Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38564376

RESUMEN

RATIONALE: The chronic lung disease bronchopulmonary dysplasia (BPD) is the most severe complication of extreme prematurity. BPD results in impaired lung alveolar and vascular development and long-term respiratory morbidity, for which only supportive therapies exist. Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) improve lung structure and function in experimental BPD. Results of clinical trials with MSCs for many disorders do not yet match the promising preclinical studies. A lack of specific criteria to define functionally distinct MSCs persists. OBJECTIVES: To determine and correlate single-cell UC-MSC transcriptomic profile with therapeutic potential. METHODS: UC-MSCs from five term donors and human neonatal dermal fibroblasts (HNDFs, control cells of mesenchymal origin) transcriptomes were investigated by single-cell RNA sequencing analysis (scRNA-seq). The lung-protective effect of UC-MSCs with a distinct transcriptome and control HNDFs was tested in vivo in hyperoxia-induced neonatal lung injury in rats. MEASUREMENTS AND MAIN RESULTS: UC-MSCs showed limited transcriptomic heterogeneity, but were different from HNDFs. Gene ontology enrichment analysis revealed distinct - progenitor-like and fibroblast-like - UC-MSC subpopulations. Only the treatment with progenitor-like UC-MSCs improved lung function and structure and attenuated pulmonary hypertension in hyperoxia-exposed rat pups. Moreover, scRNA-seq identified major histocompatibility complex class I as a molecular marker of non-therapeutic cells and associated with decreased lung retention. CONCLUSIONS: UC-MSCs with a progenitor-like transcriptome, but not with a fibroblast-like transcriptome, provide lung protection in experimental BPD. High expression of major histocompatibility complex class I is associated with reduced therapeutic benefit. scRNA-seq may be useful to identify subsets of MSCs with superior repair capacity for clinical application.

2.
Stem Cells ; 40(5): 479-492, 2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35445270

RESUMEN

Late lung development is a period of alveolar and microvascular formation, which is pivotal in ensuring sufficient and effective gas exchange. Defects in late lung development manifest in premature infants as a chronic lung disease named bronchopulmonary dysplasia (BPD). Numerous studies demonstrated the therapeutic properties of exogenous bone marrow and umbilical cord-derived mesenchymal stromal cells (MSCs) in experimental BPD. However, very little is known regarding the regenerative capacity of resident lung MSCs (L-MSCs) during normal development and in BPD. In this study we aimed to characterize the L-MSC population in homeostasis and upon injury. We used single-cell RNA sequencing (scRNA-seq) to profile in situ Ly6a+ L-MSCs in the lungs of normal and O2-exposed neonatal mice (a well-established model to mimic BPD) at 3 developmental timepoints (postnatal days 3, 7, and 14). Hyperoxia exposure increased the number and altered the expression profile of L-MSCs, particularly by increasing the expression of multiple pro-inflammatory, pro-fibrotic, and anti-angiogenic genes. In order to identify potential changes induced in the L-MSCs transcriptome by storage and culture, we profiled 15 000 Ly6a+ L-MSCs after in vitro culture. We observed great differences in expression profiles of in situ and cultured L-MSCs, particularly those derived from healthy lungs. Additionally, we have identified the location of Ly6a+/Col14a1+ L-MSCs in the developing lung and propose Serpinf1 as a novel, culture-stable marker of L-MSCs. Finally, cell communication analysis suggests inflammatory signals from immune and endothelial cells as main drivers of hyperoxia-induced changes in L-MSCs transcriptome.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Células Madre Mesenquimatosas , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/terapia , Células Endoteliales , Humanos , Hiperoxia/genética , Hiperoxia/metabolismo , Recién Nacido , Pulmón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Análisis de Secuencia de ARN
3.
J Biol Chem ; 297(5): 101172, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34624316

RESUMEN

The protein Lgl1 is a key regulator of cell polarity. We previously showed that Lgl1 is inactivated by hyperphosphorylation in glioblastoma as a consequence of PTEN tumour suppressor loss and aberrant activation of the PI 3-kinase pathway; this contributes to glioblastoma pathogenesis both by promoting invasion and repressing glioblastoma cell differentiation. Lgl1 is phosphorylated by atypical protein kinase C that has been activated by binding to a complex of the scaffolding protein Par6 and active, GTP-bound Rac. The specific Rac guanine nucleotide exchange factors that generate active Rac to promote Lgl1 hyperphosphorylation in glioblastoma are unknown. We used CRISPR/Cas9 to knockout PREX1, a PI 3-kinase pathway-responsive Rac guanine nucleotide exchange factor, in patient-derived glioblastoma cells. Knockout cells had reduced Lgl1 phosphorylation, which was reversed by re-expressing PREX1. They also had reduced motility and an altered phenotype suggestive of partial neuronal differentiation; consistent with this, RNA-seq analyses identified sets of PREX1-regulated genes associated with cell motility and neuronal differentiation. PREX1 knockout in glioblastoma cells from a second patient did not affect Lgl1 phosphorylation. This was due to overexpression of a short isoform of the Rac guanine nucleotide exchange factor TIAM1; knockdown of TIAM1 in these PREX1 knockout cells reduced Lgl1 phosphorylation. These data show that PREX1 links aberrant PI 3-kinase signaling to Lgl1 phosphorylation in glioblastoma, but that TIAM1 is also to fill this role in a subset of patients. This redundancy between PREX1 and TIAM1 is only partial, as motility was impaired in PREX1 knockout cells from both patients.


Asunto(s)
Glioblastoma/metabolismo , Glicoproteínas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/metabolismo , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Glioblastoma/genética , Glicoproteínas/genética , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Proteínas de Neoplasias/genética , Fosforilación/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/genética
4.
Br J Cancer ; 127(1): 56-68, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35318435

RESUMEN

BACKGROUND: Invasive lobular carcinoma (ILC) is the second most common type of breast cancer. As few tools exist to study ILC metastasis, we isolated ILC cells with increased invasive properties to establish a spontaneously metastasising xenograft model. METHODS: MDA-MB-134VI ILC cells were placed in transwells for 7 days. Migrated cells were isolated and expanded to create the VIVA1 cell line. VIVA1 cells were compared to parental MDA-MB-134VI cells in vitro for ILC marker expression and relative proliferative and invasive ability. An intraductally injected orthotopic xenograft model was used to assess primary and metastatic tumour growth in vivo. RESULTS: Similar to MDA-MB-134VI, VIVA1 cells retained expression of oestrogen receptor (ER) and lacked expression of E-cadherin, however showed increased invasion in vitro. Following intraductal injection, VIVA1 and MDA-MB-134VI cells had similar primary tumour growth and survival kinetics. However, macrometastases were apparent in 7/10 VIVA1-injected animals. Cells from a primary orthotopic tumour (VIVA-LIG43) were isolated and showed similar proliferative rates but were also more invasive than parental cells. Upon re-injection intraductally, VIVA-LIG43 cells had more rapid tumour growth with similar metastatic incidence and location. CONCLUSIONS: We generated a new orthotopic spontaneously metastasising xenograft model for ER+ ILC amenable for the study of ILC metastasis.


Asunto(s)
Neoplasias de la Mama , Carcinoma Ductal de Mama , Carcinoma Lobular , Animales , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Lobular/metabolismo , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Receptores de Estrógenos/metabolismo
5.
Int J Mol Sci ; 23(23)2022 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-36498937

RESUMEN

Natural Killer (NK) cell cytotoxicity and interferon-gamma (IFNγ) production are profoundly suppressed postoperatively. This dysfunction is associated with increased morbidity and cancer recurrence. NK activity depends on the integration of activating and inhibitory signals, which may be modulated by transforming growth factor-beta (TGF-ß). We hypothesized that impaired postoperative NK cell IFNγ production is due to altered signaling pathways caused by postoperative TGF-ß. NK cell receptor expression, downstream phosphorylated targets, and IFNγ production were assessed using peripheral blood mononuclear cells (PBMCs) from patients undergoing cancer surgery. Healthy NK cells were incubated in the presence of healthy/baseline/postoperative day (POD) 1 plasma and in the presence/absence of a TGF-ß-blocking monoclonal antibody (mAb) or the small molecule inhibitor (smi) SB525334. Single-cell RNA sequencing (scRNA-seq) was performed on PBMCs from six patients with colorectal cancer having surgery at baseline/on POD1. Intracellular IFNγ, activating receptors (CD132, CD212, NKG2D, DNAM-1), and downstream target (STAT5, STAT4, p38 MAPK, S6) phosphorylation were significantly reduced on POD1. Furthermore, this dysfunction was phenocopied in healthy NK cells through incubation with rTGF-ß1 or POD1 plasma and was prevented by the addition of anti-TGF-ß immunotherapeutics (anti-TGF-ß mAb or TGF-ßR smi). Targeted gene analysis revealed significant decreases in S6 and FKBP12, an increase in Shp-2, and a reduction in NK metabolism-associated transcripts on POD1. pSmad2/3 was increased and pS6 was reduced in response to rTGF-ß1 on POD1, changes that were prevented by anti-TGF-ß immunotherapeutics. Together, these results suggest that both canonical and mTOR pathways downstream of TGF-ß mediate phenotypic changes that result in postoperative NK cell dysfunction.


Asunto(s)
Células Asesinas Naturales , Neoplasias , Factor de Crecimiento Transformador beta , Humanos , Leucocitos Mononucleares/metabolismo , Neoplasias/cirugía , Receptores de Células Asesinas Naturales/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Anticuerpos Monoclonales
6.
Breast Cancer Res ; 23(1): 107, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34809697

RESUMEN

BACKGROUND: Breast cancer is a highly heterogeneous disease with multiple drivers and complex regulatory networks. Periostin (Postn) is a matricellular protein involved in a plethora of cancer types and other diseases. Postn has been shown to be involved in various processes of tumor development, such as angiogenesis, invasion, cell survival and metastasis. The expression of Postn in breast cancer cells has been correlated with a more aggressive phenotype. Despite extensive research, it remains unclear how epithelial cancer cells regulate Postn expression. METHODS: Using murine tumor models and human TMAs, we have assessed the proportion of tumor samples that have acquired Postn expression in tumor cells. Using biochemical approaches and tumor cell lines derived from Neu+ murine primary tumors, we have identified major regulators of Postn gene expression in breast cancer cell lines. RESULTS: Here, we show that, while the stromal compartment typically always expresses Postn, about 50% of breast tumors acquire Postn expression in the epithelial tumor cells. Furthermore, using an in vitro model, we show a cross-regulation between FGFR, TGFß and PI3K/AKT pathways to regulate Postn expression. In HER2-positive murine breast cancer cells, we found that basic FGF can repress Postn expression through a PKC-dependent pathway, while TGFß can induce Postn expression in a SMAD-independent manner. Postn induction following the removal of the FGF-suppressive signal is dependent on PI3K/AKT signaling. CONCLUSION: Overall, these results reveal a novel regulatory mechanism and shed light on how breast tumor cells acquire Postn expression. This complex regulation is likely to be cell type and cancer specific as well as have important therapeutic implications.


Asunto(s)
Neoplasias de la Mama/genética , Moléculas de Adhesión Celular/genética , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Células Epiteliales/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología
7.
PLoS Genet ; 14(11): e1007788, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30418965

RESUMEN

Estrogen therapy increases the risk of ovarian cancer and exogenous estradiol accelerates the onset of ovarian cancer in mouse models. Both in vivo and in vitro, ovarian surface epithelial (OSE) cells exposed to estradiol develop a subpopulation that loses cell polarity, contact inhibition, and forms multi-layered foci of dysplastic cells with increased susceptibility to transformation. Here, we use single-cell RNA-sequencing to characterize this dysplastic subpopulation and identify the transcriptional dynamics involved in its emergence. Estradiol-treated cells were characterized by up-regulation of genes associated with proliferation, metabolism, and survival pathways. Pseudotemporal ordering revealed that OSE cells occupy a largely linear phenotypic spectrum that, in estradiol-treated cells, diverges towards cell state consistent with the dysplastic population. This divergence is characterized by the activation of various cancer-associated pathways including an increase in Greb1 which was validated in fallopian tube epithelium and human ovarian cancers. Taken together, this work reveals possible mechanisms by which estradiol increases epithelial cell susceptibility to tumour initiation.


Asunto(s)
Estradiol/efectos adversos , Ovario/efectos de los fármacos , Ovario/metabolismo , Animales , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Inhibición de Contacto/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Femenino , Humanos , Proteínas de la Membrana , Ratones , Ovario/patología , Fenotipo , Proteínas/genética , Proteínas/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Análisis de la Célula Individual
8.
J Appl Biomech ; 37(6): 513-521, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34689129

RESUMEN

Patterns of interjoint coordination in the kicking legs of taekwondo players were investigated to understand movement pattern variability as a functional property of skill level. Elite and nonelite players performed roundhouse kicks against a custom-built moving target fitted with an accelerometer, and movements were recorded by motion capture. Average foot segment velocities of 13.6 and 11.4 m/s were recorded for elite and nonelite players, respectively (P < .05), corresponding to target accelerations of 87.5 and 70.8g (P < .05). Gradient values derived from piecewise linear regression of continuous relative phase curves established the comparative incoordination of nonelite taekwondo players in the form of an overshoot behavior during the crucial period leading to target impact (P < .05). This overshoot was apparent in both knee-hip and ankle-knee continuous relative phase curves. Elite players generated greater limb speed and impact force through more effective limb segment coordination. The combination of continuous relative phase and piecewise linear regression techniques allowed identification of alternate joint control approaches in the 2 groups.


Asunto(s)
Artes Marciales , Tobillo , Fenómenos Biomecánicos , Humanos , Rodilla , Extremidad Inferior
9.
Biol Reprod ; 101(3): 645-658, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31187121

RESUMEN

Ovarian cancer is a complex disease with multiple subtypes, each having distinct histopathologies and variable responses to treatment. This review highlights the technological milestones and the studies that have applied them to change our definitions of ovarian cancer. Over the past 50 years, technologies such as microarrays and next-generation sequencing have led to the discovery of molecular alterations that define each of the ovarian cancer subtypes and has enabled further subclassification of the most common subtype, high-grade serous ovarian cancer (HGSOC). Improvements in mutational profiling have provided valuable insight, such as the ubiquity of TP53 mutations in HGSOC tumors. However, the information derived from these technological advances has also revealed the immense heterogeneity of this disease, from variation between patients to compositional differences within single masses. In looking forward, the emerging technologies for single-cell and spatially resolved transcriptomics will allow us to better understand the cellular composition and structure of tumors and how these contribute to the molecular subtypes. Attempts to incorporate the complexities ovarian cancer has resulted in increasing sophistication of model systems, and the increased precision in molecular profiling of ovarian cancers has already led to the introduction of inhibitors of poly (ADP-ribose) polymerases as a new class of treatments for ovarian cancer with DNA repair deficiencies. Future endeavors to define increasingly accurate classification strategies for ovarian cancer subtypes will allow for confident prediction of disease progression and provide important insight into potentially targetable molecular mechanisms specific to each subtype.


Asunto(s)
Perfilación de la Expresión Génica , Oncología Médica , Neoplasias Ováricas/clasificación , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/genética , Transcriptoma , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Análisis Mutacional de ADN/métodos , Análisis Mutacional de ADN/tendencias , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Oncología Médica/métodos , Oncología Médica/tendencias , Técnicas de Diagnóstico Molecular/métodos , Técnicas de Diagnóstico Molecular/tendencias , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Ováricas/patología
10.
Biol Reprod ; 101(5): 961-974, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31347667

RESUMEN

The ovarian surface epithelium (OSE) is a monolayer of cells surrounding the ovary that is ruptured during ovulation. After ovulation, the wound is repaired, however, this process is poorly understood. In epithelial tissues, wound repair is mediated by an epithelial-to-mesenchymal transition (EMT). Transforming Growth Factor Beta-1 (TGFß1) is a cytokine commonly known to induce an EMT and is present throughout the ovarian microenvironment. We, therefore, hypothesized that TGFß1 induces an EMT in OSE cells and activates signaling pathways important for wound repair. Treating primary cultures of mouse OSE cells with TGFß1 induced an EMT mediated by TGFßRI signaling. The transcription factor Snail was the only EMT-associated transcription factor increased by TGFß1 and, when overexpressed, was shown to increase OSE cell migration. A polymerase chain reaction array of TGFß signaling targets determined Cyclooxygenase-2 (Cox2) to be most highly induced by TGFß1. Constitutive Cox2 expression modestly increased migration and robustly enhanced cell survival, under stress conditions similar to those observed during wound repair. The increase in Snail and Cox2 expression with TGFß1 was reproduced in human OSE cultures, suggesting these responses are conserved between mouse and human. Finally, the induction of Cox2 expression in OSE cells during ovulatory wound repair was shown in vivo, suggesting TGFß1 increases Cox2 to promote wound repair by enhancing cell survival. These data support that TGFß1 promotes ovulatory wound repair by induction of an EMT and activation of a COX2-mediated pro-survival pathway. Understanding ovulatory wound repair may give insight into why ovulation is the primary non-hereditary risk factor for ovarian cancer.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Ovario/fisiología , Cicatrización de Heridas , Animales , Supervivencia Celular , Ciclooxigenasa 2/genética , Dinoprostona/genética , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Regulación de la Expresión Génica , Ratones , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
11.
Nat Commun ; 15(1): 6550, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095365

RESUMEN

The cardiac conduction system (CCS) is a network of specialized cardiomyocytes that coordinates electrical impulse generation and propagation for synchronized heart contractions. Although the components of the CCS, including the sinoatrial node, atrioventricular node, His bundle, bundle branches, and Purkinje fibers, were anatomically discovered more than 100 years ago, their molecular constituents and regulatory mechanisms remain incompletely understood. Here, we demonstrate the transcriptomic landscape of the postnatal mouse CCS at a single-cell resolution with spatial information. Integration of single-cell and spatial transcriptomics uncover region-specific markers and zonation patterns of expression. Network inference shows heterogeneous gene regulatory networks across the CCS. Notably, region-specific gene regulation is recapitulated in vitro using neonatal mouse atrial and ventricular myocytes overexpressing CCS-specific transcription factors, Tbx3 and/or Irx3. This finding is supported by ATAC-seq of different CCS regions, Tbx3 ChIP-seq, and Irx motifs. Overall, this study provides comprehensive molecular profiles of the postnatal CCS and elucidates gene regulatory mechanisms contributing to its heterogeneity.


Asunto(s)
Sistema de Conducción Cardíaco , Proteínas de Homeodominio , Miocitos Cardíacos , Proteínas de Dominio T Box , Animales , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Redes Reguladoras de Genes , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Regulación de la Expresión Génica , Animales Recién Nacidos , Análisis de la Célula Individual , Transcriptoma , Ramos Subendocárdicos/metabolismo , Ramos Subendocárdicos/fisiología , Nodo Atrioventricular/metabolismo , Nodo Sinoatrial/metabolismo , Fascículo Atrioventricular/metabolismo
12.
Sci Rep ; 14(1): 787, 2024 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-38191799

RESUMEN

The tumour microenvironment is infiltrated by immunosuppressive cells, such as regulatory T cells (Tregs), which contribute to tumour escape and impede immunotherapy outcomes. Soluble fibrinogen-like protein 2 (sFGL2), a Treg effector protein, inhibits immune cell populations, via receptors FcγRIIB and FcγRIII, leading to downregulation of CD86 in antigen presenting cells and limiting T cell activation. Increased FGL2 expression is associated with tumour progression and poor survival in several different cancers, such as glioblastoma multiforme, lung, renal, liver, colorectal, and prostate cancer. Querying scRNA-seq human cancer data shows FGL2 is produced by cells in the tumour microenvironment (TME), particularly monocytes and macrophages as well as T cells and dendritic cells (DCs), while cancer cells have minimal expression of FGL2. We studied the role of FGL2 exclusively produced by cells in the TME, by leveraging Fgl2 knockout mice. We tested two murine models of cancer in which the role of FGL2 has not been previously studied: epithelial ovarian cancer and melanoma. We show that absence of FGL2 leads to a more activated TME, including activated DCs (CD86+, CD40+) and T cells (CD25+, TIGIT+), as well as demonstrating for the first time that the absence of FGL2 leads to more activated natural killer cells (DNAM-1+, NKG2D+) in the TME. Furthermore, the absence of FGL2 leads to prolonged survival in the B16F10 melanoma model, while the absence of FGL2 synergizes with oncolytic virus to prolong survival in the ID8-p53-/-Brca2-/- ovarian cancer model. In conclusion, targeting FGL2 is a promising cancer treatment strategy alone and in combination immunotherapies.


Asunto(s)
Fibrinógeno , Melanoma , Neoplasias Ováricas , Animales , Femenino , Humanos , Ratones , Células Presentadoras de Antígenos , Carcinoma Epitelial de Ovario , Melanoma/genética , Melanoma/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Microambiente Tumoral
13.
J Exp Med ; 221(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38869480

RESUMEN

While conventional wisdom initially postulated that PD-L1 serves as the inert ligand for PD-1, an emerging body of literature suggests that PD-L1 has cell-intrinsic functions in immune and cancer cells. In line with these studies, here we show that engagement of PD-L1 via cellular ligands or agonistic antibodies, including those used in the clinic, potently inhibits the type I interferon pathway in cancer cells. Hampered type I interferon responses in PD-L1-expressing cancer cells resulted in enhanced efficacy of oncolytic viruses in vitro and in vivo. Consistently, PD-L1 expression marked tumor explants from cancer patients that were best infected by oncolytic viruses. Mechanistically, PD-L1 promoted a metabolic shift characterized by enhanced glycolysis rate that resulted in increased lactate production. In turn, lactate inhibited type I IFN responses. In addition to adding mechanistic insight into PD-L1 intrinsic function, our results will also help guide the numerous ongoing efforts to combine PD-L1 antibodies with oncolytic virotherapy in clinical trials.


Asunto(s)
Antígeno B7-H1 , Interferón Tipo I , Viroterapia Oncolítica , Virus Oncolíticos , Animales , Femenino , Humanos , Ratones , Antígeno B7-H1/metabolismo , Antígeno B7-H1/inmunología , Antígeno B7-H1/genética , Línea Celular Tumoral , Glucólisis , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Ácido Láctico/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/metabolismo , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Transducción de Señal , Masculino
14.
Br J Sports Med ; 47(18): 1161-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22930694

RESUMEN

OBJECTIVE: The objective of this study was to assess the effect of taekwondo kicks and peak foot velocity (FVEL) on resultant head linear acceleration (RLA), head injury criterion (HIC15) and head velocity (HVEL). METHODS: Each subject (n=12) randomly performed five repetitions of the turning kick (TK), clench axe kick (CA), front leg axe kick, jump back kick (JB) and jump spinning hook kick (JH) at the average standing head height for competitors in their weight division. A Hybrid II Crash Test Dummy head was fitted with a protective taekwondo helmet and instrumented with a triaxial accelerometer and fixed to a height-adjustable frame. Resultant head linear acceleration, HVEL, FVEL data were captured and processed using Qualysis Track Manager. RESULTS: The TK (130.11 ± 51.67 g) produced a higher RLA than the CA (54.95 ± 20.08 g, p<0.001, d=1.84) and a higher HIC15 than the JH (672.74 ± 540.89 vs 300.19 ± 144.35, p<0.001, ES=0.97). There was no difference in HVEL of the TK (4.73 ± 1.67 m/s) and that of the JB (4.43 ± 0.78 m/s; p=0.977, ES<0.01). CONCLUSIONS: The TK is of concern because it is the most common technique and cause of concussion in taekwondo. Future studies should aim to understand rotational accelerations of the head.


Asunto(s)
Conmoción Encefálica/etiología , Artes Marciales/lesiones , Aceleración , Fenómenos Biomecánicos/fisiología , Conmoción Encefálica/fisiopatología , Diseño de Equipo , Pie/fisiología , Humanos , Masculino , Maniquíes , Movimiento/fisiología , Medicina Deportiva/instrumentación , Adulto Joven
15.
NPJ Syst Biol Appl ; 9(1): 61, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38086828

RESUMEN

Cancer cells often metastasize by undergoing an epithelial-mesenchymal transition (EMT). Although abundance of CD8+ T-cells in the tumor microenvironment correlates with improved survival, mesenchymal cancer cells acquire greater resistance to antitumor immunity in some cancers. We hypothesized the EMT modulates the immune response to ovarian cancer. Here we show that cancer cells from infiltrated/inflamed tumors possess more mesenchymal cells, than excluded and desert tumors. We also noted high expression of LGALS3 is associated with EMT in vivo, a finding validated with in vitro EMT models. Dissecting the cellular communications among populations in the tumor revealed that mesenchymal cancer cells in infiltrated tumors communicate through LGALS3 to LAG3 receptor expressed by CD8+ T cells. We found CD8+ T cells express high levels of LAG3, a marker of T cell exhaustion. The results indicate that EMT in ovarian cancer cells promotes interactions between cancer cells and T cells through the LGALS3 - LAG3 axis, which could increase T cell exhaustion in infiltrated tumors, dampening antitumor immunity.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias Ováricas , Humanos , Femenino , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Galectina 3/genética , Galectina 3/metabolismo , Agotamiento de Células T , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Microambiente Tumoral
16.
Cell Rep ; 42(1): 111978, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36640358

RESUMEN

Hypertranscription supports biosynthetically demanding cellular states through global transcriptome upregulation. Despite its potential widespread relevance, documented examples of hypertranscription remain few and limited to early development. Here, we demonstrate that absolute scaling of single-cell RNA-sequencing data enables the estimation of total transcript abundances per cell. We validate absolute scaling in known cases of developmental hypertranscription and apply it to adult cell types, revealing a remarkable dynamic range in transcriptional output. In adult organs, hypertranscription marks activated stem/progenitor cells with multilineage potential and is redeployed in conditions of tissue injury, where it precedes bursts of proliferation during regeneration. Our analyses identify a common set of molecular pathways associated with both adult and embryonic hypertranscription, including chromatin remodeling, DNA repair, ribosome biogenesis, and translation. These shared features across diverse cell contexts support hypertranscription as a general and dynamic cellular program that is pervasively employed during development, organ maintenance, and regeneration.


Asunto(s)
Células Madre , Transcriptoma , Transcriptoma/genética , Células Madre/metabolismo , Activación Transcripcional , Ensamble y Desensamble de Cromatina , Análisis de la Célula Individual
17.
iScience ; 26(3): 106093, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36843845

RESUMEN

Aberrant neurodevelopment is a core deficit of autism spectrum disorder (ASD). Here we ask whether a non-genetic factor, prenatal exposure to the environmental pollutant methylmercury (MeHg), is a contributing factor in ASD onset. We showed that adult mice prenatally exposed to non-apoptotic MeHg exhibited key ASD characteristics, including impaired communication, reduced sociability, and increased restrictive repetitive behaviors, whereas in the embryonic cortex, prenatal MeHg exposure caused premature neuronal differentiation. Further single-cell RNA sequencing (scRNA-seq) analysis disclosed that prenatal exposure to MeHg resulted in cortical radial glial precursors (RGPs) favoring asymmetric differentiation to directly generate cortical neurons, omitting the intermediate progenitor stage. In addition, MeHg exposure in cultured RGPs increased CREB phosphorylation and enhanced the interaction between CREB and CREB binding protein (CBP). Intriguingly, metformin, an FDA-approved drug, can reverse MeHg-induced premature neuronal differentiation via CREB/CBP repulsion. These findings provide insights into ASD etiology, its underlying mechanism, and a potential therapeutic strategy.

18.
Elife ; 122023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-37078698

RESUMEN

We sought to define the mechanism underlying lung microvascular regeneration in a model of severe acute lung injury (ALI) induced by selective lung endothelial cell ablation. Intratracheal instillation of DT in transgenic mice expressing human diphtheria toxin (DT) receptor targeted to ECs resulted in ablation of >70% of lung ECs, producing severe ALI with near complete resolution by 7 days. Using single-cell RNA sequencing, eight distinct endothelial clusters were resolved, including alveolar aerocytes (aCap) ECs expressing apelin at baseline and general capillary (gCap) ECs expressing the apelin receptor. At 3 days post-injury, a novel gCap EC population emerged characterized by de novo expression of apelin, together with the stem cell marker, protein C receptor. These stem-like cells transitioned at 5 days to proliferative endothelial progenitor-like cells, expressing apelin receptor together with the pro-proliferative transcription factor, Foxm1, and were responsible for the rapid replenishment of all depleted EC populations by 7 days post-injury. Treatment with an apelin receptor antagonist prevented ALI resolution and resulted in excessive mortality, consistent with a central role for apelin signaling in EC regeneration and microvascular repair. The lung has a remarkable capacity for microvasculature EC regeneration which is orchestrated by newly emergent apelin-expressing gCap endothelial stem-like cells that give rise to highly proliferative, apelin receptor-positive endothelial progenitors responsible for the regeneration of the lung microvasculature.


Asunto(s)
Lesión Pulmonar Aguda , Apelina , Pulmón , Animales , Ratones , Medicina Regenerativa , Apelina/genética , Apelina/metabolismo , Células Endoteliales , Ratones Transgénicos , Pulmón/irrigación sanguínea
19.
Commun Biol ; 6(1): 1152, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37957414

RESUMEN

Ovarian cancers exhibit high rates of recurrence and poor treatment response. Preclinical models that recapitulate human disease are critical to develop new therapeutic approaches. Syngeneic mouse models allow for the generation of tumours comprising the full repertoire of non-malignant cell types but have expanded in number, varying in the cell type of origin, method for transformation, and ultimately, the properties of the tumours they produce. Here we have performed a comparative analysis of high-grade serous ovarian cancer models based on transcriptomic profiling of 22 cell line models, and intrabursal and intraperitoneal tumours from 12. Among cell lines, we identify distinct signalling activity, such as elevated inflammatory signalling in STOSE and OVE16 models, and MAPK/ERK signalling in ID8 and OVE4 models; metabolic differences, such as reduced glycolysis-associated expression in several engineered ID8 subclones; and relevant functional properties, including differences in EMT activation, PD-L1 and MHC class I expression, and predicted chemosensitivity. Among tumour samples, we observe increased variability and stromal content among intrabursal tumours. Finally, we predict differences in the microenvironment of ID8 models engineered with clinically relevant mutations. We anticipate that this work will serve as a valuable resource, providing new insight to help select models for specific experimental objectives.


Asunto(s)
Neoplasias Ováricas , Animales , Ratones , Humanos , Femenino , Neoplasias Ováricas/patología , Perfilación de la Expresión Génica , Transducción de Señal , Microambiente Tumoral/genética
20.
Sci Adv ; 8(1): eabi7640, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34985957

RESUMEN

Epithelial-mesenchymal plasticity (EMP) contributes to tumor progression, promoting therapy resistance and immune cell evasion. Definitive molecular features of this plasticity have largely remained elusive due to the limited scale of most studies. Leveraging single-cell RNA sequencing data from 266 tumors spanning eight different cancer types, we identify expression patterns associated with intratumoral EMP. Integrative analysis of these programs confirmed a high degree of diversity among tumors. These diverse programs are associated with combinations of various common regulatory mechanisms initiated from cues within the tumor microenvironment. We show that inferring regulatory features can inform effective therapeutics to restrict EMP.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA