Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.523
Filtrar
1.
J Control Release ; 371: 386-405, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38844177

RESUMO

Recently, the formation of three-dimensional (3D) cell aggregates known as embryoid bodies (EBs) grown in media supplemented with HSC-specific morphogens has been utilized for the directed differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), into clinically relevant hematopoietic stem cells (HSCs). However, delivering growth factors and nutrients have become ineffective in inducing synchronous differentiation of cells due to their 3D conformation. Moreover, irregularly sized EBs often lead to the formation of necrotic cores in larger EBs, impairing differentiation. Here, we developed two gelatin microparticles (GelMPs) with different release patterns and two HSC-related growth factors conjugated to them. Slow and fast releasing GelMPs were conjugated with bone morphogenic factor-4 (BMP-4) and stem cell factor (SCF), respectively. The sequential presentation of BMP-4 and SCF in GelMPs resulted in efficient and effective hematopoietic differentiation, shown by the enhanced gene and protein expression of several mesoderm and HSC-related markers, and the increased concentration of released HSC-related cytokines. In the present study, we were able to generate CD34+, CD133+, and FLT3+ cells with similar cellular and molecular morphology as the naïve HSCs that can produce colony units of different blood cells, in vitro.


Assuntos
Proteína Morfogenética Óssea 4 , Diferenciação Celular , Gelatina , Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes Induzidas , Esferoides Celulares , Fator de Células-Tronco , Proteína Morfogenética Óssea 4/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator de Células-Tronco/metabolismo , Gelatina/química , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Animais , Humanos , Camundongos
2.
Exp Dermatol ; 33(5): e15091, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38711220

RESUMO

KIT ligand and its associated receptor KIT serve as a master regulatory system for both melanocytes and mast cells controlling survival, migration, proliferation and activation. Blockade of this pathway results in cell depletion, while overactivation leads to mastocytosis or melanoma. Expression defects are associated with pigmentary and mast cell disorders. KIT ligand regulation is complex but efficient targeting of this system would be of significant benefit to those suffering from melanocytic or mast cell disorders. Herein, we review the known associations of this pathway with cutaneous diseases and the regulators of this system both in skin and in the more well-studied germ cell system. Exogenous agents modulating this pathway will also be presented. Ultimately, we will review potential therapeutic opportunities to help our patients with melanocytic and mast cell disease processes potentially including vitiligo, hair greying, melasma, urticaria, mastocytosis and melanoma.


Assuntos
Mastócitos , Mastocitose , Melanócitos , Proteínas Proto-Oncogênicas c-kit , Fator de Células-Tronco , Humanos , Fator de Células-Tronco/metabolismo , Melanócitos/metabolismo , Mastócitos/metabolismo , Mastocitose/tratamento farmacológico , Mastocitose/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Melanoma/metabolismo , Melanoma/tratamento farmacológico , Vitiligo/metabolismo , Vitiligo/tratamento farmacológico , Vitiligo/terapia , Transtornos da Pigmentação/tratamento farmacológico , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Animais
3.
Ecotoxicol Environ Saf ; 279: 116504, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38795418

RESUMO

Cranial radiotherapy is a major treatment for leukemia and brain tumors. Our previous study found abscopal effects of cranial irradiation could cause spermatogenesis disorder in mice. However, the exact mechanisms are not yet fully understood. In the study, adult male C57BL/6 mice were administrated with 20 Gy X-ray cranial irradiation (5 Gy per day for 4 days consecutively) and sacrificed at 1, 2 and 4 weeks. Tandem Mass Tag (TMT) quantitative proteomics of testis was combined with bioinformatics analysis to identify key molecules and signal pathways related to spermatogenesis at 4 weeks after cranial irradiation. GO analysis showed that spermatogenesis was closely related to oxidative stress and inflammation. Severe oxidative stress occurred in testis, serum and brain, while serious inflammation also occurred in testis and serum. Additionally, the sex hormones related to hypothalamic-pituitary-gonadal (HPG) axis were disrupted. PI3K/Akt pathway was activated in testis, which upstream molecule SCF/C-Kit was significantly elevated. Furthermore, the proliferation and differentiation ability of spermatogonial stem cells (SSCs) were altered. These findings suggest that cranial irradiation can cause spermatogenesis disorder through brain-blood-testicular cascade oxidative stress, inflammation and the secretory dysfunction of HPG axis, and SCF/C-kit drive this process through activating PI3K/Akt pathway.


Assuntos
Irradiação Craniana , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-kit , Espermatogênese , Animais , Masculino , Espermatogênese/efeitos da radiação , Camundongos , Proteínas Proto-Oncogênicas c-kit/metabolismo , Estresse Oxidativo/efeitos da radiação , Irradiação Craniana/efeitos adversos , Testículo/efeitos da radiação , Testículo/patologia , Transdução de Sinais/efeitos da radiação , Fator de Células-Tronco/metabolismo , Inflamação
4.
Sci Rep ; 14(1): 11689, 2024 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-38778076

RESUMO

We evaluated whether serum stem cell factor (s-SCF) levels just prior to ovulation induction could indicate the ability to develop a top-quality (TQ) blastocyst by day 5. We investigated patients with normal ovarian reserve (NOR), polycystic ovary syndrome (PCOS), diminished ovarian reserve (DOR), or mild endometriosis. Our pilot research suggests a correlation between s-SCF levels and the ability to form TQ blastocysts in patients with mild endometriosis. This significant statistical difference (p < 0.05) was noted between mild endometriosis patients for whom a TQ blastocyst was obtained and those for whom it was not possible, as measured on the 8th day of stimulation and the day of oocyte retrieval. The mean SCF levels in the serum of these women on the 8th day were at 28.07 (± 2.67) pg/ml for the TQ subgroup and 53.32 (± 16.02) pg/ml for the non-TQ subgroup (p < 0.05). On oocyte retrieval day it was 33.47 (± 3.93) pg/ml and 52.23 (± 9.72) pg/ml (p < 0.05), respectively.


Assuntos
Blastocisto , Reserva Ovariana , Fator de Células-Tronco , Humanos , Feminino , Fator de Células-Tronco/sangue , Adulto , Blastocisto/citologia , Reserva Ovariana/fisiologia , Síndrome do Ovário Policístico/sangue , Endometriose/sangue , Recuperação de Oócitos , Indução da Ovulação/métodos , Projetos Piloto , Fertilização in vitro/métodos
5.
Pigment Cell Melanoma Res ; 37(4): 514-529, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38705722

RESUMO

Epidermal melanin unit integrity is crucial for skin homeostasis and pigmentation. Epidermal growth factor (EGF) receptor (EGFR) is a pivotal player in cell growth, wound healing, and maintaining skin homeostasis. However, its influence on skin pigmentation is relatively unexplored. This study investigates the impact and underlying mechanisms of EGFR inhibitors on skin pigmentation. We evaluated EGF and EGFR expression in various skin cells using quantitative real-time PCR, Western blot, and immunofluorescence. EGF and EGFR were predominantly expressed in epidermal keratinocytes, and treatment with the EGFR tyrosine kinase inhibitors (EGFR-TKIs) gefitinib and PD153035 significantly increased stem cell factor (SCF) and endothelin-1 (ET-1) expression in cultured keratinocytes. Enhanced melanocyte migration and proliferation were observed in co-culture, as evidenced by time-lapse live imaging and single-cell tracking assays. Furthermore, topical application of gefitinib to guinea pig dorsal skin induced increased pigmentation and demonstrated efficacy in mitigating rhododendrol-induced leukoderma. Suppression of EGF signaling indirectly enhanced skin pigmentation by upregulating SCF and ET-1 in epidermal keratinocytes. This novel mechanism highlights the pivotal role of EGF signaling in regulating skin pigmentation, and topical EGFR-TKI therapy at an appropriate dose may be a promising approach for depigmentation disorder management.


Assuntos
Receptores ErbB , Gefitinibe , Hipopigmentação , Queratinócitos , Melaninas , Melanócitos , Inibidores de Proteínas Quinases , Receptores ErbB/metabolismo , Animais , Melaninas/metabolismo , Melaninas/biossíntese , Humanos , Inibidores de Proteínas Quinases/farmacologia , Melanócitos/efeitos dos fármacos , Melanócitos/metabolismo , Melanócitos/patologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/patologia , Hipopigmentação/patologia , Hipopigmentação/tratamento farmacológico , Gefitinibe/farmacologia , Cobaias , Pigmentação da Pele/efeitos dos fármacos , Fator de Células-Tronco/metabolismo , Epiderme/efeitos dos fármacos , Epiderme/patologia , Epiderme/metabolismo , Proliferação de Células/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Endotelina-1/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Quinazolinas
6.
BMC Nephrol ; 25(1): 161, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38730296

RESUMO

BACKGROUND: Previous observational studies have reported that systemic inflammatory regulators are related to the development of chronic kidney disease (CKD); however, whether these associations are causal remains unclear. The current study aimed to investigate the potential causal relationships between systemic inflammatory regulators and CKD and kidney function. METHOD: We performed bidirectional two-sample Mendelian randomization (MR) analyses to infer the underlying causal associations between 41 systemic inflammatory regulators and CKD and kidney function. The inverse-variance weighting (IVW) test was used as the primary analysis method. In addition, sensitivity analyses were executed via the Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) test and the weighted median test. RESULTS: The findings revealed 12 suggestive associations between 11 genetically predicted systemic inflammatory regulators and CKD or kidney function in the forward analyses, including 4 for CKD, 3 for blood urea nitrogen (BUN), 4 for eGFRcrea and 1 for eGFRcys. In the other direction, we identified 6 significant causal associations, including CKD with granulocyte-colony stimulating factor (GCSF) (IVW ß = 0.145; 95% CI, 0.042 to 0.248; P = 0.006), CKD with stem cell factor (SCF) (IVW ß = 0.228; 95% CI, 0.133 to 0.323; P = 2.40 × 10- 6), eGFRcrea with SCF (IVW ß =-2.90; 95% CI, -3.934 to -1.867; P = 3.76 × 10- 8), eGFRcys with GCSF (IVW ß =-1.382; 95% CI, -2.404 to -0.361; P = 0.008), eGFRcys with interferon gamma (IFNg) (IVW ß =-1.339; 95% CI, -2.313 to -0.366; P = 0.007) and eGFRcys with vascular endothelial growth factor (VEGF) (IVW ß =-1.709; 95% CI, -2.720 to -0.699; P = 9.13 × 10- 4). CONCLUSIONS: Our findings support causal links between systemic inflammatory regulators and CKD or kidney function both in the forward and reverse MR analyses.


Assuntos
Análise da Randomização Mendeliana , Insuficiência Renal Crônica , Humanos , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/sangue , Taxa de Filtração Glomerular , Inflamação/genética , Fator Estimulador de Colônias de Granulócitos/sangue , Fator de Células-Tronco/genética , Fator de Células-Tronco/sangue , Rim/metabolismo , Rim/fisiopatologia , Nitrogênio da Ureia Sanguínea
8.
Int J Mol Sci ; 25(7)2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38612465

RESUMO

Ulcerative colitis (UC) is a relapsing and reoccurring inflammatory bowel disease. The treatment effect of Alhagi maurorum and stem cell extracts on UC remains unclear. The aim of the present study was to investigate the protective role of Alhagi maurorum combined with stem cell extract on the intestinal mucosal barrier in an intestinal inflammation mouse model. Sixty mice were randomly divided into a control group, model group, Alhagi group, MSC group, and MSC/Alhagi group. MSC and Alhagi extract were found to reduce the disease activity index (DAI) scores in mice with colitis, alleviate weight loss, improve intestinal inflammation in mice (p < 0.05), preserve the integrity of the ileal wall and increase the number of goblet cells and mucin in colon tissues. Little inflammatory cell infiltration was observed in the Alhagi, MSC, or MSC/Alhagi groups, and the degree of inflammation was significantly alleviated compared with that in the model group. The distribution of PCNA and TNF-alpha in the colonic tissues of the model group was more disperse than that in the normal group (p < 0.05), and the fluorescence intensity was lower. After MSC/Alhagi intervention, PCNA and TNF-alpha were distributed along the cellular membrane in the MSC/Alhagi group (p < 0.05). Compared with that in the normal control group, the intensity was slightly reduced, but it was still stronger than that in the model group. In conclusion, MSC/Alhagi can alleviate inflammatory reactions in mouse colonic tissue, possibly by strengthening the protective effect of the intestinal mucosal barrier.


Assuntos
Colite Ulcerativa , Fabaceae , Células-Tronco Mesenquimais , Animais , Camundongos , Colite Ulcerativa/tratamento farmacológico , Fator de Células-Tronco , Antígeno Nuclear de Célula em Proliferação , Fator de Necrose Tumoral alfa , Inflamação , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico
9.
Elife ; 122024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38536959

RESUMO

The cell-type-specific expression of ligand/receptor and cell-adhesion molecules is a fundamental mechanism through which neurons regulate connectivity. Here, we determine a functional relevance of the long-established mutually exclusive expression of the receptor tyrosine kinase Kit and the trans-membrane protein Kit Ligand by discrete populations of neurons in the mammalian brain. Kit is enriched in molecular layer interneurons (MLIs) of the cerebellar cortex (i.e., stellate and basket cells), while cerebellar Kit Ligand is selectively expressed by a target of their inhibition, Purkinje cells (PCs). By in vivo genetic manipulation spanning embryonic development through adulthood, we demonstrate that PC Kit Ligand and MLI Kit are required for, and capable of driving changes in, the inhibition of PCs. Collectively, these works in mice demonstrate that the Kit Ligand/Kit receptor dyad sustains mammalian central synapse function and suggest a rationale for the affiliation of Kit mutation with neurodevelopmental disorders.


Assuntos
Células de Purkinje , Fator de Células-Tronco , Camundongos , Animais , Células de Purkinje/fisiologia , Fator de Células-Tronco/metabolismo , Cerebelo/fisiologia , Córtex Cerebelar/metabolismo , Interneurônios/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Mamíferos/metabolismo
10.
Cancer Lett ; 589: 216795, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38556106

RESUMO

The immune microenvironment constructed by tumor-infiltrating immune cells and the molecular phenotype defined by hormone receptors (HRs) have been implicated as decisive factors in the regulation of breast cancer (BC) progression. Here, we found that the infiltration of mast cells (MCs) informed impaired prognoses in HR(+) BC but predicted improved prognoses in HR(-) BC. However, molecular features of MCs in different BC remain unclear. We next discovered that HR(-) BC cells were prone to apoptosis under the stimulation of MCs, whereas HR(+) BC cells exerted anti-apoptotic effects. Mechanistically, in HR(+) BC, the KIT ligand (KITLG), a major mast cell growth factor in recruiting and activating MCs, could be transcriptionally upregulated by the progesterone receptor (PGR), and elevate the production of MC-derived granulin (GRN). GRN attenuates TNFα-induced apoptosis in BC cells by competitively binding to TNFR1. Furthermore, disruption of PGR-KITLG signaling by knocking down PGR or using the specific KITLG-cKIT inhibitor iSCK03 potently enhanced the sensitivity of HR(+) BC cells to MC-induced apoptosis and exerted anti-tumor activity. Collectively, these results demonstrate that PGR-KITLG signaling in BC cells preferentially induces GRN expression in MCs to exert anti-apoptotic effects, with potential value in developing precision medicine approaches for diagnosis and treatment.


Assuntos
Neoplasias da Mama , Fator de Células-Tronco , Humanos , Feminino , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo , Mastócitos/patologia , Neoplasias da Mama/patologia , Retroalimentação , Apoptose , Microambiente Tumoral
11.
Cardiovasc Res ; 120(7): 745-755, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38507654

RESUMO

AIMS: In hypoxia, endothelial cells (ECs) proliferate, migrate, and form new vasculature in a process called angiogenesis. Recent studies have suggested that ECs rely on glycolysis to meet metabolic needs for angiogenesis in ischaemic tissues, and several studies have investigated the molecular mechanisms integrating angiogenesis and endothelial metabolism. Here, we investigated the role of stem cell factor (SCF) and its receptor, cKIT, in regulating endothelial glycolysis during hypoxia-driven angiogenesis. METHODS AND RESULTS: SCF and cKIT signalling increased the glucose uptake, lactate production, and glycolysis in human ECs under hypoxia. Mechanistically, SCF and cKIT signalling enhanced the expression of genes encoding glucose transporter 1 (GLUT1) and glycolytic enzymes via Akt- and ERK1/2-dependent increased translation of hypoxia inducible factor 1A (HIF1A). In hypoxic conditions, reduction of glycolysis and HIF-1α expression using chemical inhibitors significantly reduced the SCF-induced in vitro angiogenesis in human ECs. Compared with normal mice, mice with oxygen-induced retinopathy (OIR), characterized by ischaemia-driven pathological retinal neovascularization, displayed increased levels of SCF, cKIT, HIF-1α, GLUT1, and glycolytic enzymes in the retina. Moreover, cKIT-positive neovessels in the retina of mice with OIR showed elevated expression of GLUT1 and glycolytic enzymes. Further, blocking SCF and cKIT signalling using anti-SCF neutralizing IgG and cKIT mutant mice significantly reduced the expression of HIF-1α, GLUT1, and glycolytic enzymes and decreased the pathological neovascularization in the retina of mice with OIR. CONCLUSION: We demonstrated that SCF and cKIT signalling regulate angiogenesis by controlling endothelial glycolysis in hypoxia and elucidated the SCF/cKIT/HIF-1α axis as a novel metabolic regulation pathway during hypoxia-driven pathological angiogenesis.


Assuntos
Hipóxia Celular , Transportador de Glucose Tipo 1 , Glicólise , Subunidade alfa do Fator 1 Induzível por Hipóxia , Proteínas Proto-Oncogênicas c-kit , Transdução de Sinais , Fator de Células-Tronco , Animais , Humanos , Fator de Células-Tronco/metabolismo , Fator de Células-Tronco/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 1/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Células Endoteliais da Veia Umbilical Humana/metabolismo , Camundongos Endogâmicos C57BL , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Neovascularização Retiniana/genética , Camundongos , Neovascularização Fisiológica , Células Cultivadas , Modelos Animais de Doenças , Glucose/metabolismo
12.
Eur Rev Med Pharmacol Sci ; 28(5): 1857-1863, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38497868

RESUMO

OBJECTIVE: The clinical frailty scale (CFS) evaluates the level of frailty based on clinical examination, comorbidities, and functional and activity levels of older patients. However, there are many difficulties for internists in evaluating frailty with this scale. Therefore, simplifying the CFS with good design and application is required for better treatment outcomes. Our study was conducted to design and evaluate the correlation of a simplified clinical frailty scale (sCFS) with CFS in older patients. PATIENTS AND METHODS: We undertook a cross-sectional analysis involving 279 older patients, which comprised two steps. Step 1 involves the implementation of sCFS, a protocol that has been endorsed by the Geriatrics Professional Council (GPC). Step 2 entails the enrollment of older patients for frailty assessment using sCFS, comparing it with CFS. RESULTS: The study was conducted on 279 older patients; the average age was 75.7 ± 8.4 (years old), and men accounted for 34.8%. There was a high correlation between the sCFS and CFS (Pearson's r = 0.996; p < 0.001). The similarity of the sCFS to the CFS was very high, with Kappa coefficient = 0.984 (p < 0.001). Compared with the CFS, the sCFS had a Youden index of 98% with 100% sensitivity and 98% specificity assessed through the receiver operating characteristic (ROC) with the CFS threshold of 5. CONCLUSIONS: The sCFS can be used to assess frailty with high sensitivity and specificity.


Assuntos
Fragilidade , Geriatria , Masculino , Humanos , Idoso , Idoso de 80 Anos ou mais , Estudos Transversais , Fragilidade/diagnóstico , Pacientes , Exame Físico , Fator de Células-Tronco
13.
Biomed Pharmacother ; 173: 116318, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38401522

RESUMO

Retinal ischemia is a significant factor in various vision-threatening diseases, but effective treatments are currently lacking. This study explores the potential of stem cell factor (SCF) in regulating the neurovascular unit as a therapeutic intervention for retinal ischemic diseases. A chronic retinal ischemia model was established in Brown Norway rats using bilateral common carotid artery occlusion (BCCAO). Subsequent SCF treatment resulted in a remarkable recovery of retinal function, as indicated by electroretinogram, light/dark transition test, and optokinetic head tracking test results. Histological examination demonstrated a significant increase in the number of retinal neurons and an overall thickening of the retina. Immunofluorescence confirmed these findings and further demonstrated that SCF treatment regulated retinal remodeling. Notably, SCF treatment ameliorated the disrupted expression of synaptic markers in the control group's BCCAO rats and suppressed the activation of Müller cells and microglia. Retinal whole-mount analysis revealed a significant improvement in the abnormalities in retinal vasculature following SCF treatment. Transcriptome sequencing analysis revealed that SCF-induced transcriptome changes were closely linked to the Wnt7 pathway. Key members of the Wnt7 pathway, exhibited significant upregulation following SCF treatment. These results underscore the protective role of SCF in the neurovascular unit of retinal ischemia rats by modulating the Wnt7 pathway. SCF administration emerges as a promising therapeutic strategy for retinal ischemia-related diseases, offering potential avenues for future clinical interventions.


Assuntos
Arteriopatias Oclusivas , Doenças das Artérias Carótidas , Doenças Retinianas , Ratos , Animais , Fator de Células-Tronco , Isquemia/metabolismo , Doenças Retinianas/prevenção & controle , Doenças Retinianas/patologia , Retina , Vasos Retinianos/metabolismo , Arteriopatias Oclusivas/patologia
14.
Mol Cell Endocrinol ; 586: 112163, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38246572

RESUMO

NOTCH2 is expressed in pituitary stem cells and is necessary for stem cell maintenance, proliferation, and differentiation. However, the pathways NOTCH2 engages to affect pituitary development remain unclear. In this study, we hypothesized that glycoprotein hormone subunit A2 (GPHA2), a corneal stem cell factor and ligand for the thyroid stimulating hormone receptor (TSHR), is downstream of NOTCH2 signaling. We found Gpha2 is expressed in quiescent pituitary stem cells by RNAscope in situ hybridization and scRNA seq. In Notch2 conditional knockout pituitaries, Gpha2 mRNA is reduced compared with control littermates. We then investigated the possible functions of GPHA2. Pituitaries treated with a GPHA2 peptide do not have a change in proliferation. However, in dissociated adult pituitary cells, GPHA2 increased pCREB expression and this induction was reversed by co-treatment with a TSHR inhibitor. These data suggest GPHA2 is a NOTCH2 related stem cell factor that activates TSHR signaling, potentially impacting pituitary development.


Assuntos
Hipófise , Fator de Células-Tronco , Adulto , Humanos , Hipófise/metabolismo , Receptor Notch2/genética , Receptor Notch2/metabolismo , Receptores da Tireotropina , Fator de Células-Tronco/metabolismo , Células-Tronco/metabolismo
15.
Cancer Metastasis Rev ; 43(1): 423-440, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37796391

RESUMO

Cellular plasticity can occur naturally in an organism and is considered an adapting mechanism during the developmental stage. However, abnormal cellular plasticity is observed in different diseased conditions, including cancer. Cancer cell plasticity triggers the stimuli of epithelial-mesenchymal transition (EMT), abnormal epigenetic changes, expression of stem cell factors and implicated signaling pathways, etc., and helps in the maintenance of CSC phenotype. Conversely, CSC maintains the cancer cell plasticity, EMT, and epigenetic plasticity. EMT contributes to increased cell migration and greater diversity within tumors, while epigenetic changes, stem cell factors (OCT4, NANOG, and SOX2), and various signaling pathways allow cancer cells to maintain various phenotypes, giving rise to intra- and inter-tumoral heterogeneity. The intricate relationships between cancer cell plasticity and stem cell factors help the tumor cells adopt drug-tolerant states, evade senescence, and successfully acquire drug resistance with treatment dismissal. Inhibiting molecules/signaling pathways involved in promoting CSCs, cellular plasticity, EMT, and epigenetic plasticity might be helpful for successful cancer therapy management. This review discussed the role of cellular plasticity, EMT, and stem cell factors in tumor initiation, progression, reprogramming, and therapy resistance. Finally, we discussed how the intervention in this axis will help better manage cancers and improve patient survivability.


Assuntos
Plasticidade Celular , Neoplasias , Humanos , Fator de Células-Tronco/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Transdução de Sinais/genética , Transição Epitelial-Mesenquimal/genética , Células-Tronco , Células-Tronco Neoplásicas/patologia
16.
Postgrad Med J ; 100(1180): 84-90, 2024 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-37857513

RESUMO

BACKGROUND: It has been reported that patients with cholelithiasis may have changes in levels of cytokines and growth factors, while their causal relationships were still unclear. METHODS: This study was a bidirectional Mendelian randomization (MR) study. Datasets of 41 circulation cytokines and growth factors and the data on cholelithiasis were obtained. Six steps of strict instrumental variable filtration were set, and inverse-variance weighted analysis, MR-Egger regression, and weighted median test were used to identify the causal relationships. Benjamini-Hochberg method was used to adjust the P-values. RESULTS: After adjustments of P-values, four cytokines and growth factors were still causally associated with cholelithiasis significantly: interleukin 2 receptor alpha (adjusted P: 4.59E-02), interleukin 8 (adjusted P: 1.09E-02), monocyte-specific chemokine 3 (adjusted P: 2.73E-04), and stem cell factor (adjusted P: 2.73E-04). In the reverse MR analysis, no significant causal relationship was detected after adjustment. CONCLUSIONS: Four cytokines and growth factors, including interleukin 2 receptor alpha, interleukin 8, monocyte-specific chemokine 3, and stem cell factor, were proven to relate to cholelithiasis causally and unidirectionally.


Assuntos
Citocinas , Interleucina-8 , Humanos , Fator de Células-Tronco , Subunidade alfa de Receptor de Interleucina-2 , Análise da Randomização Mendeliana , Peptídeos e Proteínas de Sinalização Intercelular/genética , Estudo de Associação Genômica Ampla
17.
Cells ; 12(23)2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-38067124

RESUMO

Mast cells (MCs) are sentinel cells which represent an important part of the first line of defense of the immune system. MCs highly express receptors for danger-associated molecular patterns (DAMPs) such as the IL-33R and P2X7, making MCs to potentially effective sensors for IL-33 and adenosine-triphosphate (ATP), two alarmins which are released upon necrosis-induced cell damage in peripheral tissues. Besides receptors for alarmins, MCs also express the stem cell factor (SCF) receptor c-Kit, which typically mediates MC differentiation, proliferation and survival. By using bone marrow-derived MCs (BMMCs), ELISA and flow cytometry experiments, as well as p65/RelA and NFAT reporter MCs, we aimed to investigate the influence of SCF on alarmin-induced signaling pathways and the resulting cytokine production and degranulation. We found that the presence of SCF boosted the cytokine production but not degranulation in MCs which simultaneously sense ATP and IL-33 (ATP/IL-33 co-sensing). Therefore, we conclude that SCF maintains the functionality of MCs in peripheral tissues to ensure appropriate MC reactions upon cell damage, induced by pathogens or allergens.


Assuntos
Citocinas , Mastócitos , Fator de Células-Tronco , Trifosfato de Adenosina/metabolismo , Alarminas/metabolismo , Citocinas/metabolismo , Interleucina-33/metabolismo , Mastócitos/metabolismo , Fator de Células-Tronco/farmacologia , Fator de Células-Tronco/fisiologia , Masculino , Feminino , Animais , Camundongos , Camundongos Endogâmicos C57BL
19.
Front Immunol ; 14: 1294555, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38022523

RESUMO

The application of immunotherapies such as chimeric antigen receptor (CAR) T therapy or bi-specific T cell engager (BiTE) therapy to manage myeloid malignancies has proven more challenging than for B-cell malignancies. This is attributed to a shortage of leukemia-specific cell-surface antigens that distinguish healthy from malignant myeloid populations, and the inability to manage myeloid depletion unlike B-cell aplasia. Therefore, the development of targeted therapeutics for myeloid malignancies, such as acute myeloid leukemia (AML), requires new approaches. Herein, we developed a ligand-based CAR and secreted bi-specific T cell engager (sBite) to target c-kit using its cognate ligand, stem cell factor (SCF). c-kit is highly expressed on AML blasts and correlates with resistance to chemotherapy and poor prognosis, making it an ideal candidate for which to develop targeted therapeutics. We utilize γδ T cells as a cytotoxic alternative to αß T cells and a transient transfection system as both a safety precaution and switch to remove alloreactive modified cells that may hinder successful transplant. Additionally, the use of γδ T cells permits its use as an allogeneic, off-the-shelf therapeutic. To this end, we show mSCF CAR- and hSCF sBite-modified γδ T cells are proficient in killing c-kit+ AML cell lines and sca-1+ murine bone marrow cells in vitro. In vivo, hSCF sBite-modified γδ T cells moderately extend survival of NSG mice engrafted with disseminated AML, but therapeutic efficacy is limited by lack of γδ T-cell homing to murine bone marrow. Together, these data demonstrate preclinical efficacy and support further investigation of SCF-based γδ T-cell therapeutics for the treatment of myeloid malignancies.


Assuntos
Leucemia Mieloide Aguda , Camundongos , Animais , Ligantes , Receptores Proteína Tirosina Quinases , Proteínas Proto-Oncogênicas c-kit/genética , Imunoterapia Adotiva , Fator de Células-Tronco
20.
J Control Release ; 363: 670-681, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37838223

RESUMO

Herein, we present an approach for manipulating paracrine factors and signaling pathways in adipose-derived stem cells (ADSCs) to achieve highly effective tumor immunotherapy. Our method involves precise control of reactive oxygen species concentration using the CD90-maleimide-pluronic F68-chlorin e6 conjugate (CPFC) to create ACPFC, which is then attached to ADSCs through the CD90 receptor-specific interaction. By regulating the irradiated laser power, ACPFC promotes signaling pathways such as cascade-3, VEGFR2, α2ß1, C3AR1, CR1-4, and C5AR1, leading to the secretion of various inflammatory cytokines such as IFN-γ, TGF-ß, and IL-6, while inhibiting AKT, ERK, NFkB, PAR1, and PAR3/4 signaling pathways to reduce the secretion of cell growth factors like TIMP-1, TIMP-2, VEGF, Ang-2, FGF-2, and HGF. When ACPFC is injected intravenously into a tumor animal model, it autonomously targets and accumulates at the tumor site, and upon laser irradiation, it generates various anti-inflammatory factors while reducing angiogenesis growth factors. The resulting antitumor response recruits CD3+CD8+ cytotoxic T cells and CD3+CD4+ helper T cells into the tumor and spleen, leading to highly effective melanoma and pancreatic tumor treatment in mice. Our technology for regulating stem cell paracrine factors holds significant promise for the treatment of various diseases.


Assuntos
Melanoma , Fator de Células-Tronco , Camundongos , Animais , Fator de Células-Tronco/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Imunoterapia , Melanoma/metabolismo , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA