto(s)
Neoplasias , ARN Ribosómico , Ribosomas , Humanos , Neoplasias/genética , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , ARN Ribosómico/metabolismo , ARN Ribosómico/genética , Ribosomas/metabolismo , Ribosomas/genética , Procesamiento Postranscripcional del ARN/genética
6.
Int J Biol Macromol ; 268(Pt 1): 131781, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38657924

RESUMEN

Alternative splicing is a crucial regulator in stem cell biology, intricately influencing the functions of various biological macromolecules, particularly pre-mRNAs and the resultant protein isoforms. This regulatory mechanism is vital in determining stem cell pluripotency, differentiation, and proliferation. Alternative splicing's role in allowing single genes to produce multiple protein isoforms facilitates the proteomic diversity that is essential for stem cells' functional complexity. This review delves into the critical impact of alternative splicing on cellular functions, focusing on its interaction with key macromolecules and how this affects cellular behavior. We critically examine how alternative splicing modulates the function and stability of pre-mRNAs, leading to diverse protein expressions that govern stem cell characteristics, including pluripotency, self-renewal, survival, proliferation, differentiation, aging, migration, somatic reprogramming, and genomic stability. Furthermore, the review discusses the therapeutic potential of targeting alternative splicing-related pathways in disease treatment, particularly focusing on the modulation of RNA and protein interactions. We address the challenges and future prospects in this field, underscoring the need for further exploration to unravel the complex interplay between alternative splicing, RNA, proteins, and stem cell behaviors, which is crucial for advancing our understanding and therapeutic approaches in regenerative medicine and disease treatment.


Asunto(s)
Empalme Alternativo , Precursores del ARN , Células Madre , Humanos , Precursores del ARN/genética , Precursores del ARN/metabolismo , Animales , Células Madre/metabolismo , Células Madre/citología , Diferenciación Celular/genética
7.
J Exp Clin Cancer Res ; 42(1): 335, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38057867

RESUMEN

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignant tumors globally. Understanding the molecular basis of tumor progression and drug resistance can offer innovative strategies to enhance clinical outcomes for HNSCC patients. METHODS: The cytoskeletal remodeling genes associated with cisplatin resistance were screened using a PCR array. The role of alpha-actinin 1 (ACTN1) in modulating cisplatin resistance and tumorigenesis in HNSCC was evaluated both in vitro and in vivo. Co-immunoprecipitation (Co-IP), IP-mass spectrometry (MS), western blotting, dual-luciferase assay, and bioinformatics analysis were performed to elucidate the underlying mechanisms involved. RESULTS: Our study identifies ACTN1 as a crucial contributor to cisplatin resistance and tumorigenesis in HNSCC, as evidenced across cellular, animal, and patient-derived xenograft models. From a clinical perspective, overexpression of ACTN1 significantly correlates with a suboptimal response to neoadjuvant chemotherapy and reduced overall survival in HNSCC patients. Mechanistically, ACTN1 predominantly activates ß-catenin-mediated signaling by promoting the interaction between myosin heavy chain 9 (MYH9) and GSK-3ß, leading to the ubiquitin-dependent degradation of GSK-3ß. ACTN1 also interacts with integrin ß1, subsequently activating the FAK/PI3K/AKT pathway, providing an additional avenue for the activation of ß-catenin signaling. Our study also unveils that the ß-catenin/c-Myc axis transcriptionally regulates ACTN1, thereby creating a positive feedback loop promoting HNSCC tumorigenesis and drug resistance. CONCLUSIONS: These insights underscore the novel mechanisms that highlight ACTN1's pivotal role in driving HNSCC progression and resistance to chemotherapy, suggesting ACTN1 as a promising therapeutic target in HNSCC management.


Asunto(s)
Cisplatino , Neoplasias de Cabeza y Cuello , Animales , Humanos , Cisplatino/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Integrina beta1/metabolismo , Fosforilación , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Actinina/genética , Actinina/metabolismo , Línea Celular Tumoral , Carcinogénesis/genética , Transformación Celular Neoplásica , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Proliferación Celular , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo
8.
Cancers (Basel) ; 15(23)2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-38067199

RESUMEN

BACKGROUD: The stratification of head and neck squamous cell carcinoma (HNSCC) patients based on prognostic differences is critical for therapeutic guidance. This study was designed to construct a predictive signature derived from T-cell receptor-related genes (TCRRGs) to forecast the clinical outcomes in HNSCC. METHODS: We sourced gene expression profiles from The Cancer Genome Atlas (TCGA) HNSCC dataset, GSE41613, and GSE65858 datasets. Utilizing consensus clustering analysis, we identified two distinct HNSCC clusters according to TCRRG expression. A TCRRG-based signature was subsequently developed and validated across diverse independent HNSCC cohorts. Moreover, we established a nomogram model based on TCRRGs. We further explored differences in immune landscapes between high- and low-risk groups. RESULTS: The TCGA HNSCC dataset was stratified into two clusters, displaying marked variations in both overall survival (OS) and immune cell infiltration. Furthermore, we developed a robust prognostic signature based on TCRRG utilizing the TCGA HNSCC train cohort, and its prognostic efficacy was validated in the TCGA HNSCC test cohort, GSE41613, and GSE65858. Importantly, the high-risk group was characterized by a suppressive immune microenvironment, in contrast to the low-risk group. Our study successfully developed a robust TCRRG-based signature that accurately predicts clinical outcomes in HNSCC, offering valuable strategies for improved treatments.

9.
Nanomaterials (Basel) ; 12(21)2022 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-36364684

RESUMEN

With the rapid development of engineered nanomaterials (ENMs) in biomedical applications, their biocompatibility and cytotoxicity need to be evaluated properly. Recently, it has been demonstrated that inflammasome activation may be a vital contributing factor for the development of biological responses induced by ENMs. Among the inflammasome family, NLRP3 inflammasome has received the most attention because it directly interacts with ENMs to cause the inflammatory effects. However, the pathways that link ENMs to NLRP3 inflammasome have not been thoroughly summarized. Thus, we reviewed recent findings on the role of major ENMs properties in modulating NLRP3 inflammasome activation, both in vitro and in vivo, to provide a better understanding of the underlying mechanisms. In addition, the interactions between ENMs and NLRP3 inflammasome activation are summarized, which may advance our understanding of safer designs of nanomaterials and ENM-induced adverse health effects.

10.
Zhonghua Xin Xue Guan Bing Za Zhi ; 37(6): 495-500, 2009 Jun.
Artículo en Zh | MEDLINE | ID: mdl-19927628

RESUMEN

OBJECTIVE: To explore the underlying mechanism of mesenchymal stem cells (MSCs) transfer induced cardiac function improvement in failing hearts. METHODS: Congestive heart failure (CHF) was induced in rats by cauterization of the heart wall. MSCs were cultured from autologous bone marrow and injected into the border zone and the remote myocardium 5 days after cauterization. RESULTS: Ten weeks later, cardiomyocyte nucleus mitotic index, capillary density and expression of insulin-like growth factor 1 (IGF-1), hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF) were significantly increased in the border zone and significantly reduced in the remote myocardium in CHF rats (all P<0.05 vs. sham). Besides cardiac function improvement and left ventricular remodeling attenuation evidenced by hemodynamic and echocardiographic examinations, expressions of IGF-1, HGF and VEGF in the remote myocardium and in the border zone were also significantly upregulated (P<0.05 or P<0.01 vs. CHF), and cardiomyocyte nucleus mitotic index as well as capillary density were significantly increased in CHF rats with MSCs (P<0.05 or P<0.01 vs. CHF). Moreover, collagen area was significantly reduced and myocardial area was significantly increased in the border zone in these rats too. CONCLUSION: Autologous MSC implantation upregulated expressions of growth factors enhanced cardioangiogenesis which might be the underlying mechanisms for improved cardiac function and attenuated left ventricular remodeling induced by MSCs transplantation in failing rat myocardium.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/terapia , Trasplante de Células Madre Mesenquimatosas , Miocardio/metabolismo , Animales , Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Trasplante Autólogo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Remodelación Ventricular
11.
Mol Med Rep ; 17(6): 7701-7707, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29620281

RESUMEN

Mesenchymal stem cell­conditioned medium (MSC­CM) contains various cytokines (osteopontin and macrophage colony stimulating factor 1) secreted by MSCs and may modulate the immune response in tubulointerstitial fibrosis. The aim of the present study was to investigate whether MSC­CM treatment may affect B cell­dependent immune responses, which have previously been reported to facilitate the renal fibrotic processes following unilateral ureteral obstruction (UUO). In the present study, histological analysis, flow cytometry, western blotting and reverse transcription­quantitative polymerase chain reaction were performed. MSC­CM treatment was observed to impede renal infiltration of B lymphocytes and the expression of CC chemokine ligand­2. Additionally, UUO suppressed the subsequent recruitment of monocytes/macrophages to the kidney, limited local inflammation and attenuated renal fibrosis. The findings of the present study identified a potential mechanism of MSC­CM in ameliorating the UUO­kidney.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Células Madre Mesenquimatosas/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Nefritis Intersticial/etiología , Nefritis Intersticial/metabolismo , Obstrucción Ureteral/etiología , Obstrucción Ureteral/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Fibrosis , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Nefritis Intersticial/patología , Obstrucción Ureteral/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA