Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Stem Cells ; 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38864549

RESUMO

SIRT6 owns versatile types of enzymatic activities as a multitasking protein, including ribosyltransferase and deacetylase ones. To investigate the epigenetic regulations of SIRT6 on MSC fate determination via histone deacetylation, we utilized allosteric small molecules specifically controlling its histone 3 deacetylation activities. Results showed that enhanced deacetylation of SIRT6 promoted the ossific lineage commitment of MSC and finally achieved anabolic effects on hard tissues. Mechanistically, H3K9ac and H3K56ac, governed by SIRT6, in MSC orchestrated the transcriptions of crucial metabolic genes, mediating MSC fate determination. Most importantly, our data evidenced that modulating the epigenetic regulations of SIRT6, specifically via enhancing its deacetylation of H3K9ac and H3K56ac, was a promising choice to treat bone loss diseases and promote dentine regeneration. In this study, we revealed the specific roles of SIRT6's histone modification in MSC fate determination. These findings endow us with insights on SIRT6 and the promising therapeutic choices through SIRT6's epigenetic functions for hard tissues regeneration.

2.
Int J Oral Sci ; 16(1): 33, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654018

RESUMO

Precise orchestration of cell fate determination underlies the success of scaffold-based skeletal regeneration. Despite extensive studies on mineralized parenchymal tissue rebuilding, regenerating and maintaining undifferentiated mesenchyme within calvarial bone remain very challenging with limited advances yet. Current knowledge has evidenced the indispensability of rebuilding suture mesenchymal stem cell niches to avoid severe brain or even systematic damage. But to date, the absence of promising therapeutic biomaterials/scaffolds remains. The reason lies in the shortage of fundamental knowledge and methodological evidence to understand the cellular fate regulations of scaffolds. To address these issues, in this study, we systematically investigated the cellular fate determinations and transcriptomic mechanisms by distinct types of commonly used calvarial scaffolds. Our data elucidated the natural processes without scaffold transplantation and demonstrated how different scaffolds altered in vivo cellular responses. A feasible scaffold, polylactic acid electrospinning membrane (PLA), was next identified to precisely control mesenchymal ingrowth and self-renewal to rebuild non-osteogenic suture-like tissue at the defect center, meanwhile supporting proper osteointegration with defect bony edges. Especially, transcriptome analysis and cellular mechanisms underlying the well-orchestrated cell fate determination of PLA were deciphered. This study for the first time cellularly decoded the fate regulations of scaffolds in suture-bony composite defect healing, offering clinicians potential choices for regenerating such complicated injuries.


Assuntos
Regeneração Óssea , Alicerces Teciduais , Transcriptoma , Animais , Regeneração Óssea/fisiologia , Poliésteres , Crânio/cirurgia , Células-Tronco Mesenquimais , Mesoderma/citologia , Diferenciação Celular , Engenharia Tecidual/métodos , Suturas Cranianas , Materiais Biocompatíveis
3.
Elife ; 122023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-37991825

RESUMO

The role of regulated cell death in organ development, particularly the impact of non-apoptotic cell death, remains largely uncharted. Ferroptosis, a non-apoptotic cell death pathway known for its iron dependence and lethal lipid peroxidation, is currently being rigorously investigated for its pathological functions. The balance between ferroptotic stress (iron and iron-dependent lipid peroxidation) and ferroptosis supervising pathways (anti-lipid peroxidation systems) serves as the key mechanism regulating the activation of ferroptosis. Compared with other forms of regulated necrotic cell death, ferroptosis is critically related to the metabolism of lipid and iron which are also important in organ development. In our study, we examined the role of ferroptosis in organogenesis using an ex vivo tooth germ culture model, investigating the presence and impact of ferroptotic stress on tooth germ development. Our findings revealed that ferroptotic stress increased during tooth development, while the expression of glutathione peroxidase 4 (Gpx4), a crucial anti-lipid peroxidation enzyme, also escalated in dental epithelium/mesenchyme cells. The inhibition of ferroptosis was found to partially rescue erastin-impaired tooth morphogenesis. Our results suggest that while ferroptotic stress is present during tooth organogenesis, its effects are efficaciously controlled by the subsequent upregulation of Gpx4. Notably, an overabundance of ferroptotic stress, as induced by erastin, suppresses tooth morphogenesis.


Assuntos
Ferroptose , Odontogênese , Organogênese , Peroxidação de Lipídeos , Ferro
4.
Adv Healthc Mater ; 12(27): e2300713, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37498795

RESUMO

The extracellular matrix microenvironment of bone tissue comprises several physiological cues. Thus, artificial bone substitute materials with a single cue are insufficient to meet the demands for bone defect repair. Regeneration of critical-size bone defects remains challenging in orthopedic surgery. Intrinsic viscoelastic and piezoelectric cues from collagen fibers play crucial roles in accelerating bone regeneration, but scaffolds or implants providing integrated cues have seldom been reported. In this study, it is aimed to design and prepare hierarchically porous poly(methylmethacrylate)/polyethyleneimine/poly(vinylidenefluoride) composite implants presenting a similar viscoelastic and piezoelectric microenvironment to bone tissue via anti-solvent vapor-induced phase separation. The viscoelastic and piezoelectric cues of the composite implants for human bone marrow mesenchymal stem cell line stimulate and activate Piezo1 proteins associated with mechanotransduction signaling pathways. Cortical and spongy bone exhibit excellent regeneration and integration in models of critical-size bone defects on the knee joint and femur in vivo. This study demonstrates that implants with integrated physiological cues are promising artificial bone substitute materials for regenerating critical-size bone defects.


Assuntos
Substitutos Ósseos , Alicerces Teciduais , Humanos , Osteogênese , Substitutos Ósseos/farmacologia , Porosidade , Mecanotransdução Celular , Regeneração Óssea , Engenharia Tecidual
5.
Aging Cell ; 22(9): e13913, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37340571

RESUMO

Hippo-independent YAP dysfunction has been demonstrated to cause chronological aging of stromal cells by impairing the integrity of nuclear envelope (NE). In parallel with this report, we uncover that YAP activity also controls another type of cellular senescence, the replicative senescence in in vitro expansion of mesenchymal stromal cells (MSCs), but this event is Hippo phosphorylation-dependent, and there exist another NE integrity-independent downstream mechanisms of YAP. Specifically, Hippo phosphorylation causes reduced nuclear/active YAP and then decreases the level of YAP protein in the proceeding of replicative senescence. YAP/TEAD governs RRM2 expression to release replicative toxicity (RT) via licensing G1/S transition. Besides, YAP controls the core transcriptomics of RT to delay the onset of genome instability and enhances DNA damage response/repair. Hippo-off mutations of YAP (YAPS127A/S381A ) satisfactorily release RT via maintaining cell cycle and reducing genome instability, finally rejuvenating MSCs and restoring their regenerative capabilities without risks of tumorigenesis.


Assuntos
Células-Tronco Mesenquimais , Proteínas de Sinalização YAP , Humanos , Proteínas de Ciclo Celular/genética , Instabilidade Genômica , Fosforilação
6.
MedComm (2020) ; 4(3): e244, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37188325

RESUMO

Osteoporosis is a type of bone loss disease characterized by a reduction in bone mass and microarchitectural deterioration of bone tissue. With the intensification of global aging, this disease is now regarded as one of the major public health problems that often leads to unbearable pain, risk of bone fractures, and even death, causing an enormous burden at both the human and socioeconomic layers. Classic anti-osteoporosis pharmacological options include anti-resorptive and anabolic agents, whose ability to improve bone mineral density and resist bone fracture is being gradually confirmed. However, long-term or high-frequency use of these drugs may bring some side effects and adverse reactions. Therefore, an increasing number of studies are devoted to finding new pathogenesis or potential therapeutic targets of osteoporosis, and it is of great importance to comprehensively recognize osteoporosis and develop viable and efficient therapeutic approaches. In this study, we systematically reviewed literatures and clinical evidences to both mechanistically and clinically demonstrate the state-of-art advances in osteoporosis. This work will endow readers with the mechanistical advances and clinical knowledge of osteoporosis and furthermore present the most updated anti-osteoporosis therapies.

7.
Front Endocrinol (Lausanne) ; 14: 1126787, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36950693

RESUMO

Recent studies have revealed the vital role of MSCs in bone regeneration. In both self-healing bone regeneration processes and biomaterial-induced healing of bone defects beyond the critical size, MSCs show several functions, including osteogenic differentiation and thus providing seed cells. However, adverse factors such as drug intake and body senescence can significantly affect the functions of MSCs in bone regeneration. Currently, several modalities have been developed to regulate MSCs' phenotype and promote the bone regeneration process. Epigenetic regulation has received much attention because of its heritable nature. Indeed, epigenetic regulation of MSCs is involved in the pathogenesis of a variety of disorders of bone metabolism. Moreover, studies using epigenetic regulation to treat diseases are also being reported. At the same time, the effects of epigenetic regulation on MSCs are yet to be fully understood. This review focuses on recent advances in the effects of epigenetic regulation on osteogenic differentiation, proliferation, and cellular senescence in MSCs. We intend to illustrate how epigenetic regulation of MSCs orchestrates the process of bone regeneration.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Osteogênese/genética , Epigênese Genética , Regeneração Óssea/genética , Diferenciação Celular
8.
Cell Prolif ; 56(4): e13413, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36727213

RESUMO

Dynamic chromatin accessibility regulates stem cell fate determination and tissue homeostasis via controlling gene expression. As a histone-modifying enzyme that predominantly mediates methylation of lysine 27 in histone H3 (H3K27me1/2/3), Polycomb repressive complex 2 (PRC2) plays the canonical role in targeting developmental regulators during stem cell differentiation and transformation. Embryonic ectoderm development (EED), the core scaffold subunit of PRC2 and as an H3K27me3-recognizing protein, has been broadly implicated with PRC2 stabilization and allosterically stimulated PRC2. Accumulating evidences from experimental data indicate that EED-associating epigenetic modifications are indispensable for stem cell maintenance and differentiation into specific cell lineages. In this review, we discuss the most updated advances to summarize the structural architecture of EED and its contributions and underlying mechanisms to mediating lineage differentiation of different stem cells during epigenetic modification to expand our understanding of PRC2.


Assuntos
Ectoderma , Epigênese Genética , Ectoderma/metabolismo , Diferenciação Celular/genética , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Histonas/metabolismo
9.
J Vis Exp ; (191)2023 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-36715407

RESUMO

This study introduces the development of a molar extraction model in the murine mandible to provide a practicable model for studying alveolar bone regeneration and intramembranous ossification. C57/J6 mice were used to extract the mandibular first molar to establish this model. They were executed, and the bilateral mandibles harvested, at 1 week and 4 weeks post-surgery, respectively. Subsequent serial stereoscopic harvest, histological assessment, and immunofluorescence staining were performed to demonstrate successful surgery. Immediately after surgery, the stereoscopic images displayed an empty extraction socket. The hematoxylin and eosin (H&E) at 1 week and Masson staining at 4 weeks post-surgery showed that the area of the original root was partially and fully filled with bone trabeculae, respectively. The immunofluorescence staining showed that, compared with the homeostasis side, the Sp7 expression increased at 1 week post-surgery, suggesting vigorous osteogenesis in the alveolar fossa. All these results demonstrated a practicable murine tooth extraction socket healing model. Upcoming studies revealing the mechanisms of jawbone defect healing or socket healing could adopt this method.


Assuntos
Ligamento Periodontal , Alvéolo Dental , Camundongos , Animais , Alvéolo Dental/cirurgia , Dente Molar/cirurgia , Mandíbula/cirurgia , Extração Dentária
10.
Cell Rep ; 41(6): 111599, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36351390

RESUMO

Skeletal stem cells (SSCs) fuel adult bone with stemness resources to maintain homeostasis and support regeneration, which depends on the precise determination of the osteogenic lineage commitment of SSCs. In this study, using Cre-loxP reporter lineage tracking, we identified and characterized a population of NFATc1+ SSCs in bone regeneration. Pre-existing NFATc1+ SSCs are involved in early bone callus formation. Subsequently, these NFATc1+ SSCs produce osteolineage descendants in the subsequent stages of regeneration. The Ca2+-triggered transcriptional activity of NFATc1 constitutes the pre-imprinted memory of the trajectory to intrinsically orchestrate osteogenesis of SSCs. Inhibition of Ca2+/NFATc1 signaling in SSCs directly impairs osteogenesis and bone regeneration. In summary, our findings provide a mechanistic understanding of adult bone regeneration through the regulation of NFATc1+ SSCs.


Assuntos
Regeneração Óssea , Células-Tronco , Humanos , Adulto , Osteogênese/fisiologia , Osso e Ossos , Fatores de Transcrição , Diferenciação Celular , Fatores de Transcrição NFATC/genética
11.
Adv Mater ; 34(46): e2206208, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36065047

RESUMO

Exploring high-efficiency reactive oxygen species (ROS)-elimination materials is of great importance for combating oxidative stress in diverse diseases, especially stem-cell-based biotherapeutics. By mimicking the FeN active centers of natural catalase, here, an innovative concept to design ROS-elimination artificial biocatalysts with Ru catalytic centers for stem-cell protection is reported. The experimental studies and theoretical calculations have systematically disclosed the activity merits and structure diversities of different Ru sites when serving as ROS-elimination artificial biocatalysts. Benefiting from the metallic electronic structures and synergetic effects of multiple sites, the artificial biocatalysts with Ru cluster centers present exceptional ROS-elimination activity; notably, it shows much higher catalytic efficiency per Ru atom on decomposing H2 O2 when compared to the isolated single-atom Ru sites, which is more efficient than that of the natural antioxidants and recently reported state-of-the-art ROS-scavenging biocatalysts. The systematic stem-cell protection studies reveal that the catalase-like artificial biocatalysts can provide efficient rescue ability for survival, adhesion, and differentiation functions of human mesenchymal stem cells in high ROS level conditions. It is suggested that applying these artificial biocatalysts with Ru cluster centers will offer a new pathway for engineering high-performance ROS-scavenging materials in stem-cell-based therapeutics and many other ROS-related diseases.


Assuntos
Citoproteção , Estresse Oxidativo , Humanos , Catalase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Catálise
12.
Sci Adv ; 8(29): eabn4977, 2022 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-35867784

RESUMO

Given that adult stem cells (ASCs) fuel homeostasis and healing by providing tissue-specific descendants, the fidelity of ASC fate determination is crucial for regeneration. Here, we established that an epigenetic control of epithelial ASC fate fidelity via Ezh2/H3K27me3 was indispensable for incisor homeostasis and regeneration. Mechanistically, in homeostasis, H3K27me3 upstream occupies the Sonic hedgehog (Shh) promoter to directly restrain Shh expression, thereby precisely confining Shh expression. When injury occurred, Ezh2/H3K27me3 was substantially induced within inner enamel epithelium and preameloblast zones, and such epigenetic response guaranteed the fidelity of ASC commitment via pulling injury-increased Shh back to homeostatic levels, utterly underlying regeneration progression. Once losing H3K27me3-dependent restriction of Shh expression through the Cre-Loxp system totally disrupted lineage commitment and stemness exhaustion, and abolished hard tissue regeneration emerged in vivo. We next uncovered the molecular mechanisms by which injury-induced Ezh2 mediated the spatiotemporal dynamics of H3K27me3 to repress Shh expression, thus epigenetically deciding ASC fate.


Assuntos
Células-Tronco Adultas , Proteínas Hedgehog , Epigênese Genética , Epitélio/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Histonas/genética , Histonas/metabolismo
13.
Stem Cell Res Ther ; 13(1): 77, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35193674

RESUMO

BACKGROUND: Controlling the adipo-osteogenic lineage commitment of bone marrow mesenchymal stem cell (BMSC) in favor of osteogenesis is considered a promising approach for bone regeneration and repair. Accumulating evidence indicates that oxidative phosphorylation (OXPHOS) is involved in regulating cell fate decisions. As an essential cofactor for OXPHOS, nicotinamide adenine dinucleotide (NAD) has been shown to correlate with the differentiation of stem cells. However, whether NAD manipulates BMSC lineage commitment through OXPHOS remains elusive. Therefore, it is critical to investigate the potential role of NAD on energy metabolism in mediating BMSC lineage commitment. METHODS: In this study, the mitochondrial respiration and intracellular NAD+ level were firstly compared between osteogenic and adipogenic cells. For validating the role of NAD in mitochondrial OXPHOS, the inhibitor of NAD+ salvage pathway FK866 and activator P7C3 were used to manipulate the NAD+ level during osteogenesis. Furthermore, a murine femur fracture model was established to evaluate the effect of FK866 on bone fracture repair. RESULTS: We elucidated that osteogenic committed BMSCs exhibited increased OXPHOS activity and a decreased glycolysis accompanied by an elevated intracellular NAD+ level. In contrast, adipogenic committed BMSCs showed little change in OXPHOS but an upregulated activity in glycolysis and a decline in intracellular NAD+ level in vitro. Moreover, attenuates of NAD+ via salvage pathway in BMSCs diminished osteogenic commitment due to mitochondria dysfunction and reduced activity of OXPHOS. The cells were rescued by supplementing with nicotinamide mononucleotide. In addition, treatment with NAD+ inhibitor FK866 impaired bone fracture healing in vivo. CONCLUSION: Our data reveals NAD+-mediated mitochondrial OXPHOS is indispensable for osteogenic commitment in BMSCs and bone repair, which might provide a potential therapeutic target for bone repair and regeneration.


Assuntos
Fraturas Ósseas , Células-Tronco Mesenquimais , Animais , Diferenciação Celular , Fraturas Ósseas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , NAD/metabolismo , Osteogênese , Fosforilação Oxidativa
14.
Int J Oral Sci ; 14(1): 6, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-35082271

RESUMO

Inflammation-associated proteinase functions are key determinants of inflammatory stromal tissues deconstruction. As a specialized inflammatory pathological process, dental internal resorption (IR) includes both soft and hard tissues deconstruction within the dentin-pulp complex, which has been one of the main reasons for inflammatory tooth loss. Mechanisms of inflammatory matrix degradation and tissue resorption in IR are largely unclear. In this study, we used a combination of Cre-loxP reporter, flow cytometry, cell transplantation, and enzyme activities assay to mechanistically investigate the role of regenerative cells, odontoblasts (ODs), in inflammatory mineral resorption and matrices degradation. We report that inflamed ODs have strong capabilities of matrix degradation and tissue resorption. Traditionally, ODs are regarded as hard-tissue regenerative cells; however, our data unexpectedly present ODs as a crucial population that participates in IR-associated tissue deconstruction. Specifically, we uncovered that nuclear factor-kappa b (NF-κB) signaling orchestrated Tumor necrosis factor α (TNF-α)-induced matrix metalloproteinases (Mmps) and Cathepsin K (Ctsk) functions in ODs to enhance matrix degradation and tissue resorption. Furthermore, TNF-α increases Rankl/Opg ratio in ODs via NF-κB signaling by impairing Opg expression but increasing Rankl level, which utterly makes ODs cell line 17IIA11 (A11) become Trap+ and Ctsk+ multinucleated cells to perform resorptive actions. Blocking of NF-κB signaling significantly rescues matrix degradation and resorptive functions of inflamed ODs via repressing vital inflammatory proteinases Mmps and Ctsk. Utterly, via utilizing NF-κB specific small molecule inhibitors we satisfactorily attenuated inflammatory ODs-associated human dental IR in vivo. Our data reveal the underlying mechanisms of inflammatory matrix degradation and resorption via proteinase activities in IR-related pathological conditions.


Assuntos
NF-kappa B , Odontoblastos , Humanos , Metaloproteinases da Matriz/metabolismo , Minerais/metabolismo , NF-kappa B/metabolismo , Odontoblastos/metabolismo , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
15.
Adv Mater ; 34(46): e2107922, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34837252

RESUMO

Rebuilding mineralized tissues in skeletal and dental systems remains costly and challenging. Despite numerous demands and heavy clinical burden over the world, sources of autografts, allografts, and xenografts are far limited, along with massive risks including viral infections, ethic crisis, and so on. Per such dilemma, artificial scaffolds have emerged to provide efficient alternatives. To date, cell-free biomimetic mineralization (BM) and cell-dependent scaffolds have both demonstrated promising capabilities of regenerating mineralized tissues. However, BM and cell-dependent scaffolds have distinctive mechanisms for mineral genesis, which makes them methodically, synthetically, and functionally disparate. Herein, these two strategies in regenerative dentistry and orthopedics are systematically summarized at the level of mechanisms. For BM, methodological and theoretical advances are focused upon; and meanwhile, for cell-dependent scaffolds, it is demonstrated how scaffolds orchestrate osteogenic cell fate. The summary of the experimental advances and clinical progress will endow researchers with mechanistic understandings of artificial scaffolds in rebuilding hard tissues, by which better clinical choices and research directions may be approached.


Assuntos
Biomimética , Engenharia Tecidual , Alicerces Teciduais , Humanos , Diferenciação Celular , Osteogênese , Osso e Ossos
16.
Front Cell Dev Biol ; 9: 771336, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34881243

RESUMO

The imbalance between bone formation and bone resorption causes osteoporosis, which leads to severe bone fractures. It is known that increases in osteoclast numbers and activities are the main reasons for increasing bone resorption. Although extensive studies have investigated the regulation of osteoclastogenesis of bone marrow macrophages (BMMs), new pharmacological avenues still need to be unveiled for clinical purpose. Wnt ligands have been widely demonstrated as stimulators of bone formation; however, the inhibitory effect of the Wnt pathway in osteoclastogenesis is largely unknown. Here, we demonstrate that Wnt7b, a potent Wnt ligand that enhances bone formation and increases bone mass, also abolishes osteoclastogenesis in vitro. Importantly, enforced expression of Wnt in bone marrow macrophage lineage cells significantly disrupts osteoclast formation and activity, which leads to a dramatic increase in bone mass. Mechanistically, Wnt7b impacts the glucose metabolic process and AKT activation during osteoclastogenesis. Thus, we demonstrate that Wnt7b diminishes osteoclast formation, which will be beneficial for osteoporosis therapy in the future.

17.
J Hematol Oncol ; 14(1): 136, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34479611

RESUMO

Extracellular vesicles (EVs) are cell-derived membrane structures enclosing proteins, lipids, RNAs, metabolites, growth factors, and cytokines. EVs have emerged as essential intercellular communication regulators in multiple physiological and pathological processes. Previous studies revealed that mesenchymal stem cells (MSCs) could either support or suppress tumor progression in different cancers by paracrine signaling via MSC-derived EVs. Evidence suggested that MSC-derived EVs could mimic their parental cells, possessing pro-tumor and anti-tumor effects, and inherent tumor tropism. Therefore, MSC-derived EVs can be a cell-free cancer treatment alternative. This review discusses different insights regarding MSC-derived EVs' roles in cancer treatment and summarizes bioengineered MSC-derived EVs' applications as safe and versatile anti-tumor agent delivery platforms. Meanwhile, current hurdles of moving MSC-derived EVs from bench to bedside are also discussed.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Vesículas Extracelulares/metabolismo , Células-Tronco Mesenquimais/citologia , Neoplasias/terapia , Animais , Antineoplásicos/uso terapêutico , Apoptose , Proliferação de Células , Vesículas Extracelulares/transplante , Humanos , Células-Tronco Mesenquimais/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia
18.
Signal Transduct Target Ther ; 6(1): 307, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34456337

RESUMO

Wnt/ß-catenin signaling has been broadly implicated in human cancers and experimental cancer models of animals. Aberrant activation of Wnt/ß-catenin signaling is tightly linked with the increment of prevalence, advancement of malignant progression, development of poor prognostics, and even ascendence of the cancer-associated mortality. Early experimental investigations have proposed the theoretical potential that efficient repression of this signaling might provide promising therapeutic choices in managing various types of cancers. Up to date, many therapies targeting Wnt/ß-catenin signaling in cancers have been developed, which is assumed to endow clinicians with new opportunities of developing more satisfactory and precise remedies for cancer patients with aberrant Wnt/ß-catenin signaling. However, current facts indicate that the clinical translations of Wnt/ß-catenin signaling-dependent targeted therapies have faced un-neglectable crises and challenges. Therefore, in this study, we systematically reviewed the most updated knowledge of Wnt/ß-catenin signaling in cancers and relatively targeted therapies to generate a clearer and more accurate awareness of both the developmental stage and underlying limitations of Wnt/ß-catenin-targeted therapies in cancers. Insights of this study will help readers better understand the roles of Wnt/ß-catenin signaling in cancers and provide insights to acknowledge the current opportunities and challenges of targeting this signaling in cancers.


Assuntos
Proteínas de Neoplasias , Neoplasias , Proteínas Wnt , Via de Sinalização Wnt , beta Catenina , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
19.
Front Cell Dev Biol ; 9: 654958, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34434926

RESUMO

Autophagy is an intracellular self-cannibalization process delivering cytoplasmic components to lysosomes for digestion. Autophagy has been reported to be involved in pulpitis, but the regulation of autophagy during pulpitis progression is largely unknown. To figure out the epigenetic regulation of autophagy during pulpitis, we screened several groups of histone methyltransferases and demethylases in response to TNFα treatment. It was found JMJD3, a histone demethylase reducing di- and tri-methylation of H3K27, regulated the expression of several key autophagy genes via demethylation of H3K27me3 at the gene promoters. Our study highlighted the epigenetic regulation of autophagy genes during pulpitis, which will potentially provide a novel therapeutic strategy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA