Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 184(12): 3109-3124.e22, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34004145

RESUMEN

Glycans modify lipids and proteins to mediate inter- and intramolecular interactions across all domains of life. RNA is not thought to be a major target of glycosylation. Here, we challenge this view with evidence that mammals use RNA as a third scaffold for glycosylation. Using a battery of chemical and biochemical approaches, we found that conserved small noncoding RNAs bear sialylated glycans. These "glycoRNAs" were present in multiple cell types and mammalian species, in cultured cells, and in vivo. GlycoRNA assembly depends on canonical N-glycan biosynthetic machinery and results in structures enriched in sialic acid and fucose. Analysis of living cells revealed that the majority of glycoRNAs were present on the cell surface and can interact with anti-dsRNA antibodies and members of the Siglec receptor family. Collectively, these findings suggest the existence of a direct interface between RNA biology and glycobiology, and an expanded role for RNA in extracellular biology.


Asunto(s)
Membrana Celular/metabolismo , Polisacáridos/metabolismo , ARN/metabolismo , Animales , Anticuerpos/metabolismo , Secuencia de Bases , Vías Biosintéticas , Línea Celular , Supervivencia Celular , Humanos , Espectrometría de Masas , Ácido N-Acetilneuramínico/metabolismo , Poliadenilación , Polisacáridos/química , ARN/química , ARN/genética , ARN no Traducido/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Coloración y Etiquetado
2.
Cell ; 182(2): 497-514.e22, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32579974

RESUMEN

To define the cellular composition and architecture of cutaneous squamous cell carcinoma (cSCC), we combined single-cell RNA sequencing with spatial transcriptomics and multiplexed ion beam imaging from a series of human cSCCs and matched normal skin. cSCC exhibited four tumor subpopulations, three recapitulating normal epidermal states, and a tumor-specific keratinocyte (TSK) population unique to cancer, which localized to a fibrovascular niche. Integration of single-cell and spatial data mapped ligand-receptor networks to specific cell types, revealing TSK cells as a hub for intercellular communication. Multiple features of potential immunosuppression were observed, including T regulatory cell (Treg) co-localization with CD8 T cells in compartmentalized tumor stroma. Finally, single-cell characterization of human tumor xenografts and in vivo CRISPR screens identified essential roles for specific tumor subpopulation-enriched gene networks in tumorigenesis. These data define cSCC tumor and stromal cell subpopulations, the spatial niches where they interact, and the communicating gene networks that they engage in cancer.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Genómica/métodos , Neoplasias Cutáneas/metabolismo , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , RNA-Seq , Análisis de la Célula Individual , Piel/metabolismo , Neoplasias Cutáneas/patología , Transcriptoma , Trasplante Heterólogo
3.
Cell ; 166(2): 451-467, 2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-27419872

RESUMEN

Stem-cell differentiation to desired lineages requires navigating alternating developmental paths that often lead to unwanted cell types. Hence, comprehensive developmental roadmaps are crucial to channel stem-cell differentiation toward desired fates. To this end, here, we map bifurcating lineage choices leading from pluripotency to 12 human mesodermal lineages, including bone, muscle, and heart. We defined the extrinsic signals controlling each binary lineage decision, enabling us to logically block differentiation toward unwanted fates and rapidly steer pluripotent stem cells toward 80%-99% pure human mesodermal lineages at most branchpoints. This strategy enabled the generation of human bone and heart progenitors that could engraft in respective in vivo models. Mapping stepwise chromatin and single-cell gene expression changes in mesoderm development uncovered somite segmentation, a previously unobservable human embryonic event transiently marked by HOPX expression. Collectively, this roadmap enables navigation of mesodermal development to produce transplantable human tissue progenitors and uncover developmental processes. VIDEO ABSTRACT.


Asunto(s)
Mesodermo/citología , Transducción de Señal , Proteínas Morfogenéticas Óseas/metabolismo , Huesos/citología , Huesos/metabolismo , Corazón/crecimiento & desarrollo , Proteínas de Homeodominio/metabolismo , Humanos , Mesodermo/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo , Línea Primitiva/citología , Línea Primitiva/metabolismo , Análisis de la Célula Individual , Somitos/metabolismo , Células Madre , Proteínas Supresoras de Tumor/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo
4.
Nat Immunol ; 19(1): 76-84, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29180808

RESUMEN

Exciting progress in the field of cancer immunotherapy has renewed the urgency of the need for basic studies of immunoregulation in both adaptive cell lineages and innate cell lineages. Here we found a central role for major histocompatibility complex (MHC) class I in controlling the phagocytic function of macrophages. Our results demonstrated that expression of the common MHC class I component ß2-microglobulin (ß2M) by cancer cells directly protected them from phagocytosis. We further showed that this protection was mediated by the inhibitory receptor LILRB1, whose expression was upregulated on the surface of macrophages, including tumor-associated macrophages. Disruption of either MHC class I or LILRB1 potentiated phagocytosis of tumor cells both in vitro and in vivo, which defines the MHC class I-LILRB1 signaling axis as an important regulator of the effector function of innate immune cells, a potential biomarker for therapeutic response to agents directed against the signal-regulatory protein CD47 and a potential target of anti-cancer immunotherapy.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Macrófagos/inmunología , Neoplasias/inmunología , Fagocitosis/inmunología , Animales , Línea Celular Tumoral , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoterapia/métodos , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia
6.
Nature ; 571(7764): 205-210, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31270459

RESUMEN

The mammalian brain contains neurogenic niches that comprise neural stem cells and other cell types. Neurogenic niches become less functional with age, but how they change during ageing remains unclear. Here we perform single-cell RNA sequencing of young and old neurogenic niches in mice. The analysis of 14,685 single-cell transcriptomes reveals a decrease in activated neural stem cells, changes in endothelial cells and microglia, and an infiltration of T cells in old neurogenic niches. T cells in old brains are clonally expanded and are generally distinct from those in old blood, which suggests that they may experience specific antigens. T cells in old brains also express interferon-γ, and the subset of neural stem cells that has a high interferon response shows decreased proliferation in vivo. We find that T cells can inhibit the proliferation of neural stem cells in co-cultures and in vivo, in part by secreting interferon-γ. Our study reveals an interaction between T cells and neural stem cells in old brains, opening potential avenues through which to counteract age-related decline in brain function.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/citología , Movimiento Celular , Células-Madre Neurales/citología , Neurogénesis , Análisis de la Célula Individual , Nicho de Células Madre/fisiología , Linfocitos T/citología , Animales , Sangre , Proliferación Celular , Células Clonales/citología , Técnicas de Cocultivo , Células Endoteliales/citología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Análisis de Secuencia de ARN , Transducción de Señal , Linfocitos T/metabolismo , Transcriptoma/genética
7.
Nature ; 545(7655): 495-499, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28514441

RESUMEN

Programmed cell death protein 1 (PD-1) is an immune checkpoint receptor that is upregulated on activated T cells for the induction of immune tolerance. Tumour cells frequently overexpress the ligand for PD-1, programmed cell death ligand 1 (PD-L1), facilitating their escape from the immune system. Monoclonal antibodies that block the interaction between PD-1 and PD-L1, by binding to either the ligand or receptor, have shown notable clinical efficacy in patients with a variety of cancers, including melanoma, colorectal cancer, non-small-cell lung cancer and Hodgkin's lymphoma. Although it is well established that PD-1-PD-L1 blockade activates T cells, little is known about the role that this pathway may have in tumour-associated macrophages (TAMs). Here we show that both mouse and human TAMs express PD-1. TAM PD-1 expression increases over time in mouse models of cancer and with increasing disease stage in primary human cancers. TAM PD-1 expression correlates negatively with phagocytic potency against tumour cells, and blockade of PD-1-PD-L1 in vivo increases macrophage phagocytosis, reduces tumour growth and lengthens the survival of mice in mouse models of cancer in a macrophage-dependent fashion. This suggests that PD-1-PD-L1 therapies may also function through a direct effect on macrophages, with substantial implications for the treatment of cancer with these agents.


Asunto(s)
Neoplasias del Colon/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Fagocitosis , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Estadificación de Neoplasias , Fagocitosis/efectos de los fármacos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
8.
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
9.
Proc Natl Acad Sci U S A ; 114(49): E10578-E10585, 2017 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-29158380

RESUMEN

Cancer immunotherapy has emerged as a promising therapeutic intervention. However, complete and durable responses are only seen in a fraction of patients who have cancer. A key factor that limits therapeutic success is the infiltration of tumors by cells of the myeloid lineage. The inhibitory receptor signal regulatory protein-α (SIRPα) is a myeloid-specific immune checkpoint that engages the "don't eat me" signal CD47 expressed on tumors and normal tissues. We therefore developed the monoclonal antibody KWAR23, which binds human SIRPα with high affinity and disrupts its binding to CD47. Administered by itself, KWAR23 is inert, but given in combination with tumor-opsonizing monoclonal antibodies, KWAR23 greatly augments myeloid cell-dependent killing of a collection of hematopoietic and nonhematopoietic human tumor-derived cell lines. Following KWAR23 antibody treatment in a human SIRPA knockin mouse model, both neutrophils and macrophages infiltrate a human Burkitt's lymphoma xenograft and inhibit tumor growth, generating complete responses in the majority of treated animals. We further demonstrate that a bispecific anti-CD70/SIRPα antibody outperforms individually delivered antibodies in specific types of cancers. These studies demonstrate that SIRPα blockade induces potent antitumor activity by targeting multiple myeloid cell subsets that frequently infiltrate tumors. Thus, KWAR23 represents a promising candidate for combination therapy.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/farmacología , Antígenos de Diferenciación/inmunología , Linfoma de Burkitt/terapia , Fagocitosis/efectos de los fármacos , Receptores Inmunológicos/inmunología , Animales , Antígenos de Diferenciación/genética , Linfoma de Burkitt/genética , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Ligando CD27/genética , Ligando CD27/inmunología , Antígeno CD47/genética , Antígeno CD47/inmunología , Línea Celular Tumoral , Terapia Combinada/métodos , Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Inmunoterapia/métodos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Unión Proteica , Receptores Inmunológicos/genética , Transgenes , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Proc Natl Acad Sci U S A ; 113(16): 4464-9, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27035983

RESUMEN

Pancreatic neuroendocrine tumors (PanNETs) are a type of pancreatic cancer with limited therapeutic options. Consequently, most patients with advanced disease die from tumor progression. Current evidence indicates that a subset of cancer cells is responsible for tumor development, metastasis, and recurrence, and targeting these tumor-initiating cells is necessary to eradicate tumors. However, tumor-initiating cells and the biological processes that promote pathogenesis remain largely uncharacterized in PanNETs. Here we profile primary and metastatic tumors from an index patient and demonstrate that MET proto-oncogene activation is important for tumor growth in PanNET xenograft models. We identify a highly tumorigenic cell population within several independent surgically acquired PanNETs characterized by increased cell-surface protein CD90 expression and aldehyde dehydrogenase A1 (ALDHA1) activity, and provide in vitro and in vivo evidence for their stem-like properties. We performed proteomic profiling of 332 antigens in two cell lines and four primary tumors, and showed that CD47, a cell-surface protein that acts as a "don't eat me" signal co-opted by cancers to evade innate immune surveillance, is ubiquitously expressed. Moreover, CD47 coexpresses with MET and is enriched in CD90(hi)cells. Furthermore, blocking CD47 signaling promotes engulfment of tumor cells by macrophages in vitro and inhibits xenograft tumor growth, prevents metastases, and prolongs survival in vivo.


Asunto(s)
Tumores Neuroendocrinos , Neoplasias Pancreáticas , Escape del Tumor , Familia de Aldehído Deshidrogenasa 1 , Animales , Antígeno CD47/inmunología , Femenino , Humanos , Isoenzimas/inmunología , Masculino , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia , Proteínas de Neoplasias/inmunología , Tumores Neuroendocrinos/inmunología , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/terapia , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Proto-Oncogenes Mas , Retinal-Deshidrogenasa/inmunología , Antígenos Thy-1/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
bioRxiv ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38559279

RESUMEN

Glycans modify protein, lipid, and even RNA molecules to form the regulatory outer coat on cells called the glycocalyx. The changes in glycosylation have been linked to the initiation and progression of many diseases. Thus, while the significance of glycosylation is well established, a lack of accessible methods to characterize glycans has hindered the ability to understand their biological functions. Mass spectrometry (MS)-based methods have generally been at the core of most glycan profiling efforts; however, modern data-independent acquisition (DIA), which could increase sensitivity and simplify workflows, has not been benchmarked for analyzing glycans. Herein, we developed a DIA-based glycomic workflow, termed GlycanDIA, to identify and quantify glycans with high sensitivity and accuracy. The GlycanDIA workflow combined higher energy collisional dissociation (HCD)-MS/MS and staggered windows for glycomic analysis, which facilitates the sensitivity in identification and the accuracy in quantification compared to conventional data-dependent acquisition (DDA)-based glycomics. To facilitate its use, we also developed a generic search engine, GlycanDIA Finder, incorporating an iterative decoy searching for confident glycan identification and quantification from DIA data. The results showed that GlycanDIA can distinguish glycan composition and isomers from N-glycans, O-glycans, and human milk oligosaccharides (HMOs), while it also reveals information on low-abundant modified glycans. With the improved sensitivity, we performed experiments to profile N-glycans from RNA samples, which have been underrepresented due to their low abundance. Using this integrative workflow to unravel the N-glycan profile in cellular and tissue glycoRNA samples, we found that RNA-glycans have specific forms as compared to protein-glycans and are also tissue-specific differences, suggesting distinct functions in biological processes. Overall, GlycanDIA can provide comprehensive information for glycan identification and quantification, enabling researchers to obtain in-depth and refined details on the biological roles of glycosylation.

12.
Nat Biotechnol ; 42(4): 597-607, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37537499

RESUMEN

Targeted protein degradation is an emerging strategy for the elimination of classically undruggable proteins. Here, to expand the landscape of targetable substrates, we designed degraders that achieve substrate selectivity via recognition of a discrete peptide and glycan motif and achieve cell-type selectivity via antigen-driven cell-surface binding. We applied this approach to mucins, O-glycosylated proteins that drive cancer progression through biophysical and immunological mechanisms. Engineering of a bacterial mucin-selective protease yielded a variant for fusion to a cancer antigen-binding nanobody. The resulting conjugate selectively degraded mucins on cancer cells, promoted cell death in culture models of mucin-driven growth and survival, and reduced tumor growth in mouse models of breast cancer progression. This work establishes a blueprint for the development of biologics that degrade specific protein glycoforms on target cells.


Asunto(s)
Mucinas , Neoplasias , Animales , Ratones , Mucinas/metabolismo , Péptido Hidrolasas/metabolismo , Proteolisis
13.
Nat Aging ; 3(1): 121-137, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-37118510

RESUMEN

The diversity of cell types is a challenge for quantifying aging and its reversal. Here we develop 'aging clocks' based on single-cell transcriptomics to characterize cell-type-specific aging and rejuvenation. We generated single-cell transcriptomes from the subventricular zone neurogenic region of 28 mice, tiling ages from young to old. We trained single-cell-based regression models to predict chronological age and biological age (neural stem cell proliferation capacity). These aging clocks are generalizable to independent cohorts of mice, other regions of the brains, and other species. To determine if these aging clocks could quantify transcriptomic rejuvenation, we generated single-cell transcriptomic datasets of neurogenic regions for two interventions-heterochronic parabiosis and exercise. Aging clocks revealed that heterochronic parabiosis and exercise reverse transcriptomic aging in neurogenic regions, but in different ways. This study represents the first development of high-resolution aging clocks from single-cell transcriptomic data and demonstrates their application to quantify transcriptomic rejuvenation.


Asunto(s)
Envejecimiento , Rejuvenecimiento , Ratones , Animales , Envejecimiento/genética , Senescencia Celular , Encéfalo , Neurogénesis
14.
Blood Adv ; 3(18): 2713-2721, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31519647

RESUMEN

Peritoneal adhesions are pathological fibroses that ensnare organs after abdominal surgery. This dense connective tissue can cause small bowel obstruction, female infertility, and chronic abdominal pain. The pathogenesis of adhesions is a fibrotic response to tissue damage coordinated between mesothelial cells, fibroblasts, and immune cells. We have previously demonstrated that peritoneal adhesions are a consequence of mechanical injury to the mesothelial layer sustained during surgery. Neutrophils are among the first leukocytes involved in the early response to tissue damage. Here, we show that when subjected to mechanical stress, activated mesothelial cells directly recruit neutrophils and monocytes through upregulation of chemokines such as CXCL1 and monocyte chemoattractant protein 1 (MCP-1). We find that neutrophils within the adhesion sites undergo cell death and form neutrophil extracellular traps (NETosis) that contribute to pathogenesis. Conversely, tissue-resident macrophages were profoundly depleted throughout the disease time course. We show that this is distinct from traditional inflammatory kinetics such as after sham surgery or chemically induced peritonitis, and suggest that adhesions result from a primary difference in inflammatory kinetics. We find that transient depletion of circulating neutrophils significantly decreases adhesion burden, and further recruitment of monocytes with thioglycolate or MCP-1 also improves outcomes. Our findings suggest that the combination of neutrophil depletion and monocyte recruitment is sufficient to prevent adhesion formation, thus providing insight for potential clinical interventions.


Asunto(s)
Monocitos/metabolismo , Neutrófilos/metabolismo , Adherencias Tisulares/metabolismo , Animales , Femenino , Humanos , Ratones
15.
Cell Stem Cell ; 25(2): 185-192.e3, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31204177

RESUMEN

Hematopoietic cell transplantation can correct hematological and immunological disorders by replacing a diseased blood system with a healthy one, but this currently requires depleting a patient's existing hematopoietic system with toxic and non-specific chemotherapy, radiation, or both. Here we report an antibody-based conditioning protocol with reduced toxicity and enhanced specificity for robust hematopoietic stem cell (HSC) transplantation and engraftment in recipient mice. Host pre-treatment with six monoclonal antibodies targeting CD47, T cells, NK cells, and HSCs followed by donor HSC transplantation enabled stable hematopoietic system reconstitution in recipients with mismatches at half (haploidentical) or all major histocompatibility complex (MHC) genes. This approach allowed tolerance to heart tissue from HSC donor strains in haploidentical recipients, showing potential applications for solid organ transplantation without immune suppression. Fully mismatched chimeric mice developed antibody responses to nominal antigens, showing preserved functional immunity. These findings suggest approaches for transplanting immunologically mismatched HSCs and solid organs with limited toxicity.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Miocardio/inmunología , Acondicionamiento Pretrasplante/métodos , Aloinjertos/inmunología , Animales , Anticuerpos Monoclonales , Células Cultivadas , Antígenos HLA/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Trasplante de Órganos , Quimera por Radiación , Tolerancia al Trasplante , Trasplante Haploidéntico , Trasplante Homólogo
16.
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
17.
Bone ; 108: 145-155, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29305998

RESUMEN

Mechanical loading is an important aspect of post-surgical fracture care. The timing of load application relative to the injury event may differentially regulate repair depending on the stage of healing. Here, we used a novel mechanobiological model of cortical defect repair that offers several advantages including its technical simplicity and spatially confined repair program, making effects of both physical and biological interventions more easily assessed. Using this model, we showed that daily loading (5N peak load, 2Hz, 60 cycles, 4 consecutive days) during hematoma consolidation and inflammation disrupted the injury site and activated cartilage formation on the periosteal surface adjacent to the defect. We also showed that daily loading during the matrix deposition phase enhanced both bone and cartilage formation at the defect site, while loading during the remodeling phase resulted in an enlarged woven bone regenerate. All loading regimens resulted in abundant cellular proliferation throughout the regenerate and fibrous tissue formation directly above the defect demonstrating that all phases of cortical defect healing are sensitive to physical stimulation. Stress was concentrated at the edges of the defect during exogenous loading, and finite element (FE)-modeled longitudinal strain (εzz) values along the anterior and posterior borders of the defect (~2200µÎµ) was an order of magnitude larger than strain values on the proximal and distal borders (~50-100µÎµ). It is concluded that loading during the early stages of repair may impede stabilization of the injury site important for early bone matrix deposition, whereas loading while matrix deposition and remodeling are ongoing may enhance stabilization through the formation of additional cartilage and bone.


Asunto(s)
Hueso Cortical/patología , Hueso Cortical/fisiopatología , Modelos Biológicos , Cicatrización de Heridas , Animales , Fenómenos Biomecánicos , Matriz Ósea/metabolismo , Proliferación Celular , Condrogénesis , Fuerza Compresiva , Femenino , Hematoma/patología , Inflamación/patología , Ratones Endogámicos C57BL , Tamaño de los Órganos , Osteoclastos/metabolismo , Reproducibilidad de los Resultados , Estrés Mecánico , Tibia/patología , Soporte de Peso/fisiología
18.
Science ; 359(6379): 1037-1042, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29496879

RESUMEN

Interleukin-2 (IL-2) is a cytokine required for effector T cell expansion, survival, and function, especially for engineered T cells in adoptive cell immunotherapy, but its pleiotropy leads to simultaneous stimulation and suppression of immune responses as well as systemic toxicity, limiting its therapeutic use. We engineered IL-2 cytokine-receptor orthogonal (ortho) pairs that interact with one another, transmitting native IL-2 signals, but do not interact with their natural cytokine and receptor counterparts. Introduction of orthoIL-2Rß into T cells enabled the selective cellular targeting of orthoIL-2 to engineered CD4+ and CD8+ T cells in vitro and in vivo, with limited off-target effects and negligible toxicity. OrthoIL-2 pairs were efficacious in a preclinical mouse cancer model of adoptive cell therapy and may therefore represent a synthetic approach to achieving selective potentiation of engineered cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Ingeniería Celular/métodos , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores de Interleucina-2/inmunología , Animales , Células HEK293 , Humanos , Melanoma Experimental , Ratones , Receptores de Interleucina-2/genética
19.
Artículo en Inglés | MEDLINE | ID: mdl-28416577

RESUMEN

A stem cell is broadly defined as a cell that retains the capacity to self-renew, a feature that confers the ability to continuously make identical daughter cells or additional cells that will differentiate into downstream progeny. This highly regulated genetic program to retain "stemness" is under active investigation. Research in our laboratory has explored similarities and differences in embryonic, tissue-specific, and neoplastic stem cells and their terminally differentiated counterparts. In this review, we will focus on the contributions of our laboratory, in particular on the studies that identified the mouse hematopoietic stem cell (HSC) and the human leukemic stem cell. These studies have led to significant improvements in both preclinical and clinical research, including improved clinical bone marrow transplantation protocols, isolation of nonleukemic HSCs, a cancer immunotherapy currently in clinical trials, and development of a HSC reporter mouse. These studies and the current follow-up research by us and others will continue to identify the properties, function, and regulation of both normal and neoplastic stem cells.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Transformación Celular Neoplásica/metabolismo , Células Madre Hematopoyéticas/citología , Animales , Trasplante de Médula Ósea/métodos , Humanos
20.
Science ; 333(6045): 1026-30, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21852501

RESUMEN

Aneuploidy decreases cellular fitness, yet it is also associated with cancer, a disease of enhanced proliferative capacity. To investigate one mechanism by which aneuploidy could contribute to tumorigenesis, we examined the effects of aneuploidy on genomic stability. We analyzed 13 budding yeast strains that carry extra copies of single chromosomes and found that all aneuploid strains exhibited one or more forms of genomic instability. Most strains displayed increased chromosome loss and mitotic recombination, as well as defective DNA damage repair. Aneuploid fission yeast strains also exhibited defects in mitotic recombination. Aneuploidy-induced genomic instability could facilitate the development of genetic alterations that drive malignant growth in cancer.


Asunto(s)
Aneuploidia , Daño del ADN , Reparación del ADN , Genoma Fúngico , Inestabilidad Genómica , Recombinación Genética , Saccharomyces cerevisiae/genética , Segregación Cromosómica , Cromosomas Fúngicos/genética , Replicación del ADN , ADN de Hongos/genética , ADN de Hongos/metabolismo , Mutagénesis , Mutación , Neoplasias/genética , Fenotipo , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteínas de Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA