Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
NPJ Precis Oncol ; 7(1): 135, 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38092866

RESUMO

Multiple genomic and proteomic studies have suggested that peripheral blood mononuclear cells (PBMCs) respond to tumor secretomes and thus could provide possible avenues for tumor prognosis and treatment evaluation. We hypothesized that the chromatin organization of PBMCs obtained from liquid biopsies, which integrates secretome signals with gene expression programs, provides efficient biomarkers to characterize tumor signals and the efficacy of proton therapy in tumor patients. Here, we show that chromatin imaging of PBMCs combined with machine learning methods provides such robust and predictive chromatin biomarkers. We show that such chromatin biomarkers enable the classification of 10 healthy and 10 pan-tumor patients. Furthermore, we extended our pipeline to assess the tumor types and states of 30 tumor patients undergoing (proton) radiation therapy. We show that our pipeline can thereby accurately distinguish between three tumor groups with up to 89% accuracy and enables the monitoring of the treatment effects. Collectively, we show the potential of chromatin biomarkers for cancer diagnostics and therapy evaluation.

2.
Sci Rep ; 13(1): 20662, 2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-38001169

RESUMO

The heterogenous treatment response of tumor cells limits the effectiveness of cancer therapy. While this heterogeneity has been linked to cell-to-cell variability within the complex tumor microenvironment, a quantitative biomarker that identifies and characterizes treatment-resistant cell populations is still missing. Herein, we use chromatin organization as a cost-efficient readout of the cells' states to identify subpopulations that exhibit distinct responses to radiotherapy. To this end, we developed a 3D co-culture model of cancer spheroids and patient-derived fibroblasts treated with radiotherapy. Using the model we identified treatment-resistant cells that bypassed DNA damage checkpoints and exhibited an aggressive growth phenotype. Importantly, these cells featured more condensed chromatin which primed them for treatment evasion, as inhibiting chromatin condensation and DNA damage repair mechanisms improved the efficacy of not only radio- but also chemotherapy. Collectively, our work shows the potential of using chromatin organization to cost-effectively study the heterogeneous treatment susceptibility of cells and guide therapeutic design.


Assuntos
Cromatina , Neoplasias , Humanos , Técnicas de Cocultura , Neoplasias/genética , Neoplasias/radioterapia , Reparo do DNA , Biomarcadores , Microambiente Tumoral , Esferoides Celulares , Linhagem Celular Tumoral
3.
Adv Sci (Weinh) ; 10(22): e2201663, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37218524

RESUMO

Cancer cells in secondary tumors are found to form metastases more efficiently as compared to their primary tumor counterparts. This is partially due to the unfavorable microenvironments encountered by metastasizing cancer cells that result in the survival of a more metastatic phenotype from the original population. However, the role of deleterious mechanical stresses in this change of metastatic potential is unclear. Here, by forcing cancer cells to flow through small capillary-sized constrictions, it is demonstrated that mechanical deformation can select a tumor cell subpopulation that exhibits resilience to mechanical squeezing-induced cell death. Transcriptomic profiling reveals up-regulated proliferation and DNA damage response pathways in this subpopulation, which are further translated into a more proliferative and chemotherapy-resistant phenotype. These results highlight a potential link between the microenvironmental physical stresses and the enhanced malignancy of metastasizing cancer cells which may be utilized as a therapeutic strategy in preventing the metastatic spread of cancer cells.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fenótipo , Proliferação de Células , Microambiente Tumoral
4.
Nano Lett ; 22(18): 7724-7733, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-35969027

RESUMO

For more than a century, abnormal nuclei in tumor cells, presenting subnuclear invaginations and folds on the nuclear envelope, have been known to be associated with high malignancy and poor prognosis. However, current nuclear morphology analysis focuses on the features of the entire nucleus, overlooking the malignancy-related subnuclear features in nanometer scale. The main technical challenge is to probe such tiny and randomly distributed features inside cells. We here employ nanopillar arrays to guide subnuclear features into ordered patterns, enabling their quantification as a strong indicator of cell malignancy. Both breast and liver cancer cells were validated as well as the quantification of nuclear abnormality heterogeneity. The alterations of subnuclear patterns were also explored as effective readouts for drug treatment. We envision that this nanopillar-enabled quantification of subnuclear abnormal features in tumor cells opens a new angle in characterizing malignant cells and studying the unique nuclear biology in cancer.


Assuntos
Neoplasias , Membrana Nuclear , Contagem de Células , Diferenciação Celular , Núcleo Celular , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Membrana Nuclear/patologia
5.
Sci Rep ; 11(1): 23041, 2021 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-34845273

RESUMO

Tumour progression within the tissue microenvironment is accompanied by complex biomechanical alterations of the extracellular environment. While histopathology images provide robust biochemical markers for tumor progression in clinical settings, a quantitative single cell score using nuclear morphology and chromatin organization integrated with the long range mechanical coupling within the tumor microenvironment is missing. We propose that the spatial chromatin organization in individual nuclei characterises the cell state and their alterations during tumor progression. In this paper, we first built an image analysis pipeline and implemented it to classify nuclei from patient derived breast tissue biopsies of various cancer stages based on their nuclear and chromatin features. Replacing H&E with DNA binding dyes such as Hoescht stained tissue biopsies, we improved the classification accuracy. Using the nuclear morphology and chromatin organization features, we constructed a pseudo-time model to identify the chromatin state changes that occur during tumour progression. This enabled us to build a single-cell mechano-genomic score that characterises the cell state during tumor progression from a normal to a metastatic state. To gain further insights into the alterations in the local tissue microenvironments, we also used the nuclear orientations to identify spatial neighbourhoods that have been posited to drive tumor progression. Collectively, we demonstrate that image-based single cell chromatin and nuclear features are important single cell biomarkers for phenotypic mapping of tumor progression.


Assuntos
Biomarcadores/metabolismo , Núcleo Celular/metabolismo , Cromatina/química , Neoplasias/metabolismo , Biomarcadores Tumorais , Biofísica , Biópsia , Neoplasias da Mama/metabolismo , Colágeno/química , Biologia Computacional , DNA/química , Progressão da Doença , Fibroblastos/metabolismo , Genômica , Humanos , Processamento de Imagem Assistida por Computador , Técnicas In Vitro , Aprendizado de Máquina , Metástase Neoplásica , Fenótipo , Probabilidade , Ligação Proteica , Microambiente Tumoral
6.
PLoS Comput Biol ; 16(4): e1007828, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32343706

RESUMO

Lineage tracing involves the identification of all ancestors and descendants of a given cell, and is an important tool for studying biological processes such as development and disease progression. However, in many settings, controlled time-course experiments are not feasible, for example when working with tissue samples from patients. Here we present ImageAEOT, a computational pipeline based on autoencoders and optimal transport for predicting the lineages of cells using time-labeled datasets from different stages of a cellular process. Given a single-cell image from one of the stages, ImageAEOT generates an artificial lineage of this cell based on the population characteristics of the other stages. These lineages can be used to connect subpopulations of cells through the different stages and identify image-based features and biomarkers underlying the biological process. To validate our method, we apply ImageAEOT to a benchmark task based on nuclear and chromatin images during the activation of fibroblasts by tumor cells in engineered 3D tissues. We further validate ImageAEOT on chromatin images of various breast cancer cell lines and human tissue samples, thereby linking alterations in chromatin condensation patterns to different stages of tumor progression. Our results demonstrate the promise of computational methods based on autoencoding and optimal transport principles for lineage tracing in settings where existing experimental strategies cannot be used.


Assuntos
Linhagem da Célula , Biologia Computacional/métodos , Análise de Célula Única/métodos , Neoplasias da Mama , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Núcleo Celular/fisiologia , Cromatina/fisiologia , Técnicas de Cocultura , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Reprodutibilidade dos Testes
7.
Mol Biol Cell ; 31(8): 803-812, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32023167

RESUMO

Fibroblasts are a heterogeneous group of cells comprising subpopulations that have been found to be activated in the stromal microenvironment that regulates tumor initiation and growth. The underlying mechanisms of such selective activation of fibroblasts are not understood. We propose that the intrinsic geometric heterogeneity of fibroblasts modulates the nuclear mechanotransduction of signals from the microenvironment, resulting in their selective activation. To test this, we developed an engineered 3D fibroblast tumor coculture system and used high resolution images to quantify multiple cell geometry sensitive nuclear morphological and chromatin organizational features. These features were then mapped to activation levels as measured by the nuclear abundance of transcription cofactor, megakaryoblastic leukemia, and protein levels of its target, αSMA. Importantly, our results indicate the presence of activation-"primed" cell geometries that present higher activation levels, which are further enhanced in the presence of stimuli from cancer cells. Further, we show that by enriching the population of activation-primed cell geometric states by either increasing matrix rigidity or micropatterning primed cell shapes, fibroblast activation levels can be increased. Collectively, our results reveal important cellular geometric states that select for fibroblast activation within the heterogenous tumor microenvironment.


Assuntos
Fibroblastos/ultraestrutura , Microambiente Tumoral , Actinas/metabolismo , Animais , Forma Celular , Técnicas de Cocultura , Meios de Cultivo Condicionados , Humanos , Processamento de Imagem Assistida por Computador , Células MCF-7 , Mecanotransdução Celular , Camundongos , Microscopia Confocal , Análise Multivariada , Células NIH 3T3 , Esferoides Celulares , Células Estromais/metabolismo
8.
PLoS One ; 14(7): e0218757, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31314779

RESUMO

Current cancer diagnosis involves the use of nuclear morphology and chromatin condensation signatures for accurate advanced stage classification. While such diagnostic approaches rely on high resolution imaging of the cell nucleus using expensive microscopy systems, developing portable mobile microscopes to visualize nuclear and chromatin condensation patterns is desirable at clinical settings with limited infrastructure. In this study, we develop a portable fluorescent mobile microscope capable of acquiring high resolution images of the nucleus and chromatin. Using this we extracted nuclear morphometric and chromatin texture based features and were able to discriminate between normal and cancer cells with similar accuracy as wide-field fluorescence microscopy. We were also able to detect subtle changes in nuclear and chromatin features in cells subjected to compressive forces, cytoskeletal perturbations and cytokine stimulation, thereby highlighting the sensitivity of the portable microscope. Taken together, we present a versatile platform to exploit nuclear morphometrics and chromatin condensation features as physical biomarkers for point-of-care diagnostic solutions.


Assuntos
Cromatina/genética , Cromossomos/genética , Microscopia de Fluorescência , Biomarcadores Tumorais/genética , Núcleo Celular/genética , Núcleo Celular/patologia , Heterocromatina/genética , Humanos , Processamento de Imagem Assistida por Computador , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/patologia
9.
Proc Natl Acad Sci U S A ; 115(21): E4741-E4750, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29735717

RESUMO

Cells in tissues undergo transdifferentiation programs when stimulated by specific mechanical and biochemical signals. While seminal studies have demonstrated that exogenous biochemical factors can reprogram somatic cells into pluripotent stem cells, the critical roles played by mechanical signals in such reprogramming process have not been well documented. In this paper, we show that laterally confined growth of fibroblasts on micropatterned substrates induces nuclear reprogramming with high efficiency in the absence of any exogenous reprogramming factors. We provide compelling evidence on the induction of stem cell-like properties using alkaline phosphatase assays and expression of pluripotent markers. Early onset of reprogramming was accompanied with enhanced nuclear dynamics and changes in chromosome intermingling degrees, potentially facilitating rewiring of the genome. Time-lapse analysis of promoter occupancy by immunoprecipitation of H3K9Ac chromatin fragments revealed that epithelial, proliferative, and reprogramming gene promoters were progressively acetylated, while mesenchymal promoters were deacetylated by 10 days. Consistently, RNA sequencing analysis showed a systematic progression from mesenchymal to stem cell transcriptome, highlighting pathways involving mechanisms underlying nuclear reprogramming. We then demonstrated that these mechanically reprogrammed cells could be maintained as stem cells and can be redifferentiated into multiple lineages with high efficiency. Importantly, we also demonstrate the induction of cancer stemness properties in MCF7 cells grown in such laterally confined conditions. Collectively, our results highlight an important generic property of somatic cells that, when grown in laterally confined conditions, acquire stemness. Such mechanical reprogramming of somatic cells demonstrated here has important implications in tissue regeneration and disease models.


Assuntos
Neoplasias da Mama/genética , Linhagem da Célula , Reprogramação Celular , Cromatina/genética , Células-Tronco Pluripotentes Induzidas/citologia , Transcriptoma , Animais , Transdiferenciação Celular , Epigênese Genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos , Células NIH 3T3 , Células Tumorais Cultivadas
10.
Trends Cancer ; 4(4): 320-331, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29606315

RESUMO

Abnormalities in nuclear and chromatin organization are hallmarks of many diseases including cancer. In this review, we highlight our understanding of how the cellular microenvironment regulates nuclear morphology and, with it, the spatial organization of chromosomes and genes, resulting in cell type-specific genomic programs. We also discuss the molecular basis for maintaining nuclear and genomic integrity and how alterations in nuclear mechanotransduction pathways result in various diseases. Finally, we highlight the importance of digital pathology based on nuclear morphometric features combined with single-cell genomics for early cancer diagnostics.


Assuntos
Núcleo Celular/patologia , Cromatina/patologia , Mecanotransdução Celular/genética , Neoplasias/diagnóstico , Microambiente Tumoral/genética , Biomarcadores Tumorais/análise , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Humanos , Processamento de Imagem Assistida por Computador/métodos , Aprendizado de Máquina , Imagem Molecular/métodos , Mutação , Neoplasias/genética , Neoplasias/patologia , Progéria/diagnóstico , Progéria/genética , Progéria/patologia , Análise de Célula Única/métodos , Translocação Genética
11.
Cancer Res ; 78(1): 88-102, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29074538

RESUMO

Studies of genomic instability have historically focused on intrinsic mechanisms rather than extrinsic mechanisms based in the tumor microenvironment (TME). TGFß is the most abundantly secreted cytokine in the TME, where it imparts various aggressive characteristics including invasive migration, drug resistance, and epithelial-to-mesenchymal transition (EMT). Here we show that TGFß also promotes genomic instability in the form of DNA double strand breaks (DSB) in cancer cells that lack the tumor suppressor gene RUNX3 Loss of RUNX3 resulted in transcriptional downregulation of the redox regulator heme oxygenase-1 (HO-1 or HMOX1). Consequently, elevated oxidative DNA damage disrupted genomic integrity and triggered cellular senescence, which was accompanied by tumor-promoting inflammatory cytokine expression and acquisition of the senescence-associated secretory phenotype (SASP). Recapitulating the above findings, tumors harboring a TGFß gene expression signature and RUNX3 loss exhibited higher levels of genomic instability. In summary, RUNX3 creates an effective barrier against further TGFß-dependent tumor progression by preventing genomic instability. These data suggest a novel cooperation between cancer cell-extrinsic TGFß signaling and cancer cell-intrinsic RUNX3 inactivation as aggravating factors for genomic instability.Significance: RUNX3 inactivation in cancer removes an antioxidant barrier against DNA double strand breaks induced by TGFß expressed in the tumor microenvironment. Cancer Res; 78(1); 88-102. ©2017 AACR.


Assuntos
Subunidade alfa 3 de Fator de Ligação ao Core/genética , Instabilidade Genômica , Fator de Crescimento Transformador beta/metabolismo , Células A549 , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular Tumoral , Senescência Celular/genética , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Dano ao DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Genes p53 , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta/farmacologia
12.
Nat Commun ; 8(1): 2118, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29242514

RESUMO

Phosphoinositide lipids (PPIs) are enriched in the nucleus and are accumulated at DNA damage sites. Here, we investigate roles of nuclear PPIs in DNA damage response by sequestering specific PPIs with the expression of nuclear-targeted PH domains, which inhibits recruitment of Ataxia telangiectasia and Rad3-related protein (ATR) and reduces activation of Chk1. PPI-binding domains rapidly (< 1 s) accumulate at damage sites with local enrichment of PPIs. Accumulation of PIP3 in complex with the nuclear receptor protein, SF1, at damage sites requires phosphorylation by inositol polyphosphate multikinase (IPMK) and promotes nuclear actin assembly that is required for ATR recruitment. Suppressed ATR recruitment/activation is confirmed with latrunculin A and wortmannin treatment as well as IPMK or SF1 depletion. Other DNA repair pathways involving ATM and DNA-PKcs are unaffected by PPI sequestration. Together, these findings reveal that nuclear PPI metabolism mediates an early damage response through the IPMK-dependent pathway to specifically recruit ATR.


Assuntos
Dano ao DNA , Fosfatidilinositóis/metabolismo , Transdução de Sinais , Animais , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem/genética , Quinase 1 do Ponto de Checagem/metabolismo , Reparo do DNA , Humanos , Camundongos , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Interferência de RNA , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo
13.
Sci Rep ; 7(1): 17946, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-29263424

RESUMO

Current cancer diagnosis employs various nuclear morphometric measures. While these have allowed accurate late-stage prognosis, early diagnosis is still a major challenge. Recent evidence highlights the importance of alterations in mechanical properties of single cells and their nuclei as critical drivers for the onset of cancer. We here present a method to detect subtle changes in nuclear morphometrics at single-cell resolution by combining fluorescence imaging and deep learning. This assay includes a convolutional neural net pipeline and allows us to discriminate between normal and human breast cancer cell lines (fibrocystic and metastatic states) as well as normal and cancer cells in tissue slices with high accuracy. Further, we establish the sensitivity of our pipeline by detecting subtle alterations in normal cells when subjected to small mechano-chemical perturbations that mimic tumor microenvironments. In addition, our assay provides interpretable features that could aid pathological inspections. This pipeline opens new avenues for early disease diagnostics and drug discovery.


Assuntos
Núcleo Celular/ultraestrutura , Aprendizado Profundo , Neoplasias/diagnóstico , Biomarcadores Tumorais , Linhagem Celular Tumoral/ultraestrutura , Humanos , Interpretação de Imagem Assistida por Computador/métodos , Neoplasias/diagnóstico por imagem , Neoplasias/ultraestrutura , Redes Neurais de Computação , Imagem Óptica/métodos
14.
Nat Cell Biol ; 19(12): 1400-1411, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29180822

RESUMO

The DNA damage response (DDR) preserves genomic integrity. Small non-coding RNAs termed DDRNAs are generated at DNA double-strand breaks (DSBs) and are critical for DDR activation. Here we show that active DDRNAs specifically localize to their damaged homologous genomic sites in a transcription-dependent manner. Following DNA damage, RNA polymerase II (RNAPII) binds to the MRE11-RAD50-NBS1 complex, is recruited to DSBs and synthesizes damage-induced long non-coding RNAs (dilncRNAs) from and towards DNA ends. DilncRNAs act both as DDRNA precursors and by recruiting DDRNAs through RNA-RNA pairing. Together, dilncRNAs and DDRNAs fuel DDR focus formation and associate with 53BP1. Accordingly, inhibition of RNAPII prevents DDRNA recruitment, DDR activation and DNA repair. Antisense oligonucleotides matching dilncRNAs and DDRNAs impair site-specific DDR focus formation and DNA repair. We propose that DDR signalling sites, in addition to sharing a common pool of proteins, individually host a unique set of site-specific RNAs necessary for DDR activation.


Assuntos
Quebras de DNA de Cadeia Dupla , Dano ao DNA , Reparo do DNA , RNA Longo não Codificante/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Hidrolases Anidrido Ácido , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Sistema Livre de Células , Dano ao DNA/genética , Dano ao DNA/fisiologia , Reparo do DNA/genética , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA , Proteína Homóloga a MRE11/metabolismo , Camundongos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Oligonucleotídeos Antissenso/genética , RNA Polimerase II/metabolismo , RNA Longo não Codificante/genética , Transcrição Gênica , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
15.
Proc Natl Acad Sci U S A ; 114(20): E3882-E3891, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28461498

RESUMO

Cells in physiology integrate local soluble and mechanical signals to regulate genomic programs. Whereas the individual roles of these signals are well studied, the cellular responses to the combined chemical and physical signals are less explored. Here, we investigated the cross-talk between cellular geometry and TNFα signaling. We stabilized NIH 3T3 fibroblasts into rectangular anisotropic or circular isotropic geometries and stimulated them with TNFα and analyzed nuclear translocation of transcription regulators -NFκB (p65) and MKL and downstream gene-expression patterns. We found that TNFα induces geometry-dependent actin depolymerization, which enhances IκB degradation, p65 nuclear translocation, nuclear exit of MKL, and sequestration of p65 at the RNA-polymerase-II foci. Further, global transcription profile of cells under matrix-TNFα interplay reveals a geometry-dependent gene-expression pattern. At a functional level, we find cell geometry affects TNFα-induced cell proliferation. Our results provide compelling evidence that fibroblasts, depending on their geometries, elicit distinct cellular responses for the same cytokine.


Assuntos
Expressão Gênica/fisiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Núcleo Celular/metabolismo , Forma Celular/genética , Tamanho Celular , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas I-kappa B/metabolismo , Camundongos , Células NIH 3T3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transativadores/metabolismo , Fator de Transcrição RelA/metabolismo
16.
Proc Natl Acad Sci U S A ; 111(42): E4409-18, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25298531

RESUMO

The capacity to produce therapeutically relevant quantities of multipotent mesenchymal stromal cells (MSCs) via in vitro culture is a common prerequisite for stem cell-based therapies. Although culture expanded MSCs are widely studied and considered for therapeutic applications, it has remained challenging to identify a unique set of characteristics that enables robust identification and isolation of the multipotent stem cells. New means to describe and separate this rare cell type and its downstream progenitor cells within heterogeneous cell populations will contribute significantly to basic biological understanding and can potentially improve efficacy of stem and progenitor cell-based therapies. Here, we use multivariate biophysical analysis of culture-expanded, bone marrow-derived MSCs, correlating these quantitative measures with biomolecular markers and in vitro and in vivo functionality. We find that, although no single biophysical property robustly predicts stem cell multipotency, there exists a unique and minimal set of three biophysical markers that together are predictive of multipotent subpopulations, in vitro and in vivo. Subpopulations of culture-expanded stromal cells from both adult and fetal bone marrow that exhibit sufficiently small cell diameter, low cell stiffness, and high nuclear membrane fluctuations are highly clonogenic and also exhibit gene, protein, and functional signatures of multipotency. Further, we show that high-throughput inertial microfluidics enables efficient sorting of committed osteoprogenitor cells, as distinct from these mesenchymal stem cells, in adult bone marrow. Together, these results demonstrate novel methods and markers of stemness that facilitate physical isolation, study, and therapeutic use of culture-expanded, stromal cell subpopulations.


Assuntos
Separação Celular/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Multipotentes/citologia , Adulto , Antígenos de Superfície/metabolismo , Biomarcadores/metabolismo , Fenômenos Biofísicos , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular , Linhagem da Célula , Membrana Celular/metabolismo , Proliferação de Células , Tamanho Celular , Células Cultivadas , Citoplasma/metabolismo , Feto/metabolismo , Humanos , Técnicas Analíticas Microfluídicas , Análise Multivariada , Membrana Nuclear/metabolismo , Estresse Mecânico
17.
Cell ; 158(3): 633-46, 2014 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-25083873

RESUMO

ATR controls chromosome integrity and chromatin dynamics. We have previously shown that yeast Mec1/ATR promotes chromatin detachment from the nuclear envelope to counteract aberrant topological transitions during DNA replication. Here, we provide evidence that ATR activity at the nuclear envelope responds to mechanical stress. Human ATR associates with the nuclear envelope during S phase and prophase, and both osmotic stress and mechanical stretching relocalize ATR to nuclear membranes throughout the cell cycle. The ATR-mediated mechanical response occurs within the range of physiological forces, is reversible, and is independent of DNA damage signaling. ATR-defective cells exhibit aberrant chromatin condensation and nuclear envelope breakdown. We propose that mechanical forces derived from chromosome dynamics and torsional stress on nuclear membranes activate ATR to modulate nuclear envelope plasticity and chromatin association to the nuclear envelope, thus enabling cells to cope with the mechanical strain imposed by these molecular processes.


Assuntos
Membrana Nuclear/metabolismo , Estresse Mecânico , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Cromatina/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Células HeLa , Humanos , Camundongos , Células NIH 3T3 , Osmose , Proteínas Quinases/metabolismo
18.
FEBS Lett ; 588(16): 2663-70, 2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-24747426

RESUMO

Organ size is controlled by the concerted action of biochemical and physical processes. Although mechanical forces are known to regulate cell and tissue behavior, as well as organogenesis, the precise molecular events that integrate mechanical and biochemical signals to control these processes are not fully known. The recently delineated Hippo-tumor suppressor network and its two nuclear effectors, YAP and TAZ, shed light on these mechanisms. YAP and TAZ are proto-oncogene proteins that respond to complex physical milieu represented by the rigidity of the extracellular matrix, cell geometry, cell density, cell polarity and the status of the actin cytoskeleton. Here, we review the current knowledge of how YAP and TAZ function as mechanosensors and mechanotransducers. We also suggest that by deciphering the mechanical and biochemical signals controlling YAP/TAZ function, we will gain insights into new strategies for cancer treatment and organ regeneration.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mecanotransdução Celular , Neoplasias/metabolismo , Neoplasias/patologia , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Aciltransferases , Animais , Proliferação de Células , Humanos , Tamanho do Órgão , Proto-Oncogene Mas , Proteínas de Sinalização YAP
19.
Biophys J ; 103(7): 1416-28, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23062334

RESUMO

For cells to adapt to different tissues and changes in tissue mechanics, they must be able to respond to mechanical cues by changing their gene expression patterns. Biochemical signaling pathways for these responses have been elucidated, and recent evidence points to the involvement of force-induced deformation of the nucleus. However, it is still unclear how physical cues received at the plasma membrane (PM) spatiotemporally integrate to the functional chromatin organization of the cell nucleus. To investigate this issue, we applied mechanical forces through magnetic particles adhered to the PM of single cells and mapped the accompanying changes in actin polymerization, nuclear morphology, chromatin remodeling, and nuclear transport of soluble signaling intermediates using high-resolution fluorescence anisotropy imaging. Using this approach, we show the timescales associated with force-induced polymerization of actin and changes in the F/G actin ratio resulting in nuclear translocation of the G-actin-associated transcriptional cofactor, megakaryoblastic acute leukemia factor-1 (MKL). Further, this method of measuring nuclear organization at high spatiotemporal resolution with simultaneous force application revealed the physical propagation of forces to the nucleus, resulting in changes to chromatin organization, followed by nuclear deformation. We also describe a quantitative model that incorporates active stresses and chemical kinetics to evaluate the observed timescales. Our work suggests that mechanical activation of cells is accompanied by distinct timescales involved in the reorganization of actin and chromatin assembly, followed by translocation of transcription cofactors from the cytoplasm to the nucleus.


Assuntos
Núcleo Celular/metabolismo , Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/metabolismo , Fenômenos Mecânicos , Proteínas de Fusão Oncogênica/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/química , Actinas/metabolismo , Transporte Ativo do Núcleo Celular , Fenômenos Biomecânicos , Membrana Celular/metabolismo , Sobrevivência Celular , Citoplasma/metabolismo , Polarização de Fluorescência , Células HeLa , Humanos , Imãs , Modelos Biológicos , Imagem Molecular , Multimerização Proteica , Estrutura Quaternária de Proteína , Transativadores
20.
PLoS One ; 7(9): e43718, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22957031

RESUMO

Cellular differentiation programs are accompanied by large-scale changes in nuclear organization and gene expression. In this context, accompanying transitions in chromatin assembly that facilitates changes in gene expression and cell behavior in a developmental system are poorly understood. Here, we address this gap and map structural changes in chromatin organization during murine T-cell development, to describe an unusual heterogeneity in chromatin organization and associated functional correlates in T-cell lineage. Confocal imaging of DNA assembly in cells isolated from bone marrow, thymus and spleen reveal the emergence of heterogeneous patterns in DNA organization in mature T-cells following their exit from the thymus. The central DNA pattern dominated in immature precursor cells in the thymus whereas both central and peripheral DNA patterns were observed in naïve and memory cells in circulation. Naïve T-cells with central DNA patterns exhibited higher mechanical pliability in response to compressive loads in vitro and transmigration assays in vivo, and demonstrated accelerated expression of activation-induced marker CD69. T-cell activation was characterized by marked redistribution of DNA assembly to a central DNA pattern and increased nuclear size. Notably, heterogeneity in DNA patterns recovered in cells induced into quiescence in culture, suggesting an internal regulatory mechanism for chromatin reorganization. Taken together, our results uncover an important component of plasticity in nuclear organization, reflected in chromatin assembly, during T-cell development, differentiation and transmigration.


Assuntos
Movimento Celular , DNA/metabolismo , Ativação Linfocitária , Linfócitos T/imunologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Células da Medula Óssea/citologia , Linhagem da Célula , Núcleo Celular/metabolismo , Cromatina/metabolismo , Células-Tronco Hematopoéticas/citologia , Lectinas Tipo C/metabolismo , Camundongos , Microscopia Confocal/métodos , Modelos Biológicos , Modelos Estatísticos , Análise de Sequência de DNA , Baço/citologia , Linfócitos T/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA