Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 149(4): 923-35, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22559944

RESUMO

Structural genomic variations represent a major driving force of evolution, and a burst of large segmental gene duplications occurred in the human lineage during its separation from nonhuman primates. SRGAP2, a gene recently implicated in neocortical development, has undergone two human-specific duplications. Here, we find that both duplications (SRGAP2B and SRGAP2C) are partial and encode a truncated F-BAR domain. SRGAP2C is expressed in the developing and adult human brain and dimerizes with ancestral SRGAP2 to inhibit its function. In the mouse neocortex, SRGAP2 promotes spine maturation and limits spine density. Expression of SRGAP2C phenocopies SRGAP2 deficiency. It underlies sustained radial migration and leads to the emergence of human-specific features, including neoteny during spine maturation and increased density of longer spines. These results suggest that inhibition of SRGAP2 function by its human-specific paralogs has contributed to the evolution of the human neocortex and plays an important role during human brain development.


Assuntos
Encéfalo/citologia , Encéfalo/embriologia , Proteínas Ativadoras de GTPase/genética , Duplicação Gênica , Neurônios/citologia , Duplicações Segmentares Genômicas , Animais , Movimento Celular , Espinhas Dendríticas/metabolismo , Evolução Molecular , Humanos , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Estrutura Terciária de Proteína , Especificidade da Espécie
2.
Cell ; 138(5): 990-1004, 2009 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-19737524

RESUMO

During brain development, proper neuronal migration and morphogenesis is critical for the establishment of functional neural circuits. Here we report that srGAP2 negatively regulates neuronal migration and induces neurite outgrowth and branching through the ability of its F-BAR domain to induce filopodia-like membrane protrusions resembling those induced by I-BAR domains in vivo and in vitro. Previous work has suggested that in nonneuronal cells filopodia dynamics decrease the rate of cell migration and the persistence of leading edge protrusions. srGAP2 knockdown reduces leading process branching and increases the rate of neuronal migration in vivo. Overexpression of srGAP2 or its F-BAR domain has the opposite effects, increasing leading process branching and decreasing migration. These results suggest that F-BAR domains are functionally diverse and highlight the functional importance of proteins directly regulating membrane deformation for proper neuronal migration and morphogenesis.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Neurogênese , Neurônios/citologia , Animais , Movimento Celular , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Proteínas Ativadoras de GTPase , Camundongos , Pseudópodes/metabolismo
3.
Medicina (Kaunas) ; 60(2)2024 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-38399515

RESUMO

Cancer is one of the leading causes of death in the world. Various drugs have been developed to eliminate it but to no avail because a tumor can go into dormancy to avoid therapy. In the past few decades, tumor dormancy has become a popular topic in cancer therapy. Recently, there has been an important breakthrough in the study of tumor dormancy. That is, cancer cells can enter a reversible drug-tolerant persister (DTP) state to avoid therapy, but no exact mechanism has been found. The study of the link between the DTP state and diapause seems to provide an opportunity for a correct understanding of the mechanism of the DTP state. Completely treating cancer and avoiding dormancy by targeting the expression of key genes in diapause are possible. This review delves into the characteristics of the DTP state and its connection with embryonic diapause, and possible treatment strategies are summarized. The authors believe that this review will promote the development of cancer therapy.


Assuntos
Diapausa , Neoplasias , Animais , Humanos , Neoplasias/tratamento farmacológico
4.
Mol Cell Probes ; 71: 101920, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37442529

RESUMO

BACKGROUND: Aberrant expression of circRNAs is involved in the progression of hepatocellular carcinoma (HCC). This study aimed at screening the pro-tumorigenic circular RNAs (circRNAs) in HCC and the mechanisms of circCPSF6 expression influencing HCC characteristics. METHOD: circCPSF6 was identified in HCC tissues using high-throughput sequencing data, and its expression was verified in both HCC tissues and cell lines using quantitative real-time PCR (qRT-PCR). CCK-8 and Transwell assays were used to evaluate the effects of circCPSF6 on HCC proliferation and migration. A xenograft mouse model was used to investigate the effects of circCPSF6 on HCC progression in vivo, and the significance of circCPSF6 in HCC was verified both in vivo and in vitro. circCPSF6-associated miRNAs and mRNAs were identified using bioinformatic analyses. Luciferase reporter, RNA pull-down, Fluorescence in situ hybridization, and RNA immunoprecipitation assays were performed to elucidate the circCPSF6 regulatory axis in HCC. RESULT: CircCPSF6 expression was increased in HCC cell lines and tissues, and the expression of its parental mRNA was positively correlated with tumor severity and negatively correlated with survival. Mechanistic analyses of HCC cell lines showed that tumorigenesis was inhibited by circCPSF6 knockdown and promoted by its overexpression. Functional analyses revealed that circCPSF6 mediated HCC development by sponging miR-145-5p as a competing endogenous RNA. Furthermore, this sponging upregulated the miR-145-5p target gene MAP4K4, a classical pro-tumorigenic gene. CONCLUSION: Our findings reveal a regulatory network that includes the circCPSF6-miR-145-5p-MAP4K4 axis. Elements of this axis are potential HCC biomarkers, as well as targets for HCC treatment.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroRNAs , Humanos , Animais , Camundongos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , RNA Circular/genética , Hibridização in Situ Fluorescente , Linhagem Celular Tumoral , MicroRNAs/genética , MicroRNAs/metabolismo , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Serina-Treonina Quinases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética
5.
Cancer Cell Int ; 21(1): 438, 2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34419048

RESUMO

BACKGROUND: Gastric cancer is one of the most common malignant tumors of the digestive system. However, its targeted therapy develops at a slow pace. Thus, exploring the mechanisms of the malignant behavior of gastric cancer cells is crucial to exploit its treatment. Mammalian never-in-mitosis A (NIMA)-related kinases (NEKs) are considered to play a significant role in cancer cell proliferation. However, no study has reported on NIMA family proteins in gastric cancer. METHODS: Bioinformatics analysis was employed to clarify the expression patterns of NEK1-NEK11 and their effects on prognosis. The effects of NEK7 on immune infiltration and NEK7 related pathways were also analyzed. At the cell level, 5-ethynyl-2-deoxyuridine, cell cycle, and Cell Counting Kit-8 assays were utilized to clarify the effect of NEK7 on gastric cancer cell proliferation. A mouse subcutaneous model revealed the regulating effect of NEK7 on gastric cancer cell proliferation in vivo. RESULTS: Bioinformatics analysis revealed that NEK7 is upregulated in gastric cancer and is related to poor prognosis. NEK7 is also related to T-stage, which is closely associated with cell proliferation. Further analysis showed that NEK7 was correlated with infiltration of multiple immune cells as well as gastric cancer-related pathways. Cell experiments indicated the promoting effect of NEK7 on cell proliferation, while the absence of NEK7 could lead to inhibition of gastric cancer proliferation and G1/S arrest. CONCLUSION: NEK7 exerts a regulatory effect on cell proliferation and is closely related to tumor immune infiltration.

6.
Cancer Cell Int ; 19: 341, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31866766

RESUMO

Circulating tumor cells (CTCs) are a rare subset of cells found in the blood of patients with solid tumors, which function as a seed for metastases. Cancer cells metastasize through the bloodstream either as single migratory CTCs or as multicellular groupings-CTC clusters. The CTCs preserve primary tumor heterogeneity and mimic tumor properties, and may be considered as clinical biomarker, preclinical model, and therapeutic target. The potential clinical application of CTCs is being a component of liquid biopsy. CTCs are also good candidates for generating preclinical models, especially 3D organoid cultures, which could be applied in drug screening, disease modeling, genome editing, tumor immunity, and organoid biobanks. In this review, we summarize current knowledge on the value and promise of evolving CTC technologies and highlight cutting-edge research on CTCs in liquid biopsy, tumor metastasis, and organoid preclinical models. The study of CTCs offers broad pathways to develop new biomarkers for tumor patient diagnosis, prognosis, and response to therapy, as well as translational models accelerating oncologic drug development.

7.
Cancer Sci ; 109(7): 2199-2210, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29788550

RESUMO

Ubiquitin-specific protease 22 (USP22) is a member of the "death-from-cancer" signature, which plays a key role in cancer progression. Previous evidence has shown that USP22 is overexpressed and correlates with poor prognosis in glioma. The effect and mechanism of USP22 in glioma malignancy, especially cancer stemness, remain elusive. Herein, we find USP22 is more enriched in stem-like tumorspheres than differentiated glioma cells. USP22 knockdown inhibits cancer stemness in glioma cell lines. With a cell-penetrating TAT-tag protein, B cell-specific Moloney murine leukemia virus integration site 1 (BMI1), a robust glioma stem-cell marker, is found to mediate the effect of USP22 on glioma stemness. By immunofluorescence, USP22 and BMI1 are found to share similar intranuclear expression in glioma cells. By analysis with immunohistochemistry and bioinformatics, USP22 is found to positively correlate with BMI1 at the post-translational level only rather than at the transcriptional level. By immunoprecipitation and in vivo deubiquitination assay, USP22 is found to interact with and deubiquitinate BMI1 for protein stabilization. Microarray analysis shows that USP22 and BMI1 mutually regulate a series of genes involved in glioma stemness such as POSTN, HEY2, PDGFRA and ATF3. In vivo study with nude mice confirms the role of USP22 in promoting glioma tumorigenesis by regulating BMI1. All these findings indicate USP22 as a novel deubiquitinase of BMI1 in glioma. We propose a working model of the USP22-BMI1 axis, which promotes glioma stemness and tumorigenesis through oncogenic activation. Thus, targeting USP22 might be an effective strategy to treat glioma especially in those with elevated BMI1 expression.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Complexo Repressor Polycomb 1/metabolismo , Tioléster Hidrolases/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Transformação Celular Neoplásica/genética , Glioma/metabolismo , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , Proteínas Oncogênicas , Ubiquitina Tiolesterase
8.
Med Res Rev ; 37(3): 627-661, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27775833

RESUMO

Glioblastoma (GBM) is regarded as the most common primary intracranial neoplasm. Despite standard treatment with tumor resection and radiochemotherapy, the outcome remains gloomy. It is evident that a combination of oncogenic gain of function and tumor-suppressive loss of function has been attributed to glioma initiation and progression. The ubiquitin-proteasome system is a well-orchestrated system that controls the fate of most proteins by striking a dynamic balance between ubiquitination and deubiquitination of substrates, having a profound influence on the modulation of oncoproteins, tumor suppressors, and cellular signaling pathways. In recent years, deubiquitinating enzymes (DUBs) have emerged as potential anti-cancer targets due to their targeting several key proteins involved in the regulation of tumorigenesis, apoptosis, senescence, and autophagy. This review attempts to summarize recent studies of GBM-associated DUBs, their roles in various cellular processes, and discuss the relation between DUBs deregulation and gliomagenesis, especially how DUBs regulate glioma stem cells pluripotency, microenvironment, and resistance of radiation and chemotherapy through core stem-cell transcriptional factors. We also review recent achievements and progress in the development of potent and selective reversible inhibitors of DUBs, and attempted to find a potential GBM treatment by DUBs intervention.


Assuntos
Enzimas Desubiquitinantes/metabolismo , Glioblastoma/enzimologia , Glioblastoma/terapia , Terapia de Alvo Molecular , Animais , Carcinogênese/patologia , Enzimas Desubiquitinantes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia
9.
Tumour Biol ; 37(3): 2901-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26409450

RESUMO

Phenoxybenzamine hydrochloride (PHEN) is a selective antagonist of both α-adrenoceptor and calmodulin that exhibits anticancer properties. The aim of this study was to explore the anti-tumor function of PHEN in glioma. Cell proliferation assay was used to assess glioma cell growth. Migration and invasion capacity of glioma cells was monitored by wound-healing and transwell assay, respectively. Neurosphere formation test was adopted for the tumorigenesis of glioma cells, which was also confirmed by soft agar cloning formation test in vitro and a nude mouse model in vivo. Finally, we explored the potential pathway utilized by PHEN using Western blot and immunofluoresce staining. PHEN exhibited a significant inhibitory effect on the proliferation of both U251 and U87MG glioma cell lines in a positive dose-dependent manner. PHEN apparently attenuated the malignancy of glioma in terms of migration and invasion and also suppressed the tumorigenic capacity both in vitro and in vivo. Mechanism study showed that PHEN promoted tumor suppression by inhibiting the TrkB-Akt pathway. The results of the present study demonstrated that PHEN suppressed the proliferation, migration, invasion, and tumorigenesis of glioma cells, induced LINGO-1 expression, and inhibited the TrkB-Akt pathway, which may prove to be the mechanisms underlying the anti-tumor effect of PHEN on glioma cells.


Assuntos
Antineoplásicos/farmacologia , Glioma/tratamento farmacológico , Fenoxibenzamina/farmacologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Glioma/patologia , Humanos , Proteínas de Membrana/análise , Camundongos , Invasividade Neoplásica , Proteínas do Tecido Nervoso/análise , Fenoxibenzamina/uso terapêutico
10.
Neurochem Res ; 41(6): 1211-8, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26801170

RESUMO

Alzheimer's disease (AD) is the most common type of neurodegenerative dementia that affects the elderly population. Nerve growth factor (NGF) contributes to the survival, regeneration and death of neurons during aging and in neurodegenerative diseases. Recently, research has shown that NGF is related to the pathology, mechanisms and symptoms of AD. Therefore, there is a need to summarize the new advancements in NGF research and its potential therapeutic implications in AD. In this review, we will focus on NGF distribution, production, and function; the interaction of Aß and NGF; and the effect of different therapy methods on AD. In summary, we hope to describe the experimental and clinical data demonstrating the important roles of NGF for AD treatment.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Fator de Crescimento Neural/biossíntese , Terapia por Acupuntura/tendências , Doença de Alzheimer/genética , Animais , Terapia Genética/tendências , Humanos , Fator de Crescimento Neural/análise , Fator de Crescimento Neural/genética , Preparações de Plantas/uso terapêutico , Transplante de Células-Tronco/tendências
11.
Mol Cell Neurosci ; 60: 10-25, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24561795

RESUMO

The Slit-Robo GTPase activating protein 3 (srGAP3) is an important modulator of actin cytoskeletal dynamics and has an important influence on a variety of neurodevelopmental processes. Mutations in the SRGAP3 gene on chromosome 3p25 have been found in patients with intellectual disability. Genome-wide association studies and behavioral assays of knockout mice had also revealed SRGAP3 as a risk gene for schizophrenia. We have recently shown that srGAP3 protein undergoes regulated shuttling between the cytoplasm and the nucleus during neuronal development. It is shown here that nuclear-localized srGAP3 interacts with the SWI/SNF remodeling factor Brg1. This interaction is mediated by the C-terminal of srGAP3 and the ATPase motif of Brg1. In the primary cultured rat cortical neurons, the levels of nuclear-localized srGAP3 and its interaction with Brg1 have a significant impact on dendrite complexity. Furthermore, the interaction between srGAP3 and Brg1 was also involved in valproic acid (VPA) -induced neuronal differentiation of Neuro2a cells. We then show that GTP-bound Rac1 and GAP-43 may be potential mediators of nuclear srGAP3 and Brg1. Our results not only indicate a novel signaling pathway that contributes to neuronal differentiation and dendrite morphology, but also implicate a novel molecular mechanism underlying srGAP3 regulation of gene expression.


Assuntos
DNA Helicases/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Animais , Sítios de Ligação , Células COS , Células Cultivadas , Chlorocebus aethiops , Montagem e Desmontagem da Cromatina , DNA Helicases/química , DNA Helicases/genética , Proteína GAP-43/metabolismo , Proteínas Ativadoras de GTPase/genética , Células HEK293 , Humanos , Camundongos , Neurogênese , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/química , Fatores de Transcrição/genética , Ácido Valproico/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo
12.
Biochim Biophys Acta Rev Cancer ; 1879(5): 189137, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38880161

RESUMO

Cancer is a profoundly dynamic, heterogeneous and aggressive systemic ailment, with a coordinated evolution of various types of tumor niches. Hypoxia plays an indispensable role in the tumor micro-ecosystem, drastically enhancing the plasticity of cancer cells, fibroblasts and immune cells and orchestrating intercellular communication. Hypoxia-induced signals, particularly hypoxia-inducible factor-1α (HIF-1α), drive the reprogramming of genetic, transcriptional, and proteomic profiles. This leads to a spectrum of interconnected processes, including augmented survival of cancer cells, evasion of immune surveillance, metabolic reprogramming, remodeling of the extracellular matrix, and the development of resistance to conventional therapeutic modalities like radiotherapy and chemotherapy. Here, we summarize the latest research on the multifaceted effects of hypoxia, where a multitude of cellular and non-cellular elements crosstalk with each other and co-evolve in a synergistic manner. Additionally, we investigate therapeutic approaches targeting hypoxic niche, encompassing hypoxia-activated prodrugs, HIF inhibitors, nanomedicines, and combination therapies. Finally, we discuss some of the issues to be addressed and highlight the potential of emerging technologies in the treatment of cancer.

13.
Signal Transduct Target Ther ; 9(1): 92, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38637540

RESUMO

Cancer, a complex and multifactorial disease, presents a significant challenge to global health. Despite significant advances in surgical, radiotherapeutic and immunological approaches, which have improved cancer treatment outcomes, drug therapy continues to serve as a key therapeutic strategy. However, the clinical efficacy of drug therapy is often constrained by drug resistance and severe toxic side effects, and thus there remains a critical need to develop novel cancer therapeutics. One promising strategy that has received widespread attention in recent years is drug repurposing: the identification of new applications for existing, clinically approved drugs. Drug repurposing possesses several inherent advantages in the context of cancer treatment since repurposed drugs are typically cost-effective, proven to be safe, and can significantly expedite the drug development process due to their already established safety profiles. In light of this, the present review offers a comprehensive overview of the various methods employed in drug repurposing, specifically focusing on the repurposing of drugs to treat cancer. We describe the antitumor properties of candidate drugs, and discuss in detail how they target both the hallmarks of cancer in tumor cells and the surrounding tumor microenvironment. In addition, we examine the innovative strategy of integrating drug repurposing with nanotechnology to enhance topical drug delivery. We also emphasize the critical role that repurposed drugs can play when used as part of a combination therapy regimen. To conclude, we outline the challenges associated with repurposing drugs and consider the future prospects of these repurposed drugs transitioning into clinical application.


Assuntos
Reposicionamento de Medicamentos , Neoplasias , Humanos , Reposicionamento de Medicamentos/métodos , Neoplasias/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Resultado do Tratamento , Terapia Combinada , Microambiente Tumoral
14.
Int J Biol Sci ; 20(6): 2044-2071, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38617549

RESUMO

Cholesterol is crucial for cell survival and growth, and dysregulation of cholesterol homeostasis has been linked to the development of cancer. The tumor microenvironment (TME) facilitates tumor cell survival and growth, and crosstalk between cholesterol metabolism and the TME contributes to tumorigenesis and tumor progression. Targeting cholesterol metabolism has demonstrated significant antitumor effects in preclinical and clinical studies. In this review, we discuss the regulatory mechanisms of cholesterol homeostasis and the impact of its dysregulation on the hallmarks of cancer. We also describe how cholesterol metabolism reprograms the TME across seven specialized microenvironments. Furthermore, we discuss the potential of targeting cholesterol metabolism as a therapeutic strategy for tumors. This approach not only exerts antitumor effects in monotherapy and combination therapy but also mitigates the adverse effects associated with conventional tumor therapy. Finally, we outline the unresolved questions and suggest potential avenues for future investigations on cholesterol metabolism in relation to cancer.


Assuntos
Neoplasias , Humanos , Carcinogênese , Terapia Combinada , Sobrevivência Celular , Transformação Celular Neoplásica , Microambiente Tumoral
15.
Front Physiol ; 15: 1377025, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38681139

RESUMO

MG53, a member of the tripartite motif protein family, possesses multiple functionalities due to its classic membrane repair function, anti-inflammatory ability, and E3 ubiquitin ligase properties. Initially recognized for its crucial role in membrane repair, the therapeutic potential of MG53 has been extensively explored in various diseases including muscle injury, myocardial damage, acute lung injury, and acute kidney injury. However, further research has revealed that the E3 ubiquitin ligase characteristics of MG53 also contribute to the pathogenesis of certain conditions such as diabetic cardiomyopathy, insulin resistance, and metabolic syndrome. Moreover, recent studies have highlighted the anti-tumor effects of MG53 in different types of cancer, such as small cell lung cancer, liver cancer, and colorectal cancer; these effects are closely associated with their E3 ubiquitin ligase activities. In summary, MG53 is a multifunctional protein that participates in important physiological and pathological processes of multiple organs and is a promising therapeutic target for various human diseases. MG53 plays a multi-organ protective role due to its membrane repair function and its exertion of anti-tumor effects due to its E3 ubiquitin ligase properties. In addition, the controversial aspect of MG53's E3 ubiquitin ligase properties potentially causing insulin resistance and metabolic syndrome necessitates further cross-validation for clarity.

16.
MedComm (2020) ; 4(6): e450, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38084259

RESUMO

Overexpression of immediate-early genes (IEGs) has been linked to was associated with cancer progression and prognosis. In a recent study, Gu et al. reported the midnolin-proteasome pathway, a novel ubiquitin-independent proteasomal degradation. 1 The study provided the mechanism of rapid degradation for nuclear proteins with high unsteadiness. Targeting the midnolin-proteasome pathway might be beneficial for cancer therapy.

17.
Trends Cell Biol ; 33(6): 449-450, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37005211

RESUMO

Spatial omics facilitate an in-depth understanding of cell states and cell interactions. Recent work by Zhang et al. simultaneously seizes spatial epigenetic priming, differentiation, and gene regulation at nearly single-cell resolution by developing an epigenome-transcriptome comapping technology. This work displays how epigenetic features influence cell dynamics and transcriptional phenotypes at spatial and genome-wide levels.


Assuntos
Epigenoma , Transcriptoma , Transcriptoma/genética , Epigenoma/genética , Regulação da Expressão Gênica , Genoma , Tecnologia
18.
Trends Mol Med ; 29(11): 886-896, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37735022

RESUMO

Neurodegenerative diseases (NDs) are commonly observed and while no therapy is universally applicable, cell-based therapies are promising. Stem cell transplantation has been investigated, but endogenous neural stem cells (eNSCs), despite their potential, especially with the development of bioelectronic medicine and biomaterials, remain understudied. Here, we compare stem cell transplantation therapy with eNSC-based therapy and summarize the combined use of eNSCs and developing technologies. The rapid development of implantable biomaterials has resulted in electronic stimulation becoming increasingly effective and decreasingly invasive. Thus, the combination of bioelectronic medicine and eNSCs has substantial potential for the treatment of NDs.

19.
Biochim Biophys Acta Rev Cancer ; 1878(3): 188884, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36990250

RESUMO

The vagus nerve (VN) is the main parasympathetic nerve of the autonomic nervous system. It is widely distributed in the gastrointestinal tract and maintains gastrointestinal homeostasis with the sympathetic nerve under physiological conditions. The VN communicates with various components of the tumor microenvironment to positively and dynamically affect the progression of gastrointestinal tumors (GITs). The intervention in vagus innervation delays GIT progression. Developments in adeno-associated virus vectors, nanotechnology, and in vivo neurobiological techniques have enabled the creation of precisely regulated "tumor neurotherapies". The present review aimed to summarize the mechanisms of communication between the VN and the gastrointestinal TME and to explore the potential and challenges of VN-based tumor neurotherapy in GITs.


Assuntos
Neoplasias Gastrointestinais , Fenômenos Fisiológicos do Sistema Nervoso , Humanos , Nervo Vago/fisiologia , Homeostase , Microambiente Tumoral
20.
Biomed Pharmacother ; 166: 115390, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37660648

RESUMO

The tumor microenvironment (TME) is crucial in cancer progression, and the extracellular matrix (ECM) is an important TME component. Collagen is a major ECM component that contributes to tumor cell infiltration, expansion, and distant metastasis during cancer progression. Recent studies reported that collagen is deposited in the TME to form a collagen wall along which tumor cells can infiltrate and prevent drugs from working on the tumor cells. Collagen-tumor cell interaction is complex and requires the activation of multiple signaling pathways for biochemical and mechanical signaling interventions. In this review, we examine the effect of collagen deposition in the TME on tumor progression and discuss the interaction between collagen and tumor cells. This review aims to illustrate the functions and mechanisms of collagen in tumor progression in the TME and its role in tumor therapy. The findings indicated collagen in the TME appears to be a better target for cancer therapy.


Assuntos
Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Colágeno , Matriz Extracelular , Comunicação Celular , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA