Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
FASEB J ; 38(13): e23704, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38884155

RESUMO

Inflammation plays a crucial role in the development of various disease conditions or is closely associated with them. Inflammatory cytokines like TNF often engage in interactions with other cytokines and growth factors, including TGFß, to orchestrate inflammatory process. Basal/endogenous TGFß signaling is a universal presence, yet the precise way TNF communicates with TGFß signaling to regulate inflammation and influence inflammatory levels in macrophages has remained elusive. To address this question, this study utilized genetic approaches and a combination of molecular and cellular methods, including conditional TGFß receptor knockout mice, human cells, RNAseq, ATACseq and Cut & Run-seq. The results reveal that the TGFß signaling functions as a vital homeostatic pathway, curtailing uncontrolled inflammation in macrophages in response to TNF. Conversely, TNF employs two previously unrecognized mechanisms to suppress the TGFß signaling. These mechanisms encompass epigenetic inhibition and RBP-J-mediated inhibition of the TGFß signaling pathway by TNF. These mechanisms empower TNF to diminish the antagonistic influence exerted by the TGFß signaling pathway, ultimately enhancing TNF's capacity to induce heightened levels of inflammation. This reciprocal suppression dynamic between TNF and the TGFß signaling pathway holds unique physiopathological significance, as it serves as a crucial "braking" mechanism. The balance between TNF levels and the activity of the endogenous TGFß signaling pathway plays a pivotal role in determining the overall extent of inflammation. The potential for therapeutically augmenting the TGFß signaling pathway presents an intriguing avenue for countering the impact of TNF and, consequently, developing innovative strategies for inflammation control.


Assuntos
Inflamação , Macrófagos , Camundongos Knockout , Transdução de Sinais , Fator de Crescimento Transformador beta , Fator de Necrose Tumoral alfa , Animais , Fator de Crescimento Transformador beta/metabolismo , Camundongos , Macrófagos/metabolismo , Inflamação/metabolismo , Humanos , Fator de Necrose Tumoral alfa/metabolismo , Camundongos Endogâmicos C57BL
2.
J Immunol ; 211(9): 1340-1347, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37756541

RESUMO

TNF plays a crucial role in inflammation and bone resorption in various inflammatory diseases, including rheumatoid arthritis (RA). However, its direct ability to drive macrophages to differentiate into osteoclasts is limited. Although RBP-J is recognized as a key inhibitor of TNF-mediated osteoclastogenesis, the precise mechanisms that restrain TNF-induced differentiation of macrophages into osteoclasts are not fully elucidated. In this study, we identified that the Notch ligand Jagged1 is a previously unrecognized RBP-J target. The expression of Jagged1 is significantly induced by TNF mainly through RBP-J. The TNF-induced Jagged1 in turn functions as a feedback inhibitory regulator of TNF-mediated osteoclastogenesis. This feedback inhibition of osteoclastogenesis by Jagged1 does not exist in RANKL-induced mouse osteoclast differentiation, as RANKL does not induce Jagged1 expression. The Jagged1 level in peripheral blood monocytes/osteoclast precursors is decreased in RA compared with the nonerosive inflammatory disease systemic lupus erythematosus, suggesting a mechanism that contributes to increased osteoclast formation in RA. Moreover, recombinant Jagged1 suppresses human inflammatory osteoclastogenesis. Our findings identify Jagged1 as an RBP-J direct target that links TNF and Notch signaling pathways and restrains TNF-mediated osteoclastogenesis. Given that Jagged1 has no effect on TNF-induced expression of inflammatory genes, its use may present a new complementary therapeutic approach to mitigate inflammatory bone loss with little impact on the immune response in disease conditions.


Assuntos
Artrite Reumatoide , Reabsorção Óssea , Humanos , Animais , Camundongos , Osteogênese , Retroalimentação , Osteoclastos/metabolismo , Macrófagos , Artrite Reumatoide/metabolismo , Ligante RANK/metabolismo , Diferenciação Celular , Fator de Necrose Tumoral alfa/metabolismo
3.
J Am Acad Dermatol ; 90(5): 970-976, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38244613

RESUMO

BACKGROUND: Mast cell degranulation plays a pivotal role in urticaria and is also an early histologic characteristic of psoriasis. However, whether the activation of mast cells contributes to psoriasis recurrence after discontinuation of interleukin (IL)-17A blockers remains unclear. OBJECTIVE: To investigate the role of mast cells in ixekizumab treatment-associated urticaria (ITAUR) and assess the effect of urticaria eruption on psoriasis relapse. METHODS: A retrospective analysis was performed on biopsies of patients who experienced psoriasis relapse after discontinuation of ixekizumab. Transcriptomic and histopathologic features were assessed. Patterns were compared between patients with ITAUR and nonurticaria (NUR) as well as psoriasis-like mice with mast cell activation or inactivation. RESULTS: Patients with ITAUR experienced early relapse compared with NUR group after treatment withdrawal. Transcriptomic and histopathologic analyses revealed that patients with ITAUR had an elevated proportion of mast cells in resolved skin. Especially, the proportion of IL-17A+ mast cells was inversely correlated with the duration of remission. LIMITATIONS: The mechanism of mast cell activation in ITAUR has not been precisely elucidated. CONCLUSION: Ixekizumab treatment increases IL-17A+ mast cells in lesions of ITAUR, which is associated with early psoriasis relapse after ixekizumab withdrawal.


Assuntos
Anticorpos Monoclonais Humanizados , Psoríase , Urticária , Humanos , Animais , Camundongos , Interleucina-17 , Mastócitos , Estudos Retrospectivos , Psoríase/induzido quimicamente , Psoríase/tratamento farmacológico , Urticária/induzido quimicamente , Índice de Gravidade de Doença , Recidiva , Resultado do Tratamento
4.
Hepatology ; 76(6): 1706-1722, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35288960

RESUMO

BACKGROUND AND AIMS: Liver regeneration (LR) is vital for the recovery of liver function after hepatectomy. Limited regeneration capacity, together with insufficient remnant liver volume, is a risk factor for posthepatectomy liver failure (PHLF) resulting from small-for-size syndrome. Although inflammation plays an important role in controlling LR, the underlying mechanisms still remain obscure. APPROACH AND RESULTS: We identified C-C motif chemokine ligand (CCL) 5 as an important negative regulator for LR. CCL5 levels were elevated after partial hepatectomy (PHx), both in healthy donors of living donor liver transplantation (LT) and PHx mouse models. Ccl5 knockout mice displayed improved survival after 90% PHx and enhanced LR 36 h after 70% PHx. However, primary hepatocytes from Ccl5-/- mice exposed to growth factors in vitro showed no proliferation advantage compared to those from wild-type (WT) mice. Flow cytometry analysis showed that proportions of Ly6Clo macrophages were significantly increased in Ccl5-/- mice after 70% PHx. RNA-sequencing analysis revealed that sorted macrophages (CD11b+ Ly6Clo&hi ) manifested enhanced expression of reparative genes in Ccl5-/- mice compared to WT mice. Mechanistically, CCL5 induced macrophages toward proinflammatory Ly6Chi phenotype, thereby inhibiting the production of hepatocyte growth factor (HGF) through the C-C motif chemokine receptor (CCR) 1- and CCR5-mediated forkhead box O (FoxO) 3a pathways. Finally, blockade of CCL5 greatly optimized survival and boosted LR in the mouse PHx model. CONCLUSIONS: Our findings suggest that inhibition of CCL5 is a promising strategy to improve regeneration restoration by enhancing HGF secretion from reparative macrophages through the FoxO3a pathway, which may potentially reduce the mortality of PHLF.


Assuntos
Falência Hepática , Transplante de Fígado , Animais , Humanos , Camundongos , Proliferação de Células , Hepatectomia , Fator de Crescimento de Hepatócito , Hepatócitos/metabolismo , Ligantes , Fígado/metabolismo , Falência Hepática/cirurgia , Regeneração Hepática/fisiologia , Doadores Vivos , Macrófagos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Biochem Biophys Res Commun ; 590: 132-138, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-34974301

RESUMO

Osteocytes are master regulators of skeletal homeostasis. However, little is known about the molecular mechanism of their differentiation. Epigenetic regulations, especially H3K27me3 modification, play critical roles in cell differentiation. Here, we found that H3K27me3 in the loci of osteocyte-expressing genes decreased during osteocyte differentiation and that H3K27me3 demethylase, Utx, was bound to the loci of those genes. To investigate the physiological functions of Utx in vivo, we generated late osteoblast-to-osteocyte specific Utx knockout mice using Dmp1-cre mice (UtxΔOcy/ΔOcy). Micro CT analyses showed that UtxΔOcy/ΔOcy displayed osteopenic phenotypes with lower bone volume and trabecular number, and greater trabecular separation. Bone histomorphometric analysis showed that bone mineralization and formation were significantly lower in UtxΔOcy/ΔOcy. Furthermore, Dmp1 expression and the number of osteocytes were significantly decreased in UtxΔOcy/ΔOcy. These results suggest that Utx in Dmp1-expressing osteoblast/osteocyte positively regulates osteoblast-to-osteocyte differentiation through H3K27me3 modifications in osteocyte genes. Our results provide new insight into the molecular mechanism of osteocyte differentiation.


Assuntos
Diferenciação Celular , Histona Desmetilases/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Osteoblastos/citologia , Osteócitos/citologia , Animais , Sequência de Bases , Doenças Ósseas Metabólicas/genética , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/patologia , Contagem de Células , Diferenciação Celular/genética , Regulação para Baixo/genética , Epigenoma , Loci Gênicos , Histona Desmetilases/deficiência , Metilação , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/metabolismo , Osteócitos/metabolismo , Fenótipo , Processamento de Proteína Pós-Traducional , Transcriptoma/genética
6.
Cell Tissue Res ; 378(3): 561-562, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31637542

RESUMO

The authors regret that in our published paper entitled "Co-expression network analysis identified key genes in association with mesenchymal stem cell osteogenic differentiation" Cell Tissue Res (2019). https://doi.org/10.1007/s00441-019-03071-1; there is a typo in the text.

7.
Cell Tissue Res ; 378(3): 513-529, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31418071

RESUMO

Although several studies have shown that osteogenic differentiation of different mesenchymal stem cell (MSC) lines can be guided by the 3D scaffold with growth factors or biochemical agent, the key mechanism regulating osteogenic differentiation is not known yet. Here, this study was designed to investigate key genes that regulate the induction of osteogenesis by different MSC lines in different ways. Expression profiling by array (GSE58919 and GSE18043) was downloaded and analyzed using weighted gene co-expression network analysis (WGCNA) to narrow genes associated with osteogenic differentiation. A protein-protein interactive (PPI) network was built to find the key genes and the role of these key genes was confirmed by statistical analysis. To understand the function of genes associated with osteogenesis, gene ontology (GO) and the Kyoto encyclopedia of genes and genomes (KEGG) were analyzed, which showed that key genes in MSC osteogenic differentiation induced by a biochemical agent involve regulation of cell apoptosis and proliferation while key genes in MSC osteogenic differentiation induced by the 3D scaffold with growth factors involve regulation of cajal body and centromeres. Furthermore, 58 key genes are involved in Wnt signaling pathway, ion response and focal adhesion. Proteasome also played a key role in osteogenic differentiation. Seven potential key genes were found essential in the osteogenic differentiation of MSCs in the PPI network, especially the five key genes, CCT2, NOP58, FBL, EXOSC8 and SNRPD1. This study will provide important targets of MSC osteogenic differentiation that will help us understand the mechanism of osteogenic differentiation in MSCs.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Osteogênese/genética , Transporte Biológico Ativo , Adesão Celular , Diferenciação Celular , Linhagem Celular , Bases de Dados Genéticas , Redes Reguladoras de Genes , Humanos , Transporte de Íons , Células-Tronco Mesenquimais/citologia , Via de Sinalização Wnt
8.
J Gastroenterol Hepatol ; 34(3): 507-516, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30393891

RESUMO

BACKGROUND AND AIM: The thiopurines are effective in the management of patients with inflammatory bowel disease (IBD), but the association between thiopurines use and the risk of skin cancer (including nonmelanoma skin cancer [NMSC] and melanoma skin cancer) has already been sufficiently reported. However, the results of these studies are inconsistent, and thus, the objective of our analysis was to explore whether thiopurines can lead to an excess risk of skin cancer in IBD patients. METHODS: MEDLINE, EMBASE, and the Cochrane Library were searched to identify relevant studies that evaluated the risk of skin cancer in IBD patients treated with thiopurines. A random effects meta-analysis was conducted to calculate the pooled incidence rate ratios as well as risk ratios (RRs). Subgroup analysis was performed to explore the potential source of heterogeneity. RESULTS: Thirteen studies comprising 149 198 participants were included. The result suggested that thiopurines significantly increased the risk of overall skin cancer in IBD patients (random effects: RR = 1.80, 95% confidence interval [CI] 1.14-2.87, P = 0.013), among which NMSC showed an excess risk associated with thiopurines use (random effects: RR = 1.88, 95% CI 1.48-2.38, P < 0.001) while no increased risk was observed with respect to melanoma skin cancer (random effects: RR = 1.22, 95% CI 0.90-1.65, P = 0.206). Subgroup analysis regarding sample size and geographic distribution in skin cancer and follow-up duration in NMSC reached statistical significance, while other subgroups showed no significance. CONCLUSION: Exposition of thiopurines in patients with IBD is associated with a higher risk of skin cancer. Routine skin screening and daily skin protective practice are recommended for these patients.


Assuntos
Azatioprina/efeitos adversos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Melanoma/etiologia , Mercaptopurina/efeitos adversos , Neoplasias Cutâneas/etiologia , Azatioprina/uso terapêutico , Bases de Dados Bibliográficas , Humanos , Doenças Inflamatórias Intestinais/complicações , Melanoma/prevenção & controle , Mercaptopurina/uso terapêutico , Risco , Neoplasias Cutâneas/prevenção & controle
9.
Int J Med Sci ; 15(3): 257-268, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29483817

RESUMO

BMMSCs have drawn great interest in tissue engineering and regenerative medicine attributable to their multi-lineage differentiation capacity. Increasing evidence has shown that the mechanical stiffness of extracellular matrix is a critical determinant for stem cell behaviors. However, it remains unknown how matrix stiffness influences MSCs commitment with changes in cell morphology, adhesion, proliferation, self-renewal and differentiation. We employed fibronectin coated polyacrylamide hydrogels with variable stiffnesses ranging from 13 to 68 kPa to modulate the mechanical environment of BMMSCs and found that the morphology and adhesion of BMMSCs were highly dependent on mechanical stiffness. Cells became more spread and more adhesive on substrates of higher stiffness. Similarly, the proliferation of BMMSCs increased as stiffness increased. Sox2 expression was lower during 4h to 1 week on the 13-16 kPa and 62-68 kPa, in contrast, it was higher during 4h to 1 week on the 48-53 kPa. Oct4 expression on 13-16 kPa was higher than 48-53 kPa at 4h, and it has no significant differences at other time point among three different stiffness groups. On 62-68 kPa, BMMSCs were able to be induced toward osteogenic phenotype and generated a markedly high level of RUNX2, ALP, and Osteopontin. The cells exhibited a polygonal morphology and larger spreading area. These results suggest that matrix stiffness modulates commitment of BMMSCs. Our findings may eventually aid in the development of novel, effective biomaterials for the applications in tissue engineering.


Assuntos
Diferenciação Celular/genética , Células-Tronco Mesenquimais/citologia , Osteogênese/genética , Engenharia Tecidual , Adesão Celular/genética , Proliferação de Células/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Transplante de Células-Tronco Mesenquimais , Osteopontina/genética , Fatores de Transcrição SOXB1/genética , Alicerces Teciduais
10.
Differentiation ; 96: 30-39, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28753444

RESUMO

Mesenchymal stem cells (MSCs) are a compatible cellular alternative for regenerative medicine and tissue engineering because of their powerful multipotency. Matrix stiffness plays a profound role on stem cell behavior. Nevertheless, the effect of matrix stiffness on umbilical cordmesenchymal stem cells (UC-MSCs) remains unexplored. To conduct an in-depth exploration, we cultured UC-MSCs on different stiffness (Young's modulus: 13-16, 35-38, 48-53, and 62-68 kPa) polyacrylamide gels coated with fibronectin. We found that the proliferation and adhesion of UC-MSCs varied when cultured on the different matrices, and the spreading capacity was stronger as the stiffness increased (*P<0.05). Real-time quantitative PCR results showed that the soft matrix promoted adipogenic differentiation, with higher expression levels of adipocytic markers like PPARγ and C/EBPα (*P<0.05). In contrast, cells tended to differentiate into muscle when cultured on the 48-53 kPa matrix, which was validated by increased expression of myogenic makers like desminand MOYG (*P<0.05). Moreover, increased expression of osteoblastic makers (*P<0.05), such as ALP, collagen type I, osteocalcin, and Runx2, confirmed that cells differentiated into bone on the high-stiffness matrix.


Assuntos
Diferenciação Celular , Proliferação de Células , Módulo de Elasticidade , Células-Tronco Mesenquimais/citologia , Resinas Acrílicas/química , Adipócitos/citologia , Adipócitos/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Adesão Celular , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Fibronectinas/química , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Cordão Umbilical/citologia
11.
Artigo em Inglês | MEDLINE | ID: mdl-38946497

RESUMO

Meeting the exacting demands of wound healing encompasses rapid coagulation, superior exudate absorption, high antibacterial efficacy, and imperative support for cell growth. In this study, by emulating the intricate structure of natural skin, we prepare a multifunctional porous bilayer artificial skin to address these critical requirements. The bottom layer, mimicking the dermis, is crafted through freeze-drying a gel network comprising carboxymethyl chitosan (CMCs) and gelatin (GL), while the top layer, emulating the epidermis, is prepared via electrospinning poly(l-lactic acid) (PLLA) nanofibers. With protocatechuic aldehyde and gallium ion complexation (PA@Ga) as cross-linking agents, the bottom PA@Ga-CMCs/GL layer featured an adjustable pore size (78-138 µm), high hemostatic performance (67s), and excellent bacterial inhibition rate (99.9%), complemented by an impressive liquid-absorbing capacity (2000% swelling rate). The top PLLA layer, with dense micronanostructure and hydrophobic properties, worked as a shield to effectively thwarted liquid or bacterial penetration. Furthermore, accelerated wound closure, reduced inflammatory responses, and enhanced formation of hair follicles and blood vessels are achieved by the porous artificial skin covered on the surface of wound. Bilayer artificial skin integrates the advantages of nanofibers and freeze-drying porous materials to effectively replicate the protective properties of the epidermal layer of the skin, as well as the cell migration and tissue regeneration of the dermis. This bioabsorbable artificial skin demonstrates structural and functional comparability to real skin, which would advance the field of wound care through its multifaceted capabilities.

12.
J Mater Chem B ; 12(13): 3171-3190, 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38488129

RESUMO

Electrospun nanofiber membranes hold great promise as scaffolds for tissue reconstruction, mirroring the natural extracellular matrix (ECM) in their structure. However, their limited bioactive functions have hindered their effectiveness in fostering wound healing. Inorganic nanoparticles possess commendable biocompatibility, which can expedite wound healing; nevertheless, deploying them in the particle form presents challenges associated with removal or collection. To capitalize on the strengths of both components, electrospun organic/inorganic hybrid nanofibers (HNFs) have emerged as a groundbreaking solution for accelerating wound healing and maintaining stability throughout the healing process. In this review, we provide an overview of recent advancements in the utilization of HNFs for wound treatment. The review begins by elucidating various fabrication methods for hybrid nanofibers, encompassing direct electrospinning, coaxial electrospinning, and electrospinning with subsequent loading. These techniques facilitate the construction of micro-nano structures and the controlled release of inorganic ions. Subsequently, we delve into the manifold applications of HNFs in promoting the wound regeneration process. These applications encompass hemostasis, antibacterial properties, anti-inflammatory effects, stimulation of cell proliferation, and facilitation of angiogenesis. Finally, we offer insights into the prospective trends in the utilization of hybrid nanofiber-based wound dressings, charting the path forward in this dynamic field of research.


Assuntos
Nanofibras , Nanofibras/química , Estudos Prospectivos , Cicatrização , Antibacterianos/farmacologia , Bandagens
13.
Cell Prolif ; 57(1): e13519, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37322828

RESUMO

Epigenetic reprogramming during fertilization and somatic cell nuclear transfer (NT) is required for cell plasticity and competent development. Here, we characterize the epigenetic modification pattern of H4K20me3, a repressive histone signature in heterochromatin, during fertilization and NT reprogramming. Importantly, the dynamic H4K20me3 signature identified during preimplantation development in fertilized embryos differed from NT and parthenogenetic activation (PA) embryos. In fertilized embryos, only maternal pronuclei carried the canonical H4K20me3 peripheral nucleolar ring-like signature. H4K20me3 disappeared at the 2-cell stage and reappeared in fertilized embryos at the 8-cell stage and in NT and PA embryos at the 4-cell stage. H4K20me3 intensity in 4-cell, 8-cell, and morula stages of fertilized embryos was significantly lower than in NT and PA embryos, suggesting aberrant regulation of H4K20me3 in PA and NT embryos. Indeed, RNA expression of the H4K20 methyltransferase Suv4-20h2 in 4-cell fertilized embryos was significantly lower than NT embryos. Knockdown of Suv4-20h2 in NT embryos rescued the H4K20me3 pattern similar to fertilized embryos. Compared to control NT embryos, knockdown of Suv4-20h2 in NT embryos improved blastocyst development ratios (11.1% vs. 30.5%) and full-term cloning efficiencies (0.8% vs. 5.9%). Upregulation of reprogramming factors, including Kdm4b, Kdm4d, Kdm6a, and Kdm6b, as well as ZGA-related factors, including Dux, Zscan4, and Hmgpi, was observed with Suv4-20h2 knockdown in NT embryos. Collectively, these are the first findings to demonstrate that H4K20me3 is an epigenetic barrier of NT reprogramming and begin to unravel the epigenetic mechanisms of H4K20 trimethylation in cell plasticity during natural reproduction and NT reprogramming in mice.


Assuntos
Histonas , Técnicas de Transferência Nuclear , Animais , Camundongos , Histonas/genética , Histonas/metabolismo , Clonagem de Organismos , Epigênese Genética , Desenvolvimento Embrionário/genética , Reprogramação Celular/genética
15.
Mater Today Bio ; 25: 100958, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38327975

RESUMO

Cirrhosis is an aggressive disease, and over 80 % of liver cancer patients are complicated by cirrhosis, which lacks effective therapies. Transplantation of mesenchymal stem cells (MSCs) is a promising option for treating liver cirrhosis. However, this therapeutic approach is often challenged by the low homing ability and short survival time of transplanted MSCs in vivo. Therefore, a novel and efficient cell delivery system for MSCs is urgently required. This new system can effectively extend the persistence and duration of MSCs in vivo. In this study, we present novel porous microspheres with microfluidic electrospray technology for the encapsulation of bone marrow-derived MSCs (BMSCs) in the treatment of liver cirrhosis. Porous microspheres loaded with BMSCs (Mi-BMSCs) exhibit good biocompatibility and demonstrate better anti-inflammatory properties than BMSCs alone. Mi-BMSCs significantly increase the duration of BMSCs and exert potent anti-inflammatory and anti-fibrosis effects against CCl4 and TAA-induced liver cirrhosis by targeting the TGF-ß/Smad signaling pathway to ameliorate cirrhosis, which highlight the potential of Mi-BMSCs as a promising therapeutic approach for early liver cirrhosis.

16.
Theriogenology ; 221: 25-30, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38537318

RESUMO

The objectives of this study were to analyze the (1) effects of donor age and multiparity on development of in vitro fertilization (IVF) embryos after ovum pickup (OPU), (2) effects of repeated and consecutive OPU-IVF procedures on embryo development, and (3) embryo production from OPU-IVF in donors with differing embryo yields after multiple ovulation and embryo transfer technology (MOET) in Japanese Black cattle (Wagyu). Donors were pre-treated with low-dosage follicle-stimulating hormone (FSH; 200 IU total), and oocytes were collected via OPU and fertilized by IVF to generate blastocysts. The number of oocytes collected per OPU session per donor was lower in heifers (2-4 years old, 5.3 oocytes) than in primiparous and pluriparous cows (2-10 years old, 13.6-19.1 oocytes; P < 0.05). Rates of blastocyst development for oocytes from heifers (33.1%) were lower than for those from cows (2-10 years old, 44.1-54.3%; P < 0.05), and average blastocyst yield/OPU/animal was lower in heifers (3.7) than in 5-6 years old cows (10.1; P < 0.05). Donors undergoing five consecutive OPU-IVF sessions after low-dosage FSH showed similar oocyte retrieval (12.2-15.1 oocytes per OPU/animal), blastocyst development rates (35.6-45.0%), and embryo yield/OPU/animal (4.8-5.8; P > 0.05) across sessions. Additionally, embryo yield from OPU-IVF was significantly improved in animals with previous low embryo yield from MOET (5.9 vs. 2.6, respectively, P < 0.05). These results indicate that Wagyu cows with previous births can be more productive as OPU-IVF donors than heifers, and oocytes from donors undergoing to five consecutive OPU-IVF cycles are competent for embryo development without loss of embryo yield/OPU/animal. Moreover, OPU-IVF can be used for embryo production and breeding from all elite Japanese Black cattle, regardless of previous low embryo yield in routine MOET.


Assuntos
Oócitos , História Reprodutiva , Bovinos , Feminino , Animais , Fertilização in vitro/veterinária , Recuperação de Oócitos/veterinária , Recuperação de Oócitos/métodos , Hormônio Foliculoestimulante/farmacologia , Óvulo
17.
Elife ; 122023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36779851

RESUMO

M-CSF is a critical growth factor for myeloid lineage cells, including monocytes, macrophages, and osteoclasts. Tissue-resident macrophages in most organs rely on local M-CSF. However, it is unclear what specific cells in the bone marrow produce M-CSF to maintain myeloid homeostasis. Here, we found that Adipoq-lineage progenitors but not mature adipocytes in bone marrow or in peripheral adipose tissue, are a major cellular source of M-CSF, with these Adipoq-lineage progenitors producing M-CSF at levels much higher than those produced by osteoblast lineage cells. The Adipoq-lineage progenitors with high CSF1 expression also exist in human bone marrow. Deficiency of M-CSF in bone marrow Adipoq-lineage progenitors drastically reduces the generation of bone marrow macrophages and osteoclasts, leading to severe osteopetrosis in mice. Furthermore, the osteoporosis in ovariectomized mice can be significantly alleviated by the absence of M-CSF in bone marrow Adipoq-lineage progenitors. Our findings identify bone marrow Adipoq-lineage progenitors as a major cellular source of M-CSF in bone marrow and reveal their crucial contribution to bone marrow macrophage development, osteoclastogenesis, bone homeostasis, and pathological bone loss.


Assuntos
Fator Estimulador de Colônias de Macrófagos , Osteogênese , Camundongos , Humanos , Animais , Fator Estimulador de Colônias de Macrófagos/metabolismo , Medula Óssea , Diferenciação Celular , Macrófagos/metabolismo , Osteoclastos/metabolismo , Células da Medula Óssea/metabolismo , Camundongos Endogâmicos C57BL , Adiponectina/metabolismo
18.
Ital J Pediatr ; 49(1): 2, 2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36600293

RESUMO

BACKGROUND: Kawasaki disease (KD) is a kind of vasculitis with unidentified etiology. Given that the current diagnosis and therapeutic strategy of KD are mainly dependent on clinical experiences, further research to explore its pathological mechanisms is warranted. METHODS: Enzyme linked immunosorbent assay (ELISA) was used to measure the serum levels of SIGIRR, TLR4 and caspase-8. Western blotting was applied to determine protein levels, and flow cytometry was utilized to analyze cell apoptosis. Hematoxylin eosin (HE) staining and TUNEL staining were respectively used to observe coronary artery inflammation and DNA fragmentation. RESULTS: In this study, we found the level of SIGIRR was downregulated in KD serum and KD serum-treated endothelial cells. However, the level of caspase-8 was increased in serum from KD patients compared with healthy control (HC). Therefore, we hypothesized that SIGIRR-caspase-8 signaling may play an essential role in KD pathophysiology. In vitro experiments demonstrated that endothelial cell apoptosis in the setting of KD was associated with caspase-8 activation, and SIGIRR overexpression alleviated endothelial cell apoptosis via inhibiting caspase-8 activation. These findings were also recapitulated in the Candida albicans cell wall extracts (CAWS)-induced KD mouse model. CONCLUSION: Our data suggest that endothelial cell apoptosis mediated by SIGIRR-caspase-8 signaling plays a crucial role in coronary endothelial damage, providing potential targets to treat KD.


Assuntos
Células Endoteliais , Síndrome de Linfonodos Mucocutâneos , Animais , Camundongos , Humanos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Caspase 8/metabolismo , Apoptose , Transdução de Sinais
19.
PLoS One ; 18(3): e0277155, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36913356

RESUMO

Based on the relationship between the gut microbiota and colorectal cancer, we developed a new probiotic powder for treatment of colorectal cancer. Initially, we evaluated the effect of the probiotic powder on CRC using hematoxylin and eosin staining, and evaluated mouse survival rate and tumor size. We then investigated the effects of the probiotic powder on the gut microbiota, immune cells, and apoptotic proteins using 16S rDNA sequencing, flow cytometry, and western blot, respectively. The results showed that the probiotic powder improved the intestinal barrier integrity, survival rate, and reduced tumor size in CRC mice. This effect was associated with changes in the gut microbiota. Specifically, the probiotic powder increased the abundance of Bifidobacterium animalis and reduced the abundance of Clostridium cocleatum. In addition, the probiotic powder resulted in decreased numbers of CD4+ Foxp3+ Treg cells, increased numbers of IFN-γ+ CD8+ T cells and CD4+ IL-4+ Th2 cells, decreased expression of the TIGIT in CD4+ IL-4+ Th2 cells, and increased numbers of CD19+ GL-7+ B cells. Furthermore, the expression of the pro-apoptotic protein BAX was significantly increased in tumor tissues in response to the probiotic powder. In summary, the probiotic powder ameliorated CRC by regulating the gut microbiota, reducing Treg cell abundance, promoting the number of IFN-γ+ CD8+ T cells, increasing Th2 cell abundance, inhibiting the expression of TIGIT in Th2 cells, and increasing B cell abundance in the immune microenvironment of CRC, thereby increasing the expression of BAX in CRC.


Assuntos
Bifidobacterium animalis , Neoplasias Colorretais , Probióticos , Camundongos , Animais , Pós , Interleucina-4 , Proteína X Associada a bcl-2 , Probióticos/farmacologia , Probióticos/uso terapêutico , Neoplasias Colorretais/terapia , Neoplasias Colorretais/patologia , Microambiente Tumoral
20.
Macromol Biosci ; : e2300399, 2023 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-38011585

RESUMO

Sustainable and controllable drug transport is one of the most efficient ways of disease treatment. Due to high biocompatibility, good biodegradability, and low costs, chitosan and its derivatives are widely used in biomedical fields. Specifically, chitosan hydrogel enables drugs to pass through biological barriers because of their abundant amino and hydroxyl groups that can interact with human tissues. Moreover, the multi-responsive nature (pH, temperature, ions strength, and magnetic field, etc.) of chitosan hydrogels makes precise drug release a possibility. Here, the synthesis methods, modification strategies, stimuli-responsive mechanisms of chitosan-based hydrogels, and their recent progress in drug delivery are summarized. Chitosan hydrogels that carry and release drugs through subcutaneous (dealing with wound dressing), oral (dealing with gastrointestinal tract), and facial (dealing with ophthalmic, ear, and brain) are reviewed. Finally, challenges toward clinic application and the future prospects of stimuli-responsive chitosan-based hydrogels are indicated.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa