Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell ; 175(1): 43-56.e21, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30241615

RESUMEN

Stem cell regulation and hierarchical organization of human skeletal progenitors remain largely unexplored. Here, we report the isolation of a self-renewing and multipotent human skeletal stem cell (hSSC) that generates progenitors of bone, cartilage, and stroma, but not fat. Self-renewing and multipotent hSSCs are present in fetal and adult bones and can also be derived from BMP2-treated human adipose stroma (B-HAS) and induced pluripotent stem cells (iPSCs). Gene expression analysis of individual hSSCs reveals overall similarity between hSSCs obtained from different sources and partially explains skewed differentiation toward cartilage in fetal and iPSC-derived hSSCs. hSSCs undergo local expansion in response to acute skeletal injury. In addition, hSSC-derived stroma can maintain human hematopoietic stem cells (hHSCs) in serum-free culture conditions. Finally, we combine gene expression and epigenetic data of mouse skeletal stem cells (mSSCs) and hSSCs to identify evolutionarily conserved and divergent pathways driving SSC-mediated skeletogenesis. VIDEO ABSTRACT.


Asunto(s)
Desarrollo Óseo/fisiología , Huesos/citología , Células Madre Hematopoyéticas/citología , Animales , Huesos/metabolismo , Cartílago/citología , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Análisis de la Célula Individual/métodos , Células Madre/citología , Células del Estroma/citología , Transcriptoma/genética
2.
Nature ; 597(7875): 256-262, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34381212

RESUMEN

Loss of skeletal integrity during ageing and disease is associated with an imbalance in the opposing actions of osteoblasts and osteoclasts1. Here we show that intrinsic ageing of skeletal stem cells (SSCs)2 in mice alters signalling in the bone marrow niche and skews the differentiation of bone and blood lineages, leading to fragile bones that regenerate poorly. Functionally, aged SSCs have a decreased bone- and cartilage-forming potential but produce more stromal lineages that express high levels of pro-inflammatory and pro-resorptive cytokines. Single-cell RNA-sequencing studies link the functional loss to a diminished transcriptomic diversity of SSCs in aged mice, which thereby contributes to the transformation of the bone marrow niche. Exposure to a youthful circulation through heterochronic parabiosis or systemic reconstitution with young haematopoietic stem cells did not reverse the diminished osteochondrogenic activity of aged SSCs, or improve bone mass or skeletal healing parameters in aged mice. Conversely, the aged SSC lineage promoted osteoclastic activity and myeloid skewing by haematopoietic stem and progenitor cells, suggesting that the ageing of SSCs is a driver of haematopoietic ageing. Deficient bone regeneration in aged mice could only be returned to youthful levels by applying a combinatorial treatment of BMP2 and a CSF1 antagonist locally to fractures, which reactivated aged SSCs and simultaneously ablated the inflammatory, pro-osteoclastic milieu. Our findings provide mechanistic insights into the complex, multifactorial mechanisms that underlie skeletal ageing and offer prospects for rejuvenating the aged skeletal system.


Asunto(s)
Envejecimiento/patología , Huesos/patología , Senescencia Celular , Inflamación/patología , Nicho de Células Madre , Células Madre/patología , Animales , Proteína Morfogenética Ósea 2/metabolismo , Regeneración Ósea , Linaje de la Célula , Femenino , Curación de Fractura , Hematopoyesis , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Células Mieloides/citología , Osteoclastos/citología , Rejuvenecimiento
3.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34244432

RESUMEN

Natural killer (NK) cells comprise one subset of the innate lymphoid cell (ILC) family. Despite reported antitumor functions of NK cells, their tangible contribution to tumor control in humans remains controversial. This is due to incomplete understanding of the NK cell states within the tumor microenvironment (TME). Here, we demonstrate that peripheral circulating NK cells differentiate down two divergent pathways within the TME, resulting in different end states. One resembles intraepithelial ILC1s (ieILC1) and possesses potent in vivo antitumor activity. The other expresses genes associated with immune hyporesponsiveness and has poor antitumor functional capacity. Interleukin-15 (IL-15) and direct contact between the tumor cells and NK cells are required for the differentiation into CD49a+CD103+ cells, resembling ieILC1s. These data explain the similarity between ieILC1s and tissue-resident NK cells, provide insight into the origin of ieILC1s, and identify the ieILC1-like cell state within the TME to be the NK cell phenotype with the greatest antitumor activity. Because the proportions of the different ILC states vary between tumors, these findings provide a resource for the clinical study of innate immune responses against tumors and the design of novel therapy.


Asunto(s)
Neoplasias de Cabeza y Cuello/inmunología , Inmunidad Innata/inmunología , Células Asesinas Naturales/inmunología , Linfocitos/inmunología , Microambiente Tumoral/inmunología , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Antineoplásicos/metabolismo , Diferenciación Celular/inmunología , Línea Celular Tumoral , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Interleucina-15/metabolismo , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Fenotipo , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
4.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34620713

RESUMEN

In the skin, tissue injury results in fibrosis in the form of scars composed of dense extracellular matrix deposited by fibroblasts. The therapeutic goal of regenerative wound healing has remained elusive, in part because principles of fibroblast programming and adaptive response to injury remain incompletely understood. Here, we present a multimodal -omics platform for the comprehensive study of cell populations in complex tissue, which has allowed us to characterize the cells involved in wound healing across both time and space. We employ a stented wound model that recapitulates human tissue repair kinetics and multiple Rainbow transgenic lines to precisely track fibroblast fate during the physiologic response to skin injury. Through integrated analysis of single cell chromatin landscapes and gene expression states, coupled with spatial transcriptomic profiling, we are able to impute fibroblast epigenomes with temporospatial resolution. This has allowed us to reveal potential mechanisms controlling fibroblast fate during migration, proliferation, and differentiation following skin injury, and thereby reexamine the canonical phases of wound healing. These findings have broad implications for the study of tissue repair in complex organ systems.


Asunto(s)
Cicatriz/patología , Fibroblastos/metabolismo , Fibrosis/patología , Piel/lesiones , Cicatrización de Heridas/fisiología , Animales , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Matriz Extracelular/metabolismo , Femenino , Mecanotransducción Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Piel/metabolismo
5.
Orbit ; : 1-4, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38567699

RESUMEN

Disseminated intravascular coagulation (DIC) is characterized by abnormal activation of the coagulation cascade, which leads to simultaneous hypercoagulation and excessive bleeding. While it typically occurs in systemic diseases, such as infection, inflammation, obstetric complications, and malignancy, it can rarely manifest postoperatively. This case report describes a patient who presented with prolonged, refractory bleeding after ectropion repair via a lateral tarsal strip procedure. Due to the inability to control the patient's bleeding with conservative measures followed by surgical exploration and electrocautery, the patient underwent a hematologic work-up. Laboratory studies were consistent with DIC, attributed to his large burden of endovascular stents. He was treated with anticoagulation using apixaban in addition to tranexamic acid to achieve lasting hemostasis. This case highlights the importance of thorough preoperative assessments, even for minor surgical procedures, and systemic workup for atypical postoperative bleeding.

6.
Proc Natl Acad Sci U S A ; 116(50): 25115-25125, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31754028

RESUMEN

Hematopoietic stem cells (HSCs) self-renew and generate all blood cells. Recent studies with single cell transplants and lineage tracing suggest that adult HSCs are diverse in their reconstitution and lineage potentials. However, prospective isolation of these subpopulations has remained challenging. Here, we identify Neogenin-1 (NEO1) as a unique surface marker on a fraction of mouse HSCs labeled with Hoxb5, a specific reporter of long-term HSCs (LT-HSCs). We show that NEO1+Hoxb5+ LT-HSCs expand with age and respond to myeloablative stress in young mice while NEO1-Hoxb5+ LT-HSCs exhibit no significant change in number. Furthermore, NEO1+Hoxb5+ LT-HSCs are more often in the G2/S cell cycle phase compared to NEO1-Hoxb5+ LT-HSCs in both young and old bone marrow. Upon serial transplantation, NEO1+Hoxb5+ LT-HSCs exhibit myeloid-biased differentiation and reduced reconstitution while NEO1-Hoxb5+ LT-HSCs are lineage-balanced and stably reconstitute recipients. Gene expression analysis reveals erythroid and myeloid priming in the NEO1+ fraction and association of quiescence and self-renewal-related transcription factors with NEO1- LT-HSCs. Finally, transplanted NEO1+Hoxb5+ LT-HSCs rarely generate NEO1-Hoxb5+ LT-HSCs while NEO1-Hoxb5+ LT-HSCs repopulate both LT-HSC fractions. This supports a model in which dormant, balanced NEO1-Hoxb5+ LT-HSCs can hierarchically precede active, myeloid-biased NEO1+Hoxb5+ LT-HSCs.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Proteínas de Homeodominio/metabolismo , Proteínas de la Membrana/metabolismo , Envejecimiento , Animales , Femenino , Trasplante de Células Madre Hematopoyéticas , Proteínas de Homeodominio/genética , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos
7.
J Gen Intern Med ; 37(11): 2868-2869, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35581457
8.
bioRxiv ; 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38562882

RESUMEN

Single-cell RNA sequencing (scRNA-seq) has transformed our understanding of cell fate in developmental systems. However, identifying the molecular hallmarks of potency - the capacity of a cell to differentiate into other cell types - has remained challenging. Here, we introduce CytoTRACE 2, an interpretable deep learning framework for characterizing potency and differentiation states on an absolute scale from scRNA-seq data. Across 31 human and mouse scRNA-seq datasets encompassing 28 tissue types, CytoTRACE 2 outperformed existing methods for recovering experimentally determined potency levels and differentiation states covering the entire range of cellular ontogeny. Moreover, it reconstructed the temporal hierarchy of mouse embryogenesis across 62 timepoints; identified pan-tissue expression programs that discriminate major potency levels; and facilitated discovery of cellular phenotypes in cancer linked to survival and immunotherapy resistance. Our results illuminate a fundamental feature of cell biology and provide a broadly applicable platform for delineating single-cell differentiation landscapes in health and disease.

9.
J Infect Dev Ctries ; 16(3): 402-408, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35404843

RESUMEN

Coronaviruses have been responsible for the emergence of pathogenic human diseases in recent decades, especially the coronavirus disease of 2019 (COVID-19). Phylogenetic studies of RNA (ribonucleic acid) viruses suggest that most human coronaviruses originated in bats, which are suitable reservoir hosts for many zoonotic viruses because of their unique biological and physiological features. The generation of human pathogenic coronaviruses is a result of genetic adaptation in bats and/or intermediate hosts, leading to spillover events. Therefore, we propose that specifically reducing or disrupting persistent coronavirus infection in bats may consequently decrease the frequency of human coronavirus diseases. We suggest several strategies to achieve the aforementioned goal in bats, including vaccination and targeted delivery of molecular inhibitors, such as monoclonal antibodies, aptamers, antisense oligonucleotides, and siRNA by use of viral nanoparticles. Advances in global bat research with the aim of controlling coronavirus infection in these mammals are pivotal in enhancing human health worldwide.


Asunto(s)
COVID-19 , Quirópteros , Animales , Humanos , Filogenia
10.
Nat Med ; 28(2): 353-362, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35027754

RESUMEN

Severe immune-related adverse events (irAEs) occur in up to 60% of patients with melanoma treated with immune checkpoint inhibitors (ICIs). However, it is unknown whether a common baseline immunological state precedes irAE development. Here we applied mass cytometry by time of flight, single-cell RNA sequencing, single-cell V(D)J sequencing, bulk RNA sequencing and bulk T cell receptor (TCR) sequencing to study peripheral blood samples from patients with melanoma treated with anti-PD-1 monotherapy or anti-PD-1 and anti-CTLA-4 combination ICIs. By analyzing 93 pre- and early on-ICI blood samples and 3 patient cohorts (n = 27, 26 and 18), we found that 2 pretreatment factors in circulation-activated CD4 memory T cell abundance and TCR diversity-are associated with severe irAE development regardless of organ system involvement. We also explored on-treatment changes in TCR clonality among patients receiving combination therapy and linked our findings to the severity and timing of irAE onset. These results demonstrate circulating T cell characteristics associated with ICI-induced toxicity, with implications for improved diagnostics and clinical management.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Melanoma , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Melanoma/tratamiento farmacológico , Estudios Retrospectivos , Linfocitos T
11.
Sci Adv ; 8(45): eabm3548, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36351009

RESUMEN

Metastasis is responsible for most breast cancer-related deaths; however, identifying the cellular determinants of metastasis has remained challenging. Here, we identified a minority population of immature THY1+/VEGFA+ tumor epithelial cells in human breast tumor biopsies that display angiogenic features and are marked by the expression of the oncogene, LMO2. Higher abundance of LMO2+ basal cells correlated with tumor endothelial content and predicted poor distant recurrence-free survival in patients. Using MMTV-PyMT/Lmo2CreERT2 mice, we demonstrated that Lmo2 lineage-traced cells integrate into the vasculature and have a higher propensity to metastasize. LMO2 knockdown in human breast tumors reduced lung metastasis by impairing intravasation, leading to a reduced frequency of circulating tumor cells. Mechanistically, we find that LMO2 binds to STAT3 and is required for STAT3 activation by tumor necrosis factor-α and interleukin-6. Collectively, our study identifies a population of metastasis-initiating cells with angiogenic features and establishes the LMO2-STAT3 signaling axis as a therapeutic target in breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama , Neoplasias Pulmonares , Humanos , Ratones , Animales , Femenino , Neoplasias de la Mama/patología , Neoplasias Pulmonares/metabolismo , Transducción de Señal , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo
12.
Nat Commun ; 13(1): 6491, 2022 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-36310174

RESUMEN

Sexually dimorphic tissues are formed by cells that are regulated by sex hormones. While a number of systemic hormones and transcription factors are known to regulate proliferation and differentiation of osteoblasts and osteoclasts, the mechanisms that determine sexually dimorphic differences in bone regeneration are unclear. To explore how sex hormones regulate bone regeneration, we compared bone fracture repair between adult male and female mice. We found that skeletal stem cell (SSC) mediated regeneration in female mice is dependent on estrogen signaling but SSCs from male mice do not exhibit similar estrogen responsiveness. Mechanistically, we found that estrogen acts directly on the SSC lineage in mice and humans by up-regulating multiple skeletogenic pathways and is necessary for the stem cell's ability to self- renew and differentiate. Our results also suggest a clinically applicable strategy to accelerate bone healing using localized estrogen hormone therapy.


Asunto(s)
Osteoblastos , Células Madre , Humanos , Masculino , Femenino , Ratones , Animales , Osteoblastos/metabolismo , Diferenciación Celular , Osteoclastos , Estrógenos/farmacología , Estrógenos/metabolismo
13.
Science ; 371(6529)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33542111

RESUMEN

During development, cells progress from a pluripotent state to a more restricted fate within a particular germ layer. However, cranial neural crest cells (CNCCs), a transient cell population that generates most of the craniofacial skeleton, have much broader differentiation potential than their ectodermal lineage of origin. Here, we identify a neuroepithelial precursor population characterized by expression of canonical pluripotency transcription factors that gives rise to CNCCs and is essential for craniofacial development. Pluripotency factor Oct4 is transiently reactivated in CNCCs and is required for the subsequent formation of ectomesenchyme. Furthermore, open chromatin landscapes of Oct4+ CNCC precursors resemble those of epiblast stem cells, with additional features suggestive of priming for mesenchymal programs. We propose that CNCCs expand their developmental potential through a transient reacquisition of molecular signatures of pluripotency.


Asunto(s)
Cresta Neural/embriología , Células Madre Pluripotentes/fisiología , Animales , Diferenciación Celular/genética , Movimiento Celular , Embrión de Mamíferos , Estratos Germinativos/citología , Ratones , Cresta Neural/citología , Cresta Neural/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , RNA-Seq , Transcripción Genética , Transcriptoma
14.
Elife ; 92020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33236985

RESUMEN

The balance of hematopoietic stem cell (HSC) self-renewal and differentiation is critical for a healthy blood supply; imbalances underlie hematological diseases. The importance of HSCs and their progenitors have led to their extensive characterization at genomic and transcriptomic levels. However, the proteomics of hematopoiesis remains incompletely understood. Here we report a proteomics resource from mass spectrometry of mouse young adult and old adult mouse HSCs, multipotent progenitors and oligopotent progenitors; 12 cell types in total. We validated differential protein levels, including confirmation that Dnmt3a protein levels are undetected in young adult mouse HSCs until forced into cycle. Additionally, through integrating proteomics and RNA-sequencing datasets, we identified a subset of genes with apparent post-transcriptional repression in young adult mouse HSCs. In summary, we report proteomic coverage of young and old mouse HSCs and progenitors, with broader implications for understanding mechanisms for stem cell maintenance, niche interactions and fate determination.


Asunto(s)
Envejecimiento/fisiología , Regulación de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteómica , Animales , Ratones , Ratones Endogámicos C57BL , Proteoma , Procesamiento Postranscripcional del ARN
15.
Nat Med ; 26(10): 1583-1592, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32807933

RESUMEN

Osteoarthritis (OA) is a degenerative disease resulting in irreversible, progressive destruction of articular cartilage1. The etiology of OA is complex and involves a variety of factors, including genetic predisposition, acute injury and chronic inflammation2-4. Here we investigate the ability of resident skeletal stem-cell (SSC) populations to regenerate cartilage in relation to age, a possible contributor to the development of osteoarthritis5-7. We demonstrate that aging is associated with progressive loss of SSCs and diminished chondrogenesis in the joints of both mice and humans. However, a local expansion of SSCs could still be triggered in the chondral surface of adult limb joints in mice by stimulating a regenerative response using microfracture (MF) surgery. Although MF-activated SSCs tended to form fibrous tissues, localized co-delivery of BMP2 and soluble VEGFR1 (sVEGFR1), a VEGF receptor antagonist, in a hydrogel skewed differentiation of MF-activated SSCs toward articular cartilage. These data indicate that following MF, a resident stem-cell population can be induced to generate cartilage for treatment of localized chondral disease in OA.


Asunto(s)
Cartílago Articular/fisiología , Regeneración/fisiología , Células Madre/fisiología , Adulto , Animales , Cartílago Articular/citología , Diferenciación Celular , Células Cultivadas , Condrocitos/citología , Condrocitos/fisiología , Condrogénesis/fisiología , Trasplante de Tejido Fetal , Feto/citología , Xenoinjertos , Humanos , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre/citología , Ingeniería de Tejidos/métodos
16.
Cell Stem Cell ; 27(2): 284-299.e8, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32693087

RESUMEN

SMAD pathways govern epithelial proliferation, and transforming growth factor ß (TGF-ß and BMP signaling through SMAD members has distinct effects on mammary development and homeostasis. Here, we show that LEFTY1, a secreted inhibitor of NODAL/SMAD2 signaling, is produced by mammary progenitor cells and, concomitantly, suppresses SMAD2 and SMAD5 signaling to promote long-term proliferation of normal and malignant mammary epithelial cells. In contrast, BMP7, a NODAL antagonist with context-dependent functions, is produced by basal cells and restrains progenitor cell proliferation. In normal mouse epithelium, LEFTY1 expression in a subset of luminal cells and rare basal cells opposes BMP7 to promote ductal branching. LEFTY1 binds BMPR2 to suppress BMP7-induced activation of SMAD5, and this LEFTY1-BMPR2 interaction is specific to tumor-initiating cells in triple-negative breast cancer xenografts that rely on LEFTY1 for growth. These results suggest that LEFTY1 is an endogenous dual-SMAD inhibitor and that suppressing its function may represent a therapeutic vulnerability in breast cancer.


Asunto(s)
Transducción de Señal , Factor de Crecimiento Transformador beta , Animales , Carcinogénesis , Transformación Celular Neoplásica , Ratones
17.
Science ; 367(6476): 405-411, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31974247

RESUMEN

Single-cell RNA sequencing (scRNA-seq) is a powerful approach for reconstructing cellular differentiation trajectories. However, inferring both the state and direction of differentiation is challenging. Here, we demonstrate a simple, yet robust, determinant of developmental potential-the number of expressed genes per cell-and leverage this measure of transcriptional diversity to develop a computational framework (CytoTRACE) for predicting differentiation states from scRNA-seq data. When applied to diverse tissue types and organisms, CytoTRACE outperformed previous methods and nearly 19,000 annotated gene sets for resolving 52 experimentally determined developmental trajectories. Additionally, it facilitated the identification of quiescent stem cells and revealed genes that contribute to breast tumorigenesis. This study thus establishes a key RNA-based feature of developmental potential and a platform for delineation of cellular hierarchies.


Asunto(s)
Diferenciación Celular/genética , Neoplasias/genética , ARN Citoplasmático Pequeño/genética , RNA-Seq/métodos , Análisis de la Célula Individual/métodos , Transcripción Genética , Animales , Secuencia de Bases , Variación Genética , Humanos , Ratones
18.
Nat Commun ; 11(1): 4061, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792541

RESUMEN

Adhesions are fibrotic scars that form between abdominal organs following surgery or infection, and may cause bowel obstruction, chronic pain, or infertility. Our understanding of adhesion biology is limited, which explains the paucity of anti-adhesion treatments. Here we present a systematic analysis of mouse and human adhesion tissues. First, we show that adhesions derive primarily from the visceral peritoneum, consistent with our clinical experience that adhesions form primarily following laparotomy rather than laparoscopy. Second, adhesions are formed by poly-clonal proliferating tissue-resident fibroblasts. Third, using single cell RNA-sequencing, we identify heterogeneity among adhesion fibroblasts, which is more pronounced at early timepoints. Fourth, JUN promotes adhesion formation and results in upregulation of PDGFRA expression. With JUN suppression, adhesion formation is diminished. Our findings support JUN as a therapeutic target to prevent adhesions. An anti-JUN therapy that could be applied intra-operatively to prevent adhesion formation could dramatically improve the lives of surgical patients.


Asunto(s)
Adherencias Tisulares/metabolismo , Adherencias Tisulares/patología , Animales , Benzofenonas/farmacología , Sistemas CRISPR-Cas , Células Cultivadas , Doxiciclina/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/patología , Humanos , Inmunohistoquímica , Isoxazoles/farmacología , Liposomas/metabolismo , Ratones , Células 3T3 NIH , Parabiosis , ARN Mensajero/metabolismo , Tamoxifeno/farmacología
19.
Nat Biotechnol ; 37(5): 531-539, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30886438

RESUMEN

Endogenous biomarkers remain at the forefront of early disease detection efforts, but many lack the sensitivities and specificities necessary to influence disease management. Here, we describe a cell-based in vivo sensor for highly sensitive early cancer detection. We engineer macrophages to produce a synthetic reporter on adopting an M2 tumor-associated metabolic profile by coupling luciferase expression to activation of the arginase-1 promoter. After adoptive transfer in colorectal and breast mouse tumor models, the engineered macrophages migrated to the tumors and activated arginase-1 so that they could be detected by bioluminescence imaging and luciferase measured in the blood. The macrophage sensor detected tumors as small as 25-50 mm3 by blood luciferase measurements, even in the presence of concomitant inflammation, and was more sensitive than clinically used protein and nucleic acid cancer biomarkers. Macrophage sensors also effectively tracked the immunological response in muscle and lung models of inflammation, suggesting the potential utility of this approach in disease states other than cancer.


Asunto(s)
Arginasa/sangre , Detección Precoz del Cáncer , Macrófagos/inmunología , Neoplasias/sangre , Animales , Arginasa/genética , Arginasa/inmunología , Biomarcadores de Tumor/sangre , Ingeniería Celular , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Luciferasas/sangre , Luciferasas/genética , Luciferasas/inmunología , Ratones , Neoplasias/inmunología , Neoplasias/patología
20.
Antioxid Redox Signal ; 29(2): 191-204, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29113449

RESUMEN

SIGNIFICANCE: Hematopoietic stem cells (HSCs) can sustain the production of blood throughout one's lifetime. However, for proper self-renewal of its own population and differentiation to blood, the HSC requires a specialized microenvironment called the "niche." Recent Advances: Recent studies using novel mouse models have shed new light on the cellular architecture and function of the HSC niche. Here, we review the different cells that constitute the HSC niche and the molecular mechanisms that underlie HSC and niche interaction. We discuss the evidence and potential features that distinguish the HSC niche from other microenvironments in the bone marrow. The relevance of the niche in malignant transformation of the HSCs and harboring cancer metastasis to the bone is also outlined. In addition, we address how the niche may regulate reactive oxygen species levels surrounding the HSCs. Critical Issues and Future Directions: We propose future directions and remaining challenges in investigating the niche of HSCs. We discuss how a better understanding of the HSC niche may help in restoring an aged hematopoietic system, fighting against malignancies, and transplanting purified HSCs safely and effectively into patients. Antioxid. Redox Signal. 00, 000-000.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/fisiología , Nicho de Células Madre , Animales , Células de la Médula Ósea/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia/etiología , Células Madre Mesenquimatosas , Ratones , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA