Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell Commun Signal ; 22(1): 6, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38166927

RESUMEN

Ferroptosis is a newly discovered form of cell death that is featured in a wide range of diseases. Exosome therapy is a promising therapeutic option that has attracted much attention due to its low immunogenicity, low toxicity, and ability to penetrate biological barriers. In addition, emerging evidence indicates that exosomes possess the ability to modulate the progression of diverse diseases by regulating ferroptosis in damaged cells. Hence, the mechanism by which cell-derived and noncellular-derived exosomes target ferroptosis in different diseases through the system Xc-/GSH/GPX4 axis, NAD(P)H/FSP1/CoQ10 axis, iron metabolism pathway and lipid metabolism pathway associated with ferroptosis, as well as its applications in liver disease, neurological diseases, lung injury, heart injury, cancer and other diseases, are summarized here. Additionally, the role of exosome-regulated ferroptosis as an emerging repair mechanism for damaged tissues and cells is also discussed, and this is expected to be a promising treatment direction for various diseases in the future. Video Abstract.


Asunto(s)
Exosomas , Ferroptosis , Lesión Pulmonar , Humanos , Muerte Celular , NAD
2.
J Nanobiotechnology ; 20(1): 95, 2022 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-35209908

RESUMEN

BACKGROUND: The promising therapeutic strategy for the treatment of peripheral artery disease (PAD) is to restore blood supply and promote regeneration of skeletal muscle regeneration. Increasing evidence revealed that prostaglandin E2 (PGE2), a lipid signaling molecule, has significant therapeutic potential for tissue repair and regeneration. Though PGE2 has been well reported in tissue regeneration, the application of PGE2 is hampered by its short half-life in vivo and the lack of a viable system for sustained release of PGE2. RESULTS: In this study, we designed and synthesized a new PGE2 release matrix by chemically bonding PGE2 to collagen. Our results revealed that the PGE2 matrix effectively extends the half-life of PGE2 in vitro and in vivo. Moreover, the PGE2 matrix markedly improved neovascularization by increasing angiogenesis, as confirmed by bioluminescence imaging (BLI). Furthermore, the PGE2 matrix exhibits superior therapeutic efficacy in the hindlimb ischemia model through the activation of MyoD1-mediated muscle stem cells, which is consistent with accelerated structural recovery of skeletal muscle, as evidenced by histological analysis. CONCLUSIONS: Our findings highlight the chemical bonding strategy of chemical bonding PGE2 to collagen for sustained release and may facilitate the development of PGE2-based therapies to significantly improve tissue regeneration.


Asunto(s)
Dinoprostona , Neovascularización Fisiológica , Animales , Modelos Animales de Enfermedad , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Isquemia/tratamiento farmacológico , Isquemia/patología , Músculo Esquelético
3.
J Biol Chem ; 295(34): 12203-12213, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32641493

RESUMEN

Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have been shown to stimulate regeneration in the treatment of kidney injury. Renal regeneration is also thought to be stimulated by the activation of Sox9+ cells. However, whether and how the activation mechanisms underlying EV treatment and Sox9+ cell-dependent regeneration intersect is unclear. We reasoned that a high-resolution imaging platform in living animals could help to untangle this system. To test this idea, we first applied EVs derived from human placenta-derived MSCs (hP-MSCs) to a Sox9-CreERT2; R26mTmG transgenic mouse model of acute kidney injury (AKI). Then, we developed an abdominal imaging window in the mouse and tracked the Sox9+ cells in the inducible Sox9-Cre transgenic mice via in vivo lineage tracing with two-photon intravital microscopy. Our results demonstrated that EVs can travel to the injured kidneys post intravenous injection as visualized by Gaussia luciferase imaging and markedly increase the activation of Sox9+ cells. Moreover, the two-photon living imaging of lineage-labeled Sox9+ cells showed that the EVs promoted the expansion of Sox9+ cells in kidneys post AKI. Histological staining results confirmed that the descendants of Sox9+ cells contributed to nephric tubule regeneration which significantly ameliorated the renal function after AKI. In summary, intravital lineage tracing with two-photon microscopy through an embedded abdominal imaging window provides a practical strategy to investigate the beneficial functions and to clarify the mechanisms of regenerative therapies in AKI.


Asunto(s)
Lesión Renal Aguda , Vesículas Extracelulares/trasplante , Riñón/fisiología , Células Madre Mesenquimatosas/metabolismo , Regeneración , Factor de Transcripción SOX9/metabolismo , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/terapia , Animales , Vesículas Extracelulares/metabolismo , Humanos , Microscopía Intravital , Riñón/lesiones , Células Madre Mesenquimatosas/patología , Ratones , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Factor de Transcripción SOX9/genética
4.
BMC Pediatr ; 21(1): 102, 2021 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-33639900

RESUMEN

BACKGROUND: Defects of bone marrow mesenchymal stem cells (BM-MSCs) in proliferation and differentiation are involved in the pathophysiology of aplastic anemia (AA). Infusion of umbilical cord mesenchymal stem cells (UC-MSCs) may improve the efficacy of immunosuppressive therapy (IST) in childhood severe aplastic anemia (SAA). METHODS: We conducted an investigator-initiated, open-label, and prospective phase IV trial to evaluate the safety and efficacy of combination of allogenic UC-MSCs and standard IST for pediatric patients with newly diagnosed SAA. In mesenchymal stem cells (MSC) group, UC-MSCs were injected intravenously at a dose of 1 × 106/kg per week starting on the 14th day after administration of rabbit antithymocyte globulin (ATG), for a total of 3 weeks. The clinical outcomes and adverse events of patients with UC-MSCs infusion were assessed when compared with a concurrent control group in which patients received standard IST alone. RESULTS: Nine patients with a median age of 4 years were enrolled as the group with MSC, while the data of another 9 childhood SAA were analysed as the controls. Four (44%) patients in MSC group developed anaphylactic reactions which were associated with rabbit ATG. When compared with the controls, neither the improvement of blood cell counts, nor the change of T-lymphocytes after IST reached statistical significance in MSC group (both p > 0.05) and there were one (11%) patient in MSC group and two (22%) patients in the controls achieved partial response (PR) at 90 days after IST. After a median follow-up of 48 months, there was no clone evolution occurring in both groups. The 4-year estimated overall survival (OS) rate in two groups were both 88.9% ± 10.5%, while the 4-year estimated failure-free survival (FFS) rate in MSC group was lower than that in the controls (38.1% ± 17.2% vs. 66.7% ± 15.7%, p = 0.153). CONCLUSIONS: Concomitant use of IST and UC-MSCs in SAA children is safe but may not necessarily improve the early response rate and long-term outcomes. This clinical trial was registered at ClinicalTrials.gov, identifier: NCT02218437 (registered October 2013).


Asunto(s)
Anemia Aplásica , Células Madre Mesenquimatosas , Anemia Aplásica/terapia , Niño , Ciclosporina , Humanos , Estudios Prospectivos , Resultado del Tratamiento , Cordón Umbilical
5.
Appl Opt ; 57(23): 6835-6839, 2018 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-30129634

RESUMEN

A simple all-fiber sensor based on nested fiber balloon rings (FBRs) for simultaneous refractive index (RI) and temperature measurement is proposed and demonstrated. The FBR is easily fabricated by properly bending a section of single-mode fiber. The FBR with a smaller bending radius is embedded in a bigger one. Because the two separate interference dips have different RI and temperature sensitivities, which are formed by individual FBRs, the RI and temperature can be simultaneously measured by interrogating the wavelength shifts of the two interference dips. This sensor has the advantages of convenient fabrication, economical materials, probe structure, and high sensitivity and can be a candidate for simultaneous measurement of RI and temperature.

6.
Cell Physiol Biochem ; 36(5): 1991-2002, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26202359

RESUMEN

BACKGROUND: The Notch signaling pathway is implicated in a broad range of developmental processes, including cell fate decisions. This study was designed to determine the role of Notch signaling in adipogenic differentiation of human bone marrow derived MSCs (BM-MSCs). METHODS: The Notch signaling was inhibited by the γ-secretase inhibitor N-[N-(3,5-difluor- ophenacetyl-L-alanyl)]-S-phenylglycine t-butylester (DAPT). The markers involving adipogenic differentiation of MSCs, the relative pathway PTEN-PI3K/Akt/mTOR and autophagy activation were then analyzed. Furthermore, the autophagy inhibitor chloroquine (CQ) and 3-methyladenine (3-MA) were used to study the role of autophagy in the DAPT-induced the adipogenic differentiation of MSCs. RESULTS: We first confirmed the down -regulation of Notch gene expression during MSCs adipocyte differentiation, and showed that the inhibition of Notch signaling significantly enhanced adipogenic differentiation of MSCs. Furthermore, Notch inhibitor DAPT induced early autophagy by acting on PTEN-PI3K/Akt/mTOR pathway. The autophagy inhibitor CQ and 3-MA dramatically abolished the effects of DAPT-induced autophagy and adipogenic differentiation of MSCs. CONCLUSION: Our results indicate that inhibition of Notch signaling could promote MSCs adipogenesis mediated by autophagy involving PTEN-PI3K/Akt/mTOR pathway. Notch signaling could be a novel target for regulating the adipogenic differentiation of MSCs.


Asunto(s)
Tejido Adiposo/citología , Autofagia , Diferenciación Celular , Dipéptidos/farmacología , Células Madre Mesenquimatosas/citología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Notch/antagonistas & inhibidores , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Células Cultivadas , Humanos , Receptores Notch/metabolismo
7.
Cell Physiol Biochem ; 33(3): 569-80, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24603109

RESUMEN

BACKGROUND: Mesenchymal stem cells are capable of self-renewal and multi-lineage differentiation. They are used extensively to treat several diseases. Traditionally, mesenchymal stem cells are cultured in serum-containing media, typically supplemented with fetal bovine serum (FBS). However, the variability of FBS is likely to skew experimental results. Although serum-free media used to expand mesenchymal stem cells has facilitated remarkable achievements, immunomodulation of these cells in under serum-free conditions is poorly understood. We hypothesized that mesenchymal stem cells expanded in serum-free media will retain powerful immunoregulatory functions in vitro and in vivo. DESIGN AND METHODS: Immunosuppressive activity and the immunomodulatory cytokines produced by mesenchymal stem cells in serum-free media were characterized in vitro. Immunomodulation by serum-free mesenchymal stem cell expansion in monocrotaline-induced pulmonary hypertension was explored in vivo. RESULTS: Similar to cells in serum-containing media, mesenchymal stem cells expanded in serum-free media inhibited proliferation and apoptosis of CD4(+)T cells. They also exhibited strong immunosuppressive activities and secreted high levels of immunomodulatory cytokines such as PGE2, IDO1, COX2, IL-6, and IL-1ß, but not HGF. On the other hand, growth of mesenchymal stem cells in serum-free media attenuated pulmonary vascular remodeling and inhibited mRNA expression of proinflammatory cytokines TNF-α, IFN-γ, IL-6, IL-1ß, and IL-18. CONCLUSIONS: Mesenchymal stem cells in serum-free media maintained powerful immunomodulatory function in vitro and in vivo; serum-free media may replace serum-containing media for basic research and clinical applications.


Asunto(s)
Apoptosis/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Animales , Apoptosis/inmunología , Bovinos , Ciclooxigenasa 2/inmunología , Citocinas/inmunología , Dinoprostona/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Células Madre Mesenquimatosas/citología
8.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 31(6): 1319-24, 2014 Dec.
Artículo en Zh | MEDLINE | ID: mdl-25868252

RESUMEN

Recombinant protein SMB(PRG4) containing two Somatomedin B domains and a small amount of glycosylation of repetitive sequences of proteoglycan 4 was cloned according to PGR4 gene polymorphism. Mature purification process was established and recombinant protein SMB(PRG4), with high-level expression was purified. By using size-exclusion chromatogaraphy and dynamic light scattering, we found that the recombinant protein self-aggregate to dimeric form. Structure prediction and non-reducing electrophoresis revealed that SMB(PRG4), was a non-covalently bonded dimer.


Asunto(s)
Proteoglicanos/química , Proteínas Recombinantes/química , Somatomedinas/química , Glicosilación , Multimerización de Proteína
9.
Cell Prolif ; : e13679, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38801100

RESUMEN

Uncovering mechanisms of endogenous regeneration and repair through resident stem cell activation will allow us to develop specific therapies for injuries and diseases by targeting resident stem cell lineages. Sox9+ stem cells have been reported to play an essential role in acute kidney injury (AKI). However, a complete view of the Sox9+ lineage was not well investigated to accurately elucidate the functional end state and the choice of cell fate during tissue repair after AKI. To identify the mechanisms of fate determination of Sox9+ stem cells, we set up an AKI model with prostaglandin E2 (PGE2) treatment in a Sox9 lineage tracing mouse model. Single-cell RNA sequencing (scRNA-seq) was performed to analyse the transcriptomic profile of the Sox9+ lineage. Our results revealed that PGE2 could activate renal Sox9+ cells and promote the differentiation of Sox9+ cells into renal proximal tubular epithelial cells and inhibit the development of fibrosis. Furthermore, single-cell transcriptome analysis demonstrated that PGE2 could regulate the restoration of lipid metabolism homeostasis in proximal tubular epithelial cells by participating in communication with different cell types. Our results highlight the prospects for the activation of endogenous renal Sox9+ stem cells with PGE2 for the regenerative therapy of AKI.

10.
Signal Transduct Target Ther ; 9(1): 102, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38653983

RESUMEN

Patients with refractory immune thrombocytopenia (ITP) frequently encounter substantial bleeding risks and demonstrate limited responsiveness to existing therapies. Umbilical cord-derived mesenchymal stem cells (UC-MSCs) present a promising alternative, capitalizing on their low immunogenicity and potent immunomodulatory effects for treating diverse autoimmune disorders. This prospective phase I trial enrolled eighteen eligible patients to explore the safety and efficacy of UC-MSCs in treating refractory ITP. The research design included administering UC-MSCs at escalating doses of 0.5 × 106 cells/kg, 1.0 × 106 cells/kg, and 2.0 × 106 cells/kg weekly for four consecutive weeks across three cohorts during the dose-escalation phase, followed by a dose of 2.0 × 106 cells/kg weekly for the dose-expansion phase. Adverse events, platelet counts, and changes in peripheral blood immunity were monitored and recorded throughout the administration and follow-up period. Ultimately, 12 (with an addition of three patients in the 2.0 × 106 cells/kg group due to dose-limiting toxicity) and six patients were enrolled in the dose-escalation and dose-expansion phase, respectively. Thirteen patients (13/18, 72.2%) experienced one or more treatment emergent adverse events. Serious adverse events occurred in four patients (4/18, 22.2%), including gastrointestinal hemorrhage (2/4), profuse menstruation (1/4), and acute myocardial infarction (1/4). The response rates were 41.7% in the dose-escalation phase (5/12, two received 1.0 × 106 cells/kg per week, and three received 2.0 × 106 cells/kg per week) and 50.0% (3/6) in the dose-expansion phase. The overall response rate was 44.4% (8/18) among all enrolled patients. To sum up, UC-MSCs are effective and well tolerated in treating refractory ITP (ClinicalTrials.gov ID: NCT04014166).


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Púrpura Trombocitopénica Idiopática , Humanos , Femenino , Masculino , Púrpura Trombocitopénica Idiopática/terapia , Púrpura Trombocitopénica Idiopática/inmunología , Persona de Mediana Edad , Adulto , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/inmunología , Cordón Umbilical/citología , Estudios Prospectivos , Anciano
11.
Cell Physiol Biochem ; 31(6): 823-32, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23816858

RESUMEN

BACKGROUND: CD151 is highly expressed in breast cancer cells and has been shown to accelerate breast cancer by enhancing cell growth and motility, but its regulation is poorly understood. To explore post-translation regulation of CD151, for example microRNAs, will be of great importance to claim the mechanism. METHODS: A luciferase reporter assay was used to determine whether CD151 was a target of miR-124. The levels of CD151 mRNA were detected by real-time PCR and CD151 protein expression was measured by western blot and flow cytometry. The effects of miR-124 expression on growth, apoptosis, cell cycle and motility of breast cancer cells were determined. RESULTS: We discovered that miR-124 directly targets the 3' untranslated region (3'-UTR) of CD151 mRNAs and suppresses its mRNA expression and protein translation. Both siRNA of CD151 and miR-124 mimics could significantly inhibit proliferation of breast cancer cell lines via cell cycle arrest but does not induce apoptosis. Meanwhile, miR-124 mimics significantly inhibited the motility of breast cancer cells. CONCLUSION: miR-124 plays a critical role in inhibiting the invasive and metastatic potential of breast cancer cells, probably by directly targeting the CD151 genes. Our findings highlight an important role of miR-124 in the regulation of invasion and metastasis by breast cancer cells and suggest a potential application for miR-124 in breast cancer treatment.


Asunto(s)
MicroARNs/metabolismo , Tetraspanina 24/metabolismo , Regiones no Traducidas 3' , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Células MCF-7 , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Tetraspanina 24/antagonistas & inhibidores , Tetraspanina 24/genética
12.
Cytotherapy ; 15(4): 423-33, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23352460

RESUMEN

BACKGROUND AIMS: Toll-like receptors (TLRs) play an important role in innate and adaptive immunity by recognizing pathogen-associated molecular patterns (PAMPs). METHODS: In the present study, we investigated the expression and role of TLRs on human umbilical cord mesenchymal stromal cells (UC-MSCs). The proliferation, differentiation and immunoregulatory activity of UC-MSCs primed with or without TLR ligands were determined. RESULTS: At the RNA level, the expression of TLR2, 4, 6 and 9 was relatively higher than that of other TLRs. However, TLR3 and TLR4 expression were relatively higher at the protein level. UC-MSCs expressed functional TLRs by nuclear factor-κB activation and cytokine expression assay. Poly-inosinic acid:cytidylic acid [Poly(I:C)] stimulation inhibited the proliferation of UC-MSCs, but the ligand of other TLRs had no significant effect. Poly(I:C) stimulation enhanced the adipogenic differentiation capability of UC-MSCs, but lipopolysaccharide inhibited the adipogenic differentiation. Poly(I:C) and CpG-oligonucleotide promoted the immunosuppressive potentiality of UC-MSCs, accompanied with the phosphorylation of interferon regulatory factor 3 (IRF3) and increased expression of indoleamine 2,3-dioxygenase and interferon ß, whereas activation of other TLR ligands (synthetic analog fibroblast-stimulating lipopeptide-1 and lipopolysaccharide) failed to affect the immunoregulatory activity of UC-MSCs. CONCLUSIONS: Taken together, our data demonstrated that TLR activation influenced the function of UC-MSCs, which might have important implications in future efforts to explore the clinical potentials of UC-MSCs.


Asunto(s)
Células Madre Mesenquimatosas/citología , Receptores Toll-Like/metabolismo , Cordón Umbilical/citología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Activación Enzimática , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/biosíntesis , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Fosforilación , Poli I-C/farmacología , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 3/biosíntesis , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 6/biosíntesis , Receptor Toll-Like 6/metabolismo , Receptor Toll-Like 9/biosíntesis , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/biosíntesis
13.
Water Res ; 242: 120259, 2023 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-37390660

RESUMEN

Heavy metal contamination of waste activated sludge (WAS) is a key factor limiting the land application of sludge for nutrients recovery. This study proposes a novel free nitrous acid (FNA)-assisted asymmetrical alternating current electrochemistry (FNA-AACE) process to achieve high-efficiency decontamination of multi-heavy metals (Cd, Pb, and Fe) in WAS. The optimal operating conditions, the heavy metal removal performance of FNA-AACE, and the related mechanisms for maintaining the high performance were systematically investigated. During the FNA-AACE process, FNA treatment was optimal with an exposure time of 13 h at a pH of 2.9 and an FNA concentration of 0.6 mg/g TSS. Then the sludge was washed with EDTA in a recirculating leaching system under asymmetrical alternating current electrochemistry (AACE). The 6-h working and the following electrode cleaning were defined as a working circle of AACE. After three cycles of working-cleaning periods in AACE treatment, the cumulative removal efficiency of the toxic metals Cd and Pb reached over 97% and 93%, respectively, whilst that of Fe was greater than 65%. This surpasses most previously reported efficiencies and possesses a shorter treatment duration and sustainable EDTA circulation. The mechanism analysis suggested that FNA pretreatment provoked the migration of heavy metals for leaching enhancement, as well as reduced the demand for EDTA eluent concentration and increased conductivity, which can improve the AACE efficiency. Meanwhile, the AACE process absorbed the anionic chelates of heavy metals and reduced them to zero-valent particles on the electrode, regenerating the EDTA eluent and maintaining its high extraction efficiency for heavy metals. In addition, FNA-AACE could provide different electric field operation modes, allowing it to have flexibility for the real application processes. This proposed process is expected to be coupled with anaerobic digestion in wastewater treatment plants (WWTPs) for high efficiency of heavy metal decontamination, sludge reduction, and resource/energy recovery.


Asunto(s)
Metales Pesados , Aguas del Alcantarillado , Ácido Nitroso , Ácido Edético , Cadmio , Descontaminación , Electroquímica , Plomo , Metales Pesados/análisis
14.
Stem Cell Res Ther ; 14(1): 48, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36949464

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) have demonstrated remarkable therapeutic promise for acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS). MSC secretomes contain various immunoregulatory mediators that modulate both innate and adaptive immune responses. Priming MSCs has been widely considered to boost their therapeutic efficacy for a variety of diseases. Prostaglandin E2 (PGE2) plays a vital role in physiological processes that mediate the regeneration of injured organs. METHODS: This work utilized PGE2 to prime MSCs and investigated their therapeutic potential in ALI models. MSCs were obtained from human placental tissue. MSCs were transduced with firefly luciferase (Fluc)/eGFP fusion protein for real-time monitoring of MSC migration. Comprehensive genomic analyses explored the therapeutic effects and molecular mechanisms of PGE2-primed MSCs in LPS-induced ALI models. RESULTS: Our results demonstrated that PGE2-MSCs effectively ameliorated lung injury and decreased total cell numbers, neutrophils, macrophages, and protein levels in bronchoalveolar lavage fluid (BALF). Meanwhile, treating ALI mice with PGE2-MSCs dramatically reduced histopathological changes and proinflammatory cytokines while increasing anti-inflammatory cytokines. Furthermore, our findings supported that PGE2 priming improved the therapeutic efficacy of MSCs through M2 macrophage polarization. CONCLUSION: PGE2-MSC therapy significantly reduced the severity of LPS-induced ALI in mice by modulating macrophage polarization and cytokine production. This strategy boosts the therapeutic efficacy of MSCs in cell-based ALI therapy.


Asunto(s)
Lesión Pulmonar Aguda , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Embarazo , Femenino , Ratones , Humanos , Animales , Lipopolisacáridos/toxicidad , Dinoprostona/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Placenta/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/terapia , Lesión Pulmonar Aguda/metabolismo , Células Madre Mesenquimatosas/metabolismo , Citocinas/metabolismo , Inmunomodulación , Macrófagos/metabolismo , Inmunidad , Pulmón/patología
15.
Elife ; 122023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37695201

RESUMEN

Nitric oxide (NO), as a gaseous therapeutic agent, shows great potential for the treatment of many kinds of diseases. Although various NO delivery systems have emerged, the immunogenicity and long-term toxicity of artificial carriers hinder the potential clinical translation of these gas therapeutics. Mesenchymal stem cells (MSCs), with the capacities of self-renewal, differentiation, and low immunogenicity, have been used as living carriers. However, MSCs as gaseous signaling molecule (GSM) carriers have not been reported. In this study, human MSCs were genetically modified to produce mutant ß-galactosidase (ß-GALH363A). Furthermore, a new NO prodrug, 6-methyl-galactose-benzyl-oxy NONOate (MGP), was designed. MGP can enter cells and selectively trigger NO release from genetically engineered MSCs (eMSCs) in the presence of ß-GALH363A. Moreover, our results revealed that eMSCs can release NO when MGP is systemically administered in a mouse model of acute kidney injury (AKI), which can achieve NO release in a precise spatiotemporal manner and augment the therapeutic efficiency of MSCs. This eMSC and NO prodrug system provides a unique and tunable platform for GSM delivery and holds promise for regenerative therapy by enhancing the therapeutic efficiency of stem cells.


Animals are made up of cells of different types, with each type of cell specializing on a specific role. But for the body to work properly, the different types of cells must be able to coordinate with each other to respond to internal and external stimuli. This can be achieved through signaling molecules, that is, molecules released by a cell that can elicit a specific response in other cells. There are many types of different molecules, including hormones and signaling proteins. Gases can also be potent signaling molecules, participating in various biological processes. Nitric oxide (NO) is a gas signaling molecule that can freely diffuse through the membranes of cells and has roles in blood vessel constriction and other disease processes, making it a promising therapeutic agent. Unfortunately, using artificial carriers to deliver nitric oxide to the organs and tissues where it is needed can lead to issues, including immune reactions to the carrier and long-term toxicity. One way to avoid these effects is by using cells to deliver nitric oxide to the right place. Huang, Qian, Liu et al. have used mesenchymal stem cells ­ which usually develop to form connective tissues such as bone and muscle ­ to develop a cell-based NO-delivery system. The researchers genetically modified the mesenchymal stem cells to produce a compound called ß-GALH363A. On its own ß-GALH363A does not do much, but in its presence, a non-toxic, non-reactive compound developed by Huang, Qian, Liu et al., called MGP, can drive the release of NO from cells. To confirm the usefulness of their cells as a delivery system, Huang, Qian, Liu et al. transplanted some of the genetically modified mesenchymal stem cells into the kidneys of mice, and then showed that when these mice were given MGP, the levels of NO increased in the kidneys but not in other organs. This result confirms that the cell-based delivery system provides spatial and temporal control of the production of NO. These findings demonstrate a new delivery system for therapies using gas molecules, which can be controlled spatiotemporally in mice. In the future, these types of systems could be used in the clinic for long-term treatment of conditions where artificial carriers could lead to complications.


Asunto(s)
Lesión Renal Aguda , Células Madre Mesenquimatosas , Ratones , Animales , Humanos , Óxido Nítrico , Células Madre , Ingeniería Genética , Lesión Renal Aguda/terapia
16.
Adv Sci (Weinh) ; 10(3): e2204626, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36416304

RESUMEN

Endothelial cell injury plays a critical part in ischemic acute kidney injury (AKI) and participates in the progression of AKI. Targeting renal endothelial cell therapy may ameliorate vascular injury and further improve the prognosis of ischemic AKI. Here, P-selectin as a biomarker of ischemic AKI in endothelial cells is identified and P-selectin binding peptide (PBP)-engineered extracellular vesicles (PBP-EVs) with imaging and therapeutic functions are developed. The results show that PBP-EVs exhibit a selective targeting tendency to injured kidneys, while providing spatiotemporal information for the early diagnosis of AKI by quantifying the expression of P-selectin in the kidneys by molecular imaging. Meanwhile, PBP-EVs reveal superior nephroprotective functions in the promotion of renal repair and inhibition of fibrosis by alleviating inflammatory infiltration, improving reparative angiogenesis, and ameliorating maladaptive repair of the renal parenchyma. In conclusion, PBP-EVs, as an ischemic AKI theranostic system that is designed in this study, provide a spatiotemporal diagnosis in the early stages of AKI to help guide personalized therapy and exhibit superior nephroprotective effects, offering proof-of-concept data to design EV-based theranostic strategies to promote renal recovery and further improve long-term outcomes following AKI.


Asunto(s)
Lesión Renal Aguda , Vesículas Extracelulares , Humanos , Células Endoteliales/metabolismo , Selectina-P/metabolismo , Riñón/metabolismo , Isquemia/terapia , Lesión Renal Aguda/metabolismo , Vesículas Extracelulares/metabolismo
17.
Nat Commun ; 14(1): 4373, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37474525

RESUMEN

Mesenchymal stem cells (MSCs) possess potent immunomodulatory activity and have been extensively investigated for their therapeutic potential in treating inflammatory disorders. However, the mechanisms underlying the immunosuppressive function of MSCs are not fully understood, hindering the development of standardized MSC-based therapies for clinical use. In this study, we profile the single-cell transcriptomes of MSCs isolated from adipose tissue (AD), bone marrow (BM), placental chorionic membrane (PM), and umbilical cord (UC). Our results demonstrate that MSCs undergo a progressive aging process and that the cellular senescence state influences their immunosuppressive activity by downregulating PD-L1 expression. Through integrated analysis of single-cell transcriptomic and proteomic data, we identify GATA2 as a regulator of MSC senescence and PD-L1 expression. Overall, our findings highlight the roles of cell aging and PD-L1 expression in modulating the immunosuppressive efficacy of MSCs and implicating perinatal MSC therapy for clinical applications in inflammatory disorders.


Asunto(s)
Antígeno B7-H1 , Células Madre Mesenquimatosas , Humanos , Femenino , Embarazo , Regulación hacia Abajo , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Multiómica , Proteómica , Placenta/metabolismo , Senescencia Celular/genética , Células Madre Mesenquimatosas/metabolismo
18.
Cell Physiol Biochem ; 30(6): 1526-37, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23235695

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSC) play important roles in modulating the activities of T lymphocytes, dendritic cells and natural killer cells. These immunoregulatory properties of MSC suggest their therapeutic potential in autoimmune diseases. However, the effects of MSC on B cells are still poorly understood. The present study was designed to investigate the interaction between MSC and B cells both in vitro and in vivo, and to determine the possible mechanism of action. DESIGN AND METHOD: The effect of human umbilical cord mesenchymal stem cells (UC-MSC) on proliferation and differentiation of B-cells were characterized in vitro, and we also tested the immunoregulatory properties of mouse bone marrow MSC (BM-MSC) on T cell dependent and independent antibody production in vivo in mice. RESULTS: Treatment with human UC-MSC resulted in an increase of proliferation, differentiation of B cells into plasma cells and production of antibodies in vitro. Mouse BM-MSC significantly enhanced T cell dependent and independent antibodies production in vivo in mice. PGE2 partially mediated the immunosuppressive activity of human UC-MSC but IL-6 did not regulate this activity. CONCLUSION: MSC promote proliferation and differentiation of B cells in vitro and in vivo partially through PGE2 but not IL-6.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Madre Mesenquimatosas/fisiología , Células Plasmáticas/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/fisiología , Células Cultivadas , Técnicas de Cocultivo , Dinoprostona/metabolismo , Humanos , Inmunoglobulinas/biosíntesis , Factores Inmunológicos/metabolismo , Inmunomodulación , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos BALB C , Sindecano-1/metabolismo
19.
Cell Physiol Biochem ; 29(3-4): 551-60, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22508062

RESUMEN

BACKGROUND/AIMS: Human umbilical cord mesenchymal stem cells (hUC-MSCs) possess immunosuppressive activities but the mechanisms of such activities are not fully understood. Here, we investigated the role of IL-6, one of the characteristic factors of MSCs, in the immunoregulating effect of hUC-MSCs on CD4(+) T lymphocytes. METHODS: The condition media from human peripheral blood mononuclear cells (hPBMCs) or CD14+/- cell were tested if stimulating IL-6 production by hUC-MSCs. The related signaling pathway of IL-6, and the immunosuppressive activity of IL-6 on CD4(+) T lymphocytes were studied. RESULT: IL-6 production was dramatically increased by hUC-MSCs when co-culturing with resting or activated hPBMCs. CD14(+) monocytes-paracrined IL-1ß promoted the secretion of IL-6 by hUC-MSCs via JNK and NF-κB signaling pathway. Blocking of PGE2 synthesis did not affect the secretion of IL-6, anti-IL-6 antibody was not able to reverse hUC-MSCs-mediated inhibition on CD4(+) T lymphocytes. IL-6 did not mediate the suppressive activity of IL-1ß-hUC-MSCs- PGE2 on CD4(+) T cell. CONCLUSION: CD14(+) monocytes-paracrined IL-1ß promotes IL-6 secretion by hUC-MSCs through activating JNK and NF-κB signaling pathway. However, increased IL-6 production does not contribute to immunosuppressive activity of IL-1ß-hUC-MSCs- PGE2 on CD4(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inmunosupresores/farmacología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Leucocitos Mononucleares/inmunología , Células Madre Mesenquimatosas/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Dinoprostona/antagonistas & inhibidores , Dinoprostona/inmunología , Dinoprostona/farmacología , Humanos , Inmunofenotipificación , Inmunosupresores/inmunología , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Interleucina-6/metabolismo , Interleucina-6/farmacología , Receptores de Lipopolisacáridos/inmunología , Sistema de Señalización de MAP Quinasas , Activación de Macrófagos , Nitrobencenos/farmacología , Sulfonamidas/farmacología , Cordón Umbilical/citología , Cordón Umbilical/inmunología
20.
Sci Total Environ ; 833: 155272, 2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-35427618

RESUMEN

As a by-product of wastewater treatment, waste activated sludge (WAS) has complex composition, strong hydrophilic extracellular polymeric substance (EPS), which make it difficult to dewater. In this study, an electro-peroxone oxidation-Fe(III) coagulation (E-peroxone-Fe(III)) sequential conditioning approach was developed to improve WAS dewaterability. At E-peroxone oxidation stage, hydrogen peroxide was generated through 2-electron path on a carbon polytetrafluoroethylene cathode, and reacted with the sparged O3 to produce hydroxyl radicals. At the subsequent coagulation stage, Fe(III) was dosed to coagulate the small WAS fragments and release water from WAS. Along E-peroxone-Fe(III) subsequent conditioning process, the physicochemical properties of WAS, main components, functional groups and evolution of protein secondary structure, and typical amino acids in EPS, as well as the type and semi-quantitative of elements in WAS, were investigated. The results indicated that under the optimal conditions, the reductions of specific resistance to filterability (SRF) and capillary suction time (CST) for WAS equalled 78.18% and 71.06%, respectively, and its bound water content decreased from 8.87 g/g TSS to 7.67 g/g TSS. After E-peroxone oxidation, part of protein and polysaccharide migrated outside from TB-EPS to slime, the ratio of α-helix/(ß-sheet + random coil) declined, even some of organic-N disintegrated to inorganic-N. At Fe(III) coagulation stage, re-coagulation of the dispersed WAS fragments and easy extraction from inner EPS for protein and polysaccharide occurred. Furthermore, the protein secondary structure of ß-sheet increased by 13.48%, the contents of hydrophobic and hydrophilic amino acids also increased. In addition, a strong negative correlation between the hydrophobic amino acid content of Met in slime and CST or SRF (R2CST = -0.999, p < 0.05 or R2SRF = -0.948, p < 0.05) occurred, while a strong positive correlation between the hydrophilic amino acid content of Cys in TB-EPS and CST or SRF (R2CST = 0.992, p < 0.05 or R2SRF = 0.921, p < 0.05) occurred, which could be related to the WAS dewaterability.


Asunto(s)
Matriz Extracelular de Sustancias Poliméricas , Aguas del Alcantarillado , Aminoácidos , Compuestos Férricos , Oxidación-Reducción , Polisacáridos , Proteínas , Aguas del Alcantarillado/química , Eliminación de Residuos Líquidos , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA