Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59.644
Filtrar
1.
Biomaterials ; 312: 122719, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39088912

RESUMEN

Acute myeloid leukemia (AML) is a deadly form of leukemia with ineffective traditional treatment and frequent chemoresistance-associated relapse. Personalized drug screening holds promise in identifying optimal regimen, nevertheless, primary AML cells undergo spontaneous apoptosis during cultures, invalidating the drug screening results. Here, we reconstitute a 3D osteogenic niche (3DON) mimicking that in bone marrow to support primary AML cell survival and phenotype maintenance in cultures. Specifically, 3DON derived from osteogenically differentiated mesenchymal stem cells (MSC) from healthy and AML donors are co-cultured with primary AML cells. The AML cells under the AML_3DON niche showed enhanced viability, reduced apoptosis and maintained CD33+ CD34-phenotype, associating with elevated secretion of anti-apoptotic cytokines in the AML_3DON niche. Moreover, AML cells under the AML_3DON niche exhibited low sensitivity to two FDA-approved chemotherapeutic drugs, further suggesting the physiological resemblance of the AML_3DON niche. Most interestingly, AML cells co-cultured with the healthy_3DON niche are highly sensitive to the same sample drugs. This study demonstrates the differential responses of AML cells towards leukemic and healthy bone marrow niches, suggesting the impact of native cancer cell niche in drug screening, and the potential of re-engineering healthy bone marrow niche in AML patients as chemotherapeutic adjuvants overcoming chemoresistance, respectively.


Asunto(s)
Supervivencia Celular , Leucemia Mieloide Aguda , Células Madre Mesenquimatosas , Fenotipo , Microambiente Tumoral , Humanos , Leucemia Mieloide Aguda/patología , Microambiente Tumoral/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo/métodos , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Médula Ósea/patología , Médula Ósea/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos , Células de la Médula Ósea/citología , Masculino , Diferenciación Celular/efectos de los fármacos , Femenino
2.
Int J Med Sci ; 21(11): 2233-2243, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39239546

RESUMEN

Purpose: Cognitive dysfunction caused by chronic cerebral hypoperfusion (CCH) is the leading cause of vascular dementia. Therefore, it is necessary to explore the mechanism that causes cerebral injury and find an effective therapy. Methods: Bone marrow mononuclear cells (BMMNCs) were extracted to detect the activity by CCK-8 kit and verify the transfection efficiency using reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR). A CCH rat model was established. Superparamagnetic iron oxide nanoparticles (BMPs)-PEI-Slit2/BMMNCs were injected into the tail vein and intervened with an external magnetic field. Hematoxylin and eosin staining was used to observe the pathological changes in brain tissue. The Slit/Robo pathway-related proteins Slit2 and Robo4 were detected by RT-qPCR and Western blotting. Results: The neurological score of the CCH group significantly increased compared with that of the sham group (P<0.05). The levels of brain injury markers S-100ß and NSE were significantly higher in the CCH group than in the sham group (P<0.05). Neuronal apoptosis in the frontal cortex and hippocampus of CCH rats significantly increased compared with that of the sham group (P<0.05). The expression levels of Slit2 and Robo4 mRNAs and proteins in brain tissue of CCH rats significantly increased (P<0.05). The neurological function scores of CCH rats treated with BMP-PEI-Slit2/BMMNC significantly increased after Robo4 siRNA administration (P<0.05). Conclusion: BMP combination with the CCH-related gene Slit2 can effectively improve the efficiency of BMMNC transplantation in treatment.


Asunto(s)
Isquemia Encefálica , Disfunción Cognitiva , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intercelular , Proteínas del Tejido Nervioso , Animales , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratas , Disfunción Cognitiva/terapia , Disfunción Cognitiva/etiología , Isquemia Encefálica/terapia , Isquemia Encefálica/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Humanos , Masculino , Nanopartículas de Magnetita/administración & dosificación , Nanopartículas de Magnetita/química , Nanopartículas Magnéticas de Óxido de Hierro/administración & dosificación , Células de la Médula Ósea , Apoptosis/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Terapia Genética/métodos , Proteínas Roundabout
3.
Stem Cell Res Ther ; 15(1): 301, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39278909

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressive motoneuron degenerative disorder. There are still no drugs capable of slowing disease evolution or improving life quality of ALS patients. Thus, autologous stem cell therapy has emerged as an alternative treatment regime to be investigated in clinical ALS. METHOD: Using Proteomics and Protein-Protein Interaction Network analyses combined with bioinformatics, the possible cellular mechanisms and molecular targets related to mesenchymal stem cells (MSCs, 1 × 106 cells/kg, intrathecally in the lumbar region of the spine) were investigated in cerebrospinal fluid (CSF) of ALS patients who received intrathecal infusions of autologous bone marrow-derived MSCs thirty days after cell therapy. Data are available via ProteomeXchange with identifier PXD053129. RESULTS: Proteomics revealed 220 deregulated proteins in CSF of ALS subjects treated with MSCs compared to CSF collected from the same patients prior to MSCs infusion. Bioinformatics enriched analyses highlighted events of Extracellular matrix and Cell adhesion molecules as well as related key targets APOA1, APOE, APP, C4A, C5, FGA, FGB, FGG and PLG in the CSF of cell treated ALS subjects. CONCLUSIONS: Extracellular matrix and cell adhesion molecules as well as their related highlighted components have emerged as key targets of autologous MSCs in CSF of ALS patients. TRIAL REGISTRATION: Clinicaltrial.gov identifier NCT0291768. Registered 28 September 2016.


Asunto(s)
Esclerosis Amiotrófica Lateral , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Proteómica , Trasplante Autólogo , Humanos , Esclerosis Amiotrófica Lateral/líquido cefalorraquídeo , Esclerosis Amiotrófica Lateral/terapia , Esclerosis Amiotrófica Lateral/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteómica/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Masculino , Femenino , Persona de Mediana Edad , Apolipoproteínas E/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/líquido cefalorraquídeo , Anciano , Apolipoproteína A-I/líquido cefalorraquídeo , Apolipoproteína A-I/metabolismo , Adulto , Células de la Médula Ósea/metabolismo , Mapas de Interacción de Proteínas
4.
Stem Cell Res Ther ; 15(1): 304, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39278922

RESUMEN

BACKGROUND: Although bone marrow-derived cells with high aldehyde dehydrogenase activity (ALDHbr) have shown therapeutic potential against various diseases in animal studies, clinical trials have failed to show concurrent findings. We aimed to clarify the optimal conditions for the efficacy of ALDHbr cells by using a murine bleomycin-induced pulmonary fibrosis model. METHODS: We intravenously transferred male or female donor C57BL/6 mice-derived ALDHbr cells into recipient C57BL/6 mice under various conditions, and used mCherry-expressing mice as a donor to trace the transferred ALDHbr cells. RESULTS: Pulmonary fibrosis improved significantly when (1) female-derived, not male-derived, and (2) lineage (Lin)-negative, not lineage-positive, ALDHbr cells were transferred during the (3) fibrotic, not inflammatory, phase. Consistent with the RNA-sequencing results, female-derived Lin-/ALDHbr cells were more resistant to oxidative stress than male-derived cells in vitro, and transferred female-derived Lin-/ALDHbr cells were more viable than male-derived cells in the fibrotic lung. The mechanism underlying the antifibrotic effects of Lin-/ALDHbr cells was strongly associated with reduction of oxidative stress. CONCLUSIONS: Our results indicated that Lin-/ALDHbr cell therapy could ameliorate pulmonary fibrosis by reducing oxidative stress and suggested that their efficacy was mediated by sex-related differences. Thus, sex-awareness strategies may be important for clinical application of bone marrow ALDHbr cells as a therapeutic tool.


Asunto(s)
Aldehído Deshidrogenasa , Células de la Médula Ósea , Ratones Endogámicos C57BL , Fibrosis Pulmonar , Animales , Ratones , Femenino , Masculino , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/inducido químicamente , Células de la Médula Ósea/citología , Aldehído Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa/genética , Bleomicina , Modelos Animales de Enfermedad , Estrés Oxidativo
5.
J Orthop Surg Res ; 19(1): 572, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39285416

RESUMEN

BACKGROUND: Osteoporosis results from decreased bone mass and disturbed bone structure. Human bone marrow mesenchymal stem cells (hBMSCs) demonstrate robust osteogenic differentiation, a critical process for bone formation. This research was designed to examine the functions of LINC01133 in osteogenic differentiation. METHODS: Differentially expressed lncRNAs affecting osteogenic differentiation in hBMSCs were identified from the GEO database. A total of 74 osteoporosis patients and 70 controls were enrolled. hBMSCs were stimulated to undergo osteogenic differentiation using an osteogenic differentiation medium (OM). RT-qPCR was performed to evaluate LINC01133 levels and osteogenesis-related genes such as osteocalcin, osteopontin, and RUNX2. An alkaline phosphates (ALP) activity assay was conducted to assess osteogenic differentiation. Cell apoptosis was detected using flow cytometry. Dual luciferase reporter assay and RIP assay were employed to investigate the association between miR-214-3p and LINC01133 or CTNNB1. Loss or gain of function assays were conducted to elucidate the impact of LINC01133 and miR-214-3p on osteogenic differentiation of hBMSCs. RESULTS: LINC01133 and CTNNB1 expression decreased in osteoporotic patients but increased in OM-cultured hBMSCs, whereas miR-214-3p showed an opposite trend. Depletion of LINC01133 suppressed the expression of genes associated with bone formation and ALP activity triggered by OM in hBMSCs, leading to increased cell apoptosis. Nevertheless, this suppression was partially counteracted by the reduced miR-214-3p levels. Mechanistically, LINC01133 and CTNNB1 were identified as direct targets of miR-214-3p. CONCLUSIONS: Our study highlights the role of LINC01133 in positively regulating CTNNB1 expression by inhibiting miR-214-3p, thereby promoting osteogenic differentiation of BMSCs. These findings may provide valuable insights into bone regeneration in osteoporosis.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas , MicroARNs , Osteogénesis , Osteoporosis , ARN Largo no Codificante , Regulación hacia Arriba , beta Catenina , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/genética , Osteogénesis/fisiología , Diferenciación Celular/genética , ARN Largo no Codificante/genética , beta Catenina/genética , beta Catenina/metabolismo , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/patología , Células Cultivadas , Femenino , Persona de Mediana Edad , Masculino , Apoptosis/genética , Células de la Médula Ósea/metabolismo
6.
Front Immunol ; 15: 1374838, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39281683

RESUMEN

Dendritic cells (DCs) are professional antigen-presenting cells, which are key components of the immune system and involved in early immune responses. DCs are specialized in capturing, processing, and presenting antigens to facilitate immune interactions. Chickens infected with avian influenza virus (AIV) demonstrate a wide range of clinical symptoms, based on pathogenicity of the virus. Low pathogenic avian influenza (LPAI) viruses typically induce mild clinical signs, whereas high pathogenic avian influenza (HPAI) induce more severe disease, which can lead to death. For this study, chicken bone marrow-derived DC (ckBM-DC)s were produced and infected with high and low pathogenic avian influenza viruses of H5N2 or H7N3 subtypes to characterize innate immune responses, study effect on cell morphologies, and evaluate virus replication. A strong proinflammatory response was observed at 8 hours post infection, via upregulation of chicken interleukin-1ß and stimulation of the interferon response pathway. Microscopically, the DCs underwent morphological changes from classic elongated dendrites to a more general rounded shape that eventually led to cell death with the presence of scattered cellular debris. Differences in onset of morphologic changes were observed between H5 and H7 subtypes. Increases in viral titers demonstrated that both HPAI and LPAI are capable of infecting and replicating in DCs. The increase in activation of infected DCs may be indicative of a dysregulated immune response typically seen with HPAI infections.


Asunto(s)
Pollos , Citocinas , Células Dendríticas , Gripe Aviar , Animales , Células Dendríticas/inmunología , Células Dendríticas/virología , Pollos/virología , Gripe Aviar/inmunología , Gripe Aviar/virología , Gripe Aviar/patología , Citocinas/metabolismo , Citocinas/inmunología , Virus de la Influenza A/inmunología , Replicación Viral , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/virología
7.
FASEB J ; 38(17): e23892, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39230563

RESUMEN

Mesenchymal stromal stem cells (MSCs) or skeletal stem cells (SSCs) play a major role in tissue repair due to their robust ability to differentiate into osteoblasts, chondrocytes, and adipocytes. Complex cell signaling cascades tightly regulate this differentiation. In osteogenic differentiation, Runt-related transcription factor 2 (RUNX2) and ALP activity are essential. Furthermore, during the latter stages of osteogenic differentiation, mineral formation mediated by the osteoblast occurs with the secretion of a collagenous extracellular matrix and calcium deposition. Activation of nuclear factor erythroid 2-related factor 2 (NRF2), an important transcription factor against oxidative stress, inhibits osteogenic differentiation and mineralization via modulation of RUNX2 function; however, the exact role of NRF2 in osteoblastogenesis remains unclear. Here, we demonstrate that NRF2 activation in human bone marrow-derived stromal cells (HBMSCs) suppressed osteogenic differentiation. NRF2 activation increased the expression of STRO-1 and KITLG (stem cell markers), indicating NRF2 protects HBMSCs stemness against osteogenic differentiation. In contrast, NRF2 activation enhanced mineralization, which is typically linked to osteogenic differentiation. We determined that these divergent results were due in part to the modulation of cellular calcium flux genes by NRF2 activation. The current findings demonstrate a dual role for NRF2 as a HBMSC maintenance factor as well as a central factor in mineralization, with implications therein for elucidation of bone formation and cellular Ca2+ kinetics, dystrophic calcification and, potentially, application in the modulation of bone formation.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas , Factor 2 Relacionado con NF-E2 , Osteoblastos , Osteogénesis , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Osteogénesis/fisiología , Diferenciación Celular/fisiología , Osteoblastos/metabolismo , Osteoblastos/citología , Calcificación Fisiológica/fisiología , Células Cultivadas , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética
8.
PLoS One ; 19(9): e0309455, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39231178

RESUMEN

Hemorrhage, a condition that accompanies most physical trauma cases, remains an important field of study, a field that has been extensively studied in the immunological context for myeloid and lymphoid cells, but not as much for erythroid cells. In this study, we studied the immunological response of murine erythroid cells to acute blood loss using flow cytometry, NanoString immune transcriptome profiling, and BioPlex cytokine secretome profiling. We observed that acute blood loss forces the differentiation of murine erythroid cells in both bone marrow and spleen and that there was an up-regulation of several immune response genes, in particular pathogen-associated molecular pattern sensing gene Clec5a in post-acute blood loss murine bone marrow erythroid cells. We believe that the up-regulation of the Clec5a gene in bone marrow erythroid cells could help bone marrow erythroid cells detect and eliminate pathogens with the help of reactive oxygen species and antimicrobial proteins calprotectin and cathelicidin, the genes of which (S100a8, S100a9, and Camp) dominate the expression in bone marrow erythroid cells of mice.


Asunto(s)
Diferenciación Celular , Quimiocina CCL3 , Células Eritroides , Antígenos Comunes de Leucocito , Animales , Ratones , Células Eritroides/metabolismo , Células Eritroides/citología , Quimiocina CCL3/metabolismo , Quimiocina CCL3/genética , Antígenos Comunes de Leucocito/metabolismo , Lectinas Tipo C/metabolismo , Lectinas Tipo C/genética , Ratones Endogámicos C57BL , Calgranulina A/metabolismo , Calgranulina A/genética , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Calgranulina B/metabolismo , Calgranulina B/genética , Masculino
9.
Int J Mol Sci ; 25(17)2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39273235

RESUMEN

Ionizing radiation exposure can cause damage to diverse tissues and organs, with the hematopoietic system being the most sensitive. However, limited information is available regarding the radiosensitivity of various hematopoietic cell populations in the bone marrow due to the high heterogeneity of the hematopoietic system. In this study, we observed that granulocyte-macrophage progenitors, hematopoietic stem/progenitor cells, and B cells within the bone marrow showed the highest sensitivity, exhibiting a rapid decrease in cell numbers following irradiation. Nonetheless, neutrophils, natural killer (NK) cells, T cells, and dendritic cells demonstrated a certain degree of radioresistance, with neutrophils exhibiting the most pronounced resistance. By employing single-cell transcriptome sequencing, we investigated the early responsive genes in various cell types following irradiation, revealing that distinct gene expression profiles emerged between radiosensitive and radioresistant cells. In B cells, radiation exposure led to a specific upregulation of genes associated with mitochondrial respiratory chain complexes, suggesting a connection between these complexes and cell radiosensitivity. In neutrophils, radiation exposure resulted in fewer gene alterations, indicating their potential for distinct mechanisms in radiation resistance. Collectively, this study provides insights into the molecular mechanism for the heterogeneity of radiosensitivity among the various bone marrow hematopoietic cell populations.


Asunto(s)
Radiación Ionizante , Análisis de la Célula Individual , Transcriptoma , Animales , Ratones , Análisis de la Célula Individual/métodos , Transcriptoma/efectos de la radiación , Células de la Médula Ósea/efectos de la radiación , Células de la Médula Ósea/metabolismo , Ratones Endogámicos C57BL , Tolerancia a Radiación/genética , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/efectos de la radiación , Células Madre Hematopoyéticas/metabolismo , Neutrófilos/efectos de la radiación , Neutrófilos/metabolismo
10.
Chin J Dent Res ; 27(3): 215-224, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39221982

RESUMEN

OBJECTIVE: To investigate whether bone marrow mesenchymal stem cells (BMMSCs) modulate periodontal bone repair through the hydroxylase domain-containing protein 2 (PHD2)/hypoxia- inducible factor-1 (HIF-1) signalling pathway in response to inflammatory conditions. METHODS: Osteogenic differentiation of PHD2 shRNA-modified BMMSCs and the possible mechanism were explored in an inflammatory microenvironment stimulated by porphyromonas gingivalis lipopolysaccharide (Pg-LPS) in vitro. The effect of PHD2 gene-modified BMMSCs on periodontal bone loss was evaluated with experimental periodontitis. RESULTS: Pg-LPS stimulation greatly impaired the osteogenic differentiation of BMMSCs, whereas the silence of PHD2 significantly enhanced the osteogenesis of BMMSCs. More importantly, increased level of vascular endothelial growth factor (VEGF) was detected under Pg-LPS stimulation, which was verified to be associated with the augmented osteogenesis. In experimental periodontitis, PHD2-modified BMMSCs transplantation elevated osteogenic parameters and the expression of VEGF in periodontal tissue. CONCLUSION: This study highlighted that PHD2 gene silencing could be a feasible approach to combat inflammatory bone loss by rescuing the dysfunction of seed cells.


Asunto(s)
Prolina Dioxigenasas del Factor Inducible por Hipoxia , Células Madre Mesenquimatosas , Osteogénesis , ARN Interferente Pequeño , Animales , ARN Interferente Pequeño/genética , Osteogénesis/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Porphyromonas gingivalis , Periodontitis/terapia , Periodontitis/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Diferenciación Celular , Lipopolisacáridos , Pérdida de Hueso Alveolar , Ratones , Masculino , Células de la Médula Ósea , Regeneración Ósea/genética
11.
Chin J Dent Res ; 27(3): 225-234, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39221983

RESUMEN

OBJECTIVE: To reveal the role and mechanism of cannabinoid receptor 1 (CB1) and mitochondria in promoting osteogenic differentiation of periodontal ligament stem cells (PDLSCs) in the inflammatory microenvironment. METHODS: Bidirectional mitochondrial transfer was performed in bone mesenchymal stem cells (BMSCs) and PDLSCs. Laser confocal microscopy and quantitative flow cytometry were used to observe the mitochondrial transfer and quantitative mitochondrial transfer efficiency. Realtime reverse transcription polymerase chain reaction (RT-PCR) was employed to detect gene expression. Alkaline phosphatase (ALP) activity, alizarin red staining (ARS) and quantitative calcium ion analysis were used to evaluate the degree of osteogenic differentiation of PDLSCs. RESULTS: Bidirectional mitochondrial transfer was observed between BMSCs and PDLSCs. The indirect co-culture system could simulate intercellular mitochondrial transfer. Compared with the conditioned medium (CM) for BMSCs, that for HA-CB1 BMSCs could significantly enhance the mineralisation ability of PDLSCs. The mineralisation ability of PDLSCs could not be enhanced after removing the mitochondria in CM for HA-CB1 BMSCs. The expression level of HO-1, PGC-1α, NRF-1, ND1 and HK2 was significantly increased in HA-CB1 BMSCs. CONCLUSION: CM for HA-CB1 BMSCs could significantly enhance the damaged osteogenic differentiation ability of PDLSCs in the inflammatory microenvironment, and the mitochondria of CM played an important role. CB1 was related to the activation of the HO-1/PGC-1α/NRF-1 mitochondrial biogenesis pathway, and significantly increased the mitochondrial content in BMSCs.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas , Mitocondrias , Osteogénesis , Ligamento Periodontal , Receptor Cannabinoide CB1 , Adolescente , Humanos , Células de la Médula Ósea , Células Cultivadas , Técnicas de Cocultivo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Mitocondrias/metabolismo , Osteogénesis/fisiología , Ligamento Periodontal/citología , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB1/genética
12.
BMC Res Notes ; 17(1): 253, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39252057

RESUMEN

OBJECTIVES: Current data suggests that Bacille Calmette-Guerin (BCG) vaccination contributes to nonspecific enhancement of resistance to various infections. Thus, BCG vaccination induces both specific immunity against mycobacteria and non-specific "trained immunity" against various pathogens. To understand the fundamental mechanisms of "trained" immunity, studies of transcriptome changes occurring during BCG vaccination in innate immunity cells, as well as in their precursors, are necessary. Furthermore, this data possesses important significance for practical applications associated with the development of recombinant BCG strains aimed to enhance innate immunity against diverse infectious agents. DATA DESCRIPTION: We performed RNA sequencing of innate immune cells derived from murine bone marrow and spleen three days after subcutaneous BCG vaccination. Using fluorescence-activated cell sorting we obtained three cell populations for each mouse from both control and BCG vaccinated groups: bone marrow monocytes and neutrophils and splenic NK-cells. Then double-indexed cDNA libraries for Illumina sequencing from the collected samples were prepared, the resulting cDNA library mix was subjected to NovaSeq 6000 sequencing. This paper describes the collection of 24 RNA sequencing samples comprising 4 sets of immune cell populations obtained from subcutaneously BCG-vaccinated and control mice.


Asunto(s)
Vacuna BCG , Inmunidad Innata , Bazo , Transcriptoma , Animales , Vacuna BCG/inmunología , Vacuna BCG/administración & dosificación , Ratones , Transcriptoma/genética , Bazo/inmunología , Vacunación/métodos , Células Asesinas Naturales/inmunología , Ratones Endogámicos C57BL , Inyecciones Subcutáneas , Monocitos/inmunología , Femenino , Neutrófilos/inmunología , Análisis de Secuencia de ARN/métodos , Células de la Médula Ósea/inmunología
13.
J Ovarian Res ; 17(1): 184, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39267091

RESUMEN

Ovarian insufficiency is one of the common reproductive disorders affecting women with limited therapeutic aids. Mesenchymal stem cells have been investigated in such disorders before yet, the exact mechanism of MSCs in ovarian regeneration regarding their epigenetic regulation remains elusive. The current study is to investigate the role of the bone marrow-derived mesenchymal stem cells (BM-MSCs) lncRNA (Neat-1 and Hotair1) and miRNA (mir-21-5p, mir-144-5p, and mir-664-5p) in mitigating ovarian granulosa cell apoptosis as well as searching BM-MSCs in altering the expression of ovarian and hypothalamic IGF-1 - kisspeptin system in connection to HPG axis in a cyclophosphamide-induced ovarian failure rat model. Sixty mature female Sprague Dawley rats were divided into 3 equal groups; control group, premature ovarian insufficiency (POI) group, and POI + BM-MSCs. POI female rat model was established with cyclophosphamide. The result revealed that BM-MSCs and their conditioned media displayed a significant expression level of Neat-1, Hotair-1, mir-21-5p, mir-144-5p, and mir-664-5p. Moreover, BM-MSCs transplantation in POI rats improves; the ovarian and hypothalamic IGF-1 - kisspeptin, HPG axis, ovarian granulosa cell apoptosis, steroidogenesis, angiogenesis, energy balance, and oxidative stress. BM-MSCs expressed higher levels of antiapoptotic lncRNAs and microRNAs that mitigate ovarian insufficiency.


Asunto(s)
Apoptosis , Ciclofosfamida , Factor I del Crecimiento Similar a la Insulina , Células Madre Mesenquimatosas , MicroARNs , Insuficiencia Ovárica Primaria , ARN Largo no Codificante , Ratas Sprague-Dawley , Animales , Femenino , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ciclofosfamida/efectos adversos , Ratas , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Insuficiencia Ovárica Primaria/metabolismo , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/inducido químicamente , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Ovario/metabolismo , Células de la Médula Ósea/metabolismo , Angiogénesis
14.
Int J Mol Sci ; 25(17)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39273096

RESUMEN

In recent years, with the advent of a super-aged society, lifelong dental care has gained increasing emphasis, and implant therapy for patients with an edentulous jaw has become a significant option. However, for implant therapy to be suitable for elderly patients with reduced regenerative and immunological capabilities, higher osteoconductive and antimicrobial properties are required on the implant surfaces. Silicon nitride, a non-oxide ceramic known for its excellent mechanical properties and biocompatibility, has demonstrated high potential for inducing hard tissue differentiation and exhibiting antibacterial properties. In this study, silicon nitride was deposited on pure titanium metal surfaces and evaluated for its biocompatibility and antibacterial properties. The findings indicate that silicon nitride improves the hydrophilicity of the material surface, enhancing the initial adhesion of rat bone marrow cells and promoting hard tissue differentiation. Additionally, the antibacterial properties were assessed using Staphylococcus aureus, revealing that the silicon nitride-coated surfaces exhibited significant antibacterial activity. Importantly, no cytotoxicity was observed, suggesting that silicon nitride-coated titanium could serve as a novel implant material.


Asunto(s)
Antibacterianos , Materiales Biocompatibles Revestidos , Compuestos de Silicona , Staphylococcus aureus , Propiedades de Superficie , Titanio , Titanio/química , Titanio/farmacología , Animales , Antibacterianos/farmacología , Antibacterianos/química , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Ratas , Staphylococcus aureus/efectos de los fármacos , Compuestos de Silicona/química , Compuestos de Silicona/farmacología , Ensayo de Materiales , Adhesión Celular/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos
15.
Zhonghua Xue Ye Xue Za Zhi ; 45(7): 651-659, 2024 Jul 14.
Artículo en Chino | MEDLINE | ID: mdl-39231769

RESUMEN

Objective: To analyze the clinical characteristics and prognosis of patients with myelodysplastic syndrome (MDS) with a bone marrow nucleated erythroid cell proportion of greater than or equal to 50% (MDS-E) . Methods: The clinical characteristics and prognostic factors of patients with MDS-E were retrospectively analyzed by collecting the case data of 1 436 newly treated patients with MDS diagnosed in the Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences from May 2014 to June 2023. Results: A total of 1 436 newly diagnosed patients with complete data were included in the study, of which 337 (23.5%) patients with MDS-E had a younger age of onset and lower neutrophil and platelet counts compared with those in patients with an erythroid cell proportion of less than 50% (MDS-NE) (all P<0.05). The proportion of MDS cases with ring sideroblasts (MDS-RS) was higher in the MDS-E group than in the MDS-NE group, and multi-hit TP53 mutations were more enriched in the MDS-E group than in the MDS-NE group (all P<0.05). Among patients with MDS-RS, the frequency of complex karyotypes and the TP53 mutation rate were significantly lower in the MDS-E group than in the MDS-NE group (0 vs 11.9%, P=0.048 and 2.4% vs 15.1%, P=0.053, respectively). Among patients with TP53 mutations, the frequencies of complex karyotypes and multi-hit TP53 mutations were significantly higher in the MDS-E group than in the MDS-NE group (87.5% vs 64.6%, P=0.003 and 84.0% vs 54.2%, P<0.001, respectively). Survival analysis of patients with MDS-RS found that the overall survival (OS) in the MDS-E group was better than that in the MDS-NE group [not reached vs 63 (95% CI 53.3-72.7) months, P=0.029]. Among patients with TP53 mutations and excess blasts, the OS in the MDS-E group was worse than that in the MDS-NE group [6 (95% CI 2.2-9.8) months vs 12 (95% CI 8.9-15.1) months, P=0.022]. Multivariate analysis showed that age of ≥65 years (HR=2.47, 95% CI 1.43-4.26, P=0.001), mean corpuscular volume (MCV) of ≤100 fl (HR=2.62, 95% CI 1.54-4.47, P<0.001), and TP53 mutation (HR=2.31, 95% CI 1.29-4.12, P=0.005) were poor prognostic factors independent of the Revised International Prognostic Scoring System (IPSS-R) prognosis stratification in patients with MDS-E. Conclusion: Among patients with MDS-RS, MDS-E was strongly associated with a lower proportion of complex karyotypes and TP53 mutations, and the OS in the MDS-E group was longer than that in the MDS-NE group. Among patients with TP53 mutations, MDS-E was strongly associated with complex karyotypes and multi-hit TP53 mutations, and among TP53-mutated patients with excess blasts, the OS in the MDS-E group was shorter than that in the MDS-NE group. Age of ≥65 years, MCV of ≤100 fl, and TP53 mutation were independent adverse prognostic factors affecting OS in patients with MDS-E.


Asunto(s)
Mutación , Síndromes Mielodisplásicos , Humanos , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/diagnóstico , Pronóstico , Estudios Retrospectivos , Médula Ósea/patología , Células de la Médula Ósea , Masculino , Femenino , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/genética , Persona de Mediana Edad
16.
Toxicology ; 508: 153932, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39179171

RESUMEN

Hydroquinone (HQ), a metabolite of benzene, is frequently utilized as a surrogate for benzene in in vitro studies and is associated with the development of acute myeloid leukemia (AML). In the hemotoxicity caused by benzene and HQ, cell apoptosis plays a key role. However, the molecular mechanisms underlying HQ are unknown. Studies have indicated that Suv39h1 is involved in regulating cell division and proliferation by regulating histone H3K9me3. Meanwhile, the Wnt/ß-catenin signaling pathway also plays a significant role in cell proliferation and apoptosis. Therefore, this study was aimed at exploring the regulatory role of Suv39h1 and the Wnt/ß-catenin signaling pathway in the effects of HQ on bone marrow mesenchymal stem cells (BMSCs), as well as its influence on cell proliferation and apoptosis. The results demonstrated that HQ elevated the levels of Suv39h1 and H3K9me3 and activated the Wnt/ß-catenin signaling pathway by upregulating ß-catenin, Wnt2b, C-myc, and Cyclin D1 and downregulating Wnt5a, resulting in an increase in cell growth and a decrease in apoptosis. Suv39h1 knockdown inhibited the Wnt/ß-catenin signaling pathway. Meanwhile, inhibition of the Wnt/ß-catenin signaling pathway resulted in the down-regulation of Suv39h1 and H3K9me3 in BMSCs. They both promoted cell proliferation and inhibited apoptosis in the effects of HQ on BMSCs by downregulating the expression of Cyt-C, Bax, Caspase 3, and Caspase 9 and upregulating the expression of Bcl-xl. Therefore, we concluded that Suv39h1 and the Wnt/ß-catenin signaling pathway may mutually regulate each other in the effects of HQ on BMSCs in order to ameliorate the altered function of BMSCs.


Asunto(s)
Apoptosis , Proliferación Celular , Hidroquinonas , Células Madre Mesenquimatosas , Vía de Señalización Wnt , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Apoptosis/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Animales , Hidroquinonas/toxicidad , Células Cultivadas , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , beta Catenina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Masculino
17.
Mol Med Rep ; 30(4)2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-39129299

RESUMEN

Tanshinone IIA (Tan IIA) may have therapeutic effects on avascular necrosis of the femoral head (ANFH) by targeting bone marrow mesenchymal stem cells (BMSCs). The effect and underlying mechanism of Tan IIA on adipogenesis and osteogenesis ability of BMSCs remain to be elucidated. In the present study BMSCs were treated with osteogenic or adipogenic differentiation medium with or without Tan IIA under hypoxic environment. Osteogenic differentiation potential was evaluated by alkaline phosphatase (ALP) measurement, alizarin red staining and reverse transcription­quantitative (RT­q) PCR of osteogenic marker genes. Adipogenic differentiation potential was evaluated with oil red staining and RT­qPCR of adipogenic marker genes. Detailed mechanism was explored by RNA­seq and small molecular treatment during osteogenesis and adipogenesis of BMSCs. ALP level, mineralized nodules and expression level of osteogenic marker genes significantly increased following Tan IIA treatment during osteogenic differentiation of BMSCs. Lipid droplet and expression levels of adipogenic marker genes significantly decreased following Tan IIA treatment during adipogenic differentiation of BMSCs. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses of RNA­seq data indicated increased Akt and TGFß signaling following Tan IIA treatment. Further western blot assay confirmed that Tan IIA significantly activated Akt/cAMP response element­binding protein signaling and TGFß/Smad3 signaling. Application of Akti1/2 (an Akt inhibitor) significantly decreased the promotion effect of osteogenesis induced by Tan IIA, while the addition of SB431542 significantly reduced inhibition effect of adipogenesis caused by Tan IIA. Tan IIA could promote osteogenic differentiation potential of BMSCs by activating AKT signaling and suppress adipogenic differentiation potential of BMSCs by activating TGFß signaling.


Asunto(s)
Abietanos , Adipogénesis , Diferenciación Celular , Células Madre Mesenquimatosas , Osteogénesis , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Osteogénesis/efectos de los fármacos , Abietanos/farmacología , Adipogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Cultivadas , Proteína smad3/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/citología
18.
J Bone Miner Res ; 39(9): 1356-1370, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39126376

RESUMEN

The skeleton is a metabolically active organ undergoing continuous remodeling initiated by bone marrow stem cells (BMSCs). Recent research has demonstrated that BMSCs adapt the metabolic pathways to drive the osteogenic differentiation and bone formation, but the mechanism involved remains largely elusive. Here, using a comprehensive targeted metabolome and transcriptome profiling, we revealed that one-carbon metabolism was promoted following osteogenic induction of BMSCs. Methotrexate (MTX), an inhibitor of one-carbon metabolism that blocks S-adenosylmethionine (SAM) generation, led to decreased N6-methyladenosine (m6A) methylation level and inhibited osteogenic capacity. Increasing intracellular SAM generation through betaine addition rescued the suppressed m6A content and osteogenesis in MTX-treated cells. Using S-adenosylhomocysteine (SAH) to inhibit the m6A level, the osteogenic activity of BMSCs was consequently impeded. We also demonstrated that the pro-osteogenic effect of m6A methylation mediated by one-carbon metabolism could be attributed to HIF-1α and glycolysis pathway. This was supported by the findings that dimethyloxalyl glycine rescued the osteogenic potential in MTX-treated and SAH-treated cells by upregulating HIF-1α and key glycolytic enzymes expression. Importantly, betaine supplementation attenuated MTX-induced m6A methylation decrease and bone loss via promoting the abundance of SAM in rat. Collectively, these results revealed that one-carbon metabolite SAM was a potential promoter in BMSC osteogenesis via the augmentation of m6A methylation, and the cross talk between metabolic reprogramming, epigenetic modification, and transcriptional regulation of BMSCs might provide strategies for bone regeneration.


The bone is a self-renewing tissue that continues to reshape throughout life. Bone marrow mesenchymal stem cells (BMSCs) are essential for bone homeostasis as they are capable of osteogenic differentiation. Recent evidence suggests that BMSCs drive the osteogenic differentiation through metabolic reprogramming, but the mechanism remains unclear. In this paper, we explored the metabolic alteration following osteogenic induction of BMSCs and found that one-carbon metabolism was obviously promoted in this process. The underlining mechanisms of the osteogenic potential driven by one-carbon metabolism seem to be its contribution on N6-methyladenosine (m6A) methylation and consequent glycolysis level by providing methyl donor. We demonstrated that one-carbon metabolism-mediated m6A methylation was a potential promoter in BMSC osteogenesis, and metabolic-epigenetic coupling might provide novel therapeutic targets for bone regeneration.


Asunto(s)
Adenosina , Carbono , Osteogénesis , Ratas Sprague-Dawley , S-Adenosilmetionina , Animales , S-Adenosilmetionina/metabolismo , S-Adenosilmetionina/farmacología , Osteogénesis/efectos de los fármacos , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosina/metabolismo , Metilación/efectos de los fármacos , Carbono/metabolismo , Carbono/farmacología , Ratas , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Metotrexato/farmacología , Glucólisis/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos
19.
Int J Mol Sci ; 25(16)2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39201546

RESUMEN

Philadelphia-Negative Myeloproliferative neoplasms (MPNs) are a diverse group of blood cancers leading to excessive production of mature blood cells. These chronic diseases, including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), can significantly impact patient quality of life and are still incurable in the vast majority of the cases. This review examines the mechanobiology within a bone marrow niche, emphasizing the role of mechanical cues and the primary cilium in the pathophysiology of MPNs. It discusses the influence of extracellular matrix components, cell-cell and cell-matrix interactions, and mechanosensitive structures on hematopoietic stem cell (HSC) behavior and disease progression. Additionally, the potential implications of the primary cilium as a chemo- and mechanosensory organelle in bone marrow cells are explored, highlighting its involvement in signaling pathways crucial for hematopoietic regulation. This review proposes future research directions to better understand the dysregulated bone marrow niche in MPNs and to identify novel therapeutic targets.


Asunto(s)
Cilios , Trastornos Mieloproliferativos , Humanos , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Trastornos Mieloproliferativos/fisiopatología , Cilios/metabolismo , Cilios/patología , Animales , Médula Ósea/patología , Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Mecanotransducción Celular , Matriz Extracelular/metabolismo , Transducción de Señal , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología
20.
Front Immunol ; 15: 1439510, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39188716

RESUMEN

Background and aim: Bone marrow stem cells (BM-SCs) and their progeny play a central role in tissue repair and regeneration. In patients with chronic liver failure, bone marrow (BM) reserve is severally compromised and they showed marked defects in the resolution of injury and infection, leading to liver failure and the onset of decompensation. Whether BM failure is the cause or consequence of liver failure during cirrhosis is not known. In this study, we aimed to determine the underlying relationship between BM failure and regeneration failure in cirrhosis. Methodology: C57Bl/6(J) mice were used to develop chronic liver injury through intra-peritoneal administration of carbon tetrachloride (CCl4) for 15 weeks (0.1-0.5 ml/kg). Animals were sacrificed to study the transition of cirrhosis and BM defects. To restore the BM-SC reserve; healthy BM cells were infused via intra-BM infusion and assessed for changes in liver injury, regeneration, and BM-SC reserve. Results: Using a CCl4-induced animal - model of cirrhosis, we showed the loss of BM-SCs reserve occurred before regeneration failure and the onset of non-acute decompensation. Intra-BM infusion of healthy BM cells induced the repopulation of native hematopoietic stem cells (HSCs) in cirrhotic BM. Restoring BM-HSCs reserve augments liver macrophage-mediated clearance of infection and inflammation dampens neutrophil-mediated inflammation, accelerates fibrosis regression, enhances hepatocyte proliferation, and delays the onset of non-acute decompensation. Conclusion: These findings suggest that loss of BM-HSCs reserve underlies the compromised innate immune function of the liver, drives regeneration failure, and the onset of non-acute decompensation. We further provide the proof-of-concept that rejuvenating BM-HSC reserve can serve as a potential therapeutic approach for preventing regeneration failure and transition to decompensated cirrhosis.


Asunto(s)
Tetracloruro de Carbono , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas , Cirrosis Hepática , Regeneración Hepática , Ratones Endogámicos C57BL , Animales , Ratones , Cirrosis Hepática/terapia , Cirrosis Hepática/inmunología , Cirrosis Hepática/patología , Masculino , Hígado/patología , Trasplante de Médula Ósea , Células de la Médula Ósea
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA