Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(11): eadh9547, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38489372

RESUMEN

Solid tumors, especially those with aberrant MYCN activation, often harbor an immunosuppressive microenvironment to fuel malignant growth and trigger treatment resistance. Despite this knowledge, there are no effective strategies to tackle this problem. We found that chemokine-like factor (CKLF) is highly expressed by various solid tumor cells and transcriptionally up-regulated by MYCN. Using the MYCN-driven high-risk neuroblastoma as a model system, we demonstrated that as early as the premalignant stage, tumor cells secrete CKLF to attract CCR4-expressing CD4+ cells, inducing immunosuppression and tumor aggression. Genetic depletion of CD4+ T regulatory cells abolishes the immunorestrictive and protumorigenic effects of CKLF. Our work supports that disrupting CKLF-mediated cross-talk between tumor and CD4+ suppressor cells represents a promising immunotherapeutic approach to battling MYCN-driven tumors.


Asunto(s)
Quimiocinas , Proteínas con Dominio MARVEL , Proteína Proto-Oncogénica N-Myc , Neuroblastoma , Humanos , Línea Celular Tumoral , Quimiocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas con Dominio MARVEL/metabolismo , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuroblastoma/terapia , Microambiente Tumoral
2.
medRxiv ; 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-38106222

RESUMEN

Polycystic kidney disease (PKD) is an important cause of end stage renal disease, but treatment options are limited. While later stages of the disease have been extensively studied, mechanisms driving the initial conversion of renal tubules into cysts are not understood. To identify factors that promote the initiation of cysts we deleted polycystin-2 ( Pkd2 ) in mice and surveyed transcriptional changes before and immediately after cysts developed. We identified 74 genes which we term cyst initiation candidates (CICs). To identify conserved changes with relevance to human disease we compared these murine CICs to single cell transcriptomic data derived from patients with PKD and from healthy controls. Tumor-associated calcium signal transducer 2 ( Tacstd2 ) stood out as an epithelial-expressed gene whose levels were elevated prior to cystic transformation and further increased with disease progression. Human tissue biopsies and organoids show that TACSTD2 protein is low in normal kidney cells but is elevated in cyst lining cells. While TACSTD2 has not been studied in PKD, it has been studied in cancer where it is highly expressed in solid tumors while showing minimal expression in normal tissue. This property is being exploited by antibody drug conjugates that target TACSTD2 for the delivery of cytotoxic drugs. Our finding that Tacstd2 is highly expressed in cysts, but not normal tissue, suggests that it should be explored as a candidate for drug development in PKD. More immediately, our work suggests that PKD patients undergoing TACSTD2 treatment for cancer should be monitored for kidney effects. One Sentence Summary: The oncogene, tumor-associated calcium signal transducer 2 (Tacstd2) mRNA increased in abundance shortly after Pkd2 loss and may be a driver of cyst initiation in polycystic kidney disease.

3.
Cancer Discov ; 13(10): 2128-2130, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37794841

RESUMEN

SUMMARY: GABA signaling by melanoma cells was found by Tagore and colleagues to trigger keratinocyte-driven growth of melanomas. This study reveals new roles for nonneuronal signaling by a neurotransmitter in regulating tumor initiation and outgrowth. See related article by Tagore et al., p. 2270 (4).


Asunto(s)
Melanoma , Humanos , Melanoma/genética , Melanoma/patología , Queratinocitos , Transformación Celular Neoplásica , Transducción de Señal , Ácido gamma-Aminobutírico
4.
Elife ; 122023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-37021774

RESUMEN

Tissue-resident stem and progenitor cells are present in many adult organs, where they are important for organ homeostasis and repair in response to injury. However, the signals that activate these cells and the mechanisms governing how these cells renew or differentiate are highly context-dependent and incompletely understood, particularly in non-hematopoietic tissues. In the skin, melanocyte stem and progenitor cells are responsible for replenishing mature pigmented melanocytes. In mammals, these cells reside in the hair follicle bulge and bulb niches where they are activated during homeostatic hair follicle turnover and following melanocyte destruction, as occurs in vitiligo and other skin hypopigmentation disorders. Recently, we identified melanocyte progenitors in adult zebrafish skin. To elucidate mechanisms governing melanocyte progenitor renewal and differentiation we analyzed individual transcriptomes from thousands of melanocyte lineage cells during the regeneration process. We identified transcriptional signatures for progenitors, deciphered transcriptional changes and intermediate cell states during regeneration, and analyzed cell-cell signaling changes to discover mechanisms governing melanocyte regeneration. We identified KIT signaling via the RAS/MAPK pathway as a regulator of melanocyte progenitor direct differentiation and asymmetric division. Our findings show how activation of different subpopulations of mitfa-positive cells underlies cellular transitions required to properly reconstitute the melanocyte pigmentary system following injury.


Asunto(s)
Melanocitos , Pez Cebra , Animales , Pez Cebra/fisiología , Melanocitos/metabolismo , Piel , Células Madre/metabolismo , Folículo Piloso , Transducción de Señal , Diferenciación Celular , Mamíferos
5.
Nat Commun ; 13(1): 4109, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35840569

RESUMEN

Melanomas and other solid tumors commonly have increased ploidy, with near-tetraploid karyotypes being most frequently observed. Such karyotypes have been shown to arise through whole-genome doubling events that occur during early stages of tumor progression. The generation of tetraploid cells via whole-genome doubling is proposed to allow nascent tumor cells the ability to sample various pro-tumorigenic genomic configurations while avoiding the negative consequences that chromosomal gains or losses have in diploid cells. Whereas a high prevalence of whole-genome doubling events has been established, the means by which whole-genome doubling arises is unclear. Here, we find that BRAFV600E, the most common mutation in melanomas, can induce whole-genome doubling via cytokinesis failure in vitro and in a zebrafish melanoma model. Mechanistically, BRAFV600E causes decreased activation and localization of RhoA, a critical cytokinesis regulator. BRAFV600E activity during G1/S phases of the cell cycle is required to suppress cytokinesis. During G1/S, BRAFV600E activity causes inappropriate centriole amplification, which is linked in part to inhibition of RhoA and suppression of cytokinesis. Together these data suggest that common abnormalities of melanomas linked to tumorigenesis - amplified centrosomes and whole-genome doubling events - can be induced by oncogenic BRAF and other mutations that increase RAS/MAPK pathway activity.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf , Animales , Línea Celular Tumoral , Citocinesis/genética , Melanoma/genética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Tetraploidía , Pez Cebra/genética , Pez Cebra/metabolismo
6.
Nat Commun ; 13(1): 3732, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35768444

RESUMEN

Melanoma is commonly driven by activating mutations in the MAP kinase BRAF; however, oncogenic BRAF alone is insufficient to promote melanomagenesis. Instead, its expression induces a transient proliferative burst that ultimately ceases with the development of benign nevi comprised of growth-arrested melanocytes. The tumor suppressive mechanisms that restrain nevus melanocyte proliferation remain poorly understood. Here we utilize cell and murine models to demonstrate that oncogenic BRAF leads to activation of the Hippo tumor suppressor pathway, both in melanocytes in vitro and nevus melanocytes in vivo. Mechanistically, we show that oncogenic BRAF promotes both ERK-dependent alterations in the actin cytoskeleton and whole-genome doubling events, which independently reduce RhoA activity to promote Hippo activation. We also demonstrate that functional impairment of the Hippo pathway enables oncogenic BRAF-expressing melanocytes to bypass nevus formation and rapidly form melanomas. Our data reveal that the Hippo pathway enforces the stable arrest of nevus melanocytes and represents a critical barrier to melanoma development.


Asunto(s)
Melanoma , Nevo , Neoplasias Cutáneas , Animales , Melanocitos/metabolismo , Melanoma/patología , Ratones , Mutación , Nevo/genética , Nevo/metabolismo , Nevo/patología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/patología
7.
Cell Rep ; 39(12): 110995, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35732120

RESUMEN

Dysregulated cellular metabolism is a cancer hallmark for which few druggable oncoprotein targets have been identified. Increased fatty acid (FA) acquisition allows cancer cells to meet their heightened membrane biogenesis, bioenergy, and signaling needs. Excess FAs are toxic to non-transformed cells but surprisingly not to cancer cells. Molecules underlying this cancer adaptation may provide alternative drug targets. Here, we demonstrate that diacylglycerol O-acyltransferase 1 (DGAT1), an enzyme integral to triacylglyceride synthesis and lipid droplet formation, is frequently up-regulated in melanoma, allowing melanoma cells to tolerate excess FA. DGAT1 over-expression alone transforms p53-mutant zebrafish melanocytes and co-operates with oncogenic BRAF or NRAS for more rapid melanoma formation. Antagonism of DGAT1 induces oxidative stress in melanoma cells, which adapt by up-regulating cellular reactive oxygen species defenses. We show that inhibiting both DGAT1 and superoxide dismutase 1 profoundly suppress tumor growth through eliciting intolerable oxidative stress.


Asunto(s)
Diacilglicerol O-Acetiltransferasa , Melanoma , Animales , Diacilglicerol O-Acetiltransferasa/genética , Diacilglicerol O-Acetiltransferasa/metabolismo , Proteínas Oncogénicas/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno , Triglicéridos , Pez Cebra/metabolismo
8.
J Invest Dermatol ; 142(3 Pt A): 499-506.e1, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35184798

RESUMEN

Skin diseases affect nearly one third of the world's population. Disease types range from oncologic to inflammatory, and outcomes can be as severe as death and disfigurement. Although many skin diseases have been modeled in murine models, the advantages of zebrafish models have led to recent increasing use in modeling human disease. Their rapid development, comparable skin architecture, tractable genetics, unparalleled optical properties, and straightforward drug screens make them an excellent model to study skin disease. In this review, we discuss the attributes of the zebrafish model system as well as current zebrafish models for dermatologic diseases, including melanoma, squamous cell carcinoma, vitiligo, epidermal bullosa, psoriasis, and wounding.


Asunto(s)
Melanoma , Vitíligo , Animales , Modelos Animales de Enfermedad , Humanos , Melanoma/patología , Ratones , Proyectos de Investigación , Pez Cebra
9.
Dev Cell ; 56(20): 2783-2784, 2021 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-34699785

RESUMEN

In this issue of Developmental Cell, Campbell et al. (2021) show that melanoma cells with distinct invasive or proliferative gene signatures can form heterotypic clusters that extravasate collectively and readily seed the growth of metastatic lesions. These findings highlight interactions between heterogenous tumor cells as being critical for metastasis.


Asunto(s)
Melanoma , Recuento de Células , Movimiento Celular , Humanos
10.
Cancer Cell ; 39(5): 610-631, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33545064

RESUMEN

There is a lack of appropriate melanoma models that can be used to evaluate the efficacy of novel therapeutic modalities. Here, we discuss the current state of the art of melanoma models including genetically engineered mouse, patient-derived xenograft, zebrafish, and ex vivo and in vitro models. We also identify five major challenges that can be addressed using such models, including metastasis and tumor dormancy, drug resistance, the melanoma immune response, and the impact of aging and environmental exposures on melanoma progression and drug resistance. Additionally, we discuss the opportunity for building models for rare subtypes of melanomas, which represent an unmet critical need. Finally, we identify key recommendations for melanoma models that may improve accuracy of preclinical testing and predict efficacy in clinical trials, to help usher in the next generation of melanoma therapies.


Asunto(s)
Modelos Animales de Enfermedad , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Microambiente Tumoral/inmunología , Animales , Humanos , Inmunidad/inmunología , Inmunoterapia/métodos , Melanoma/patología , Neoplasias Cutáneas/patología
12.
Pigment Cell Melanoma Res ; 34(2): 280-287, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33283422

RESUMEN

Melanoma arises from the melanocyte lineage and is the most aggressive and lethal form of skin cancer. There are several genetic, genomic, and cellular changes associated with melanoma initiation. Here, we discuss these alterations and the melanoma cells of origin in which they are proposed to promote melanomagenesis.


Asunto(s)
Carcinogénesis/patología , Regulación Neoplásica de la Expresión Génica , Melanocitos/patología , Melanoma/patología , Neoplasias Cutáneas/patología , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Humanos , Melanocitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo
13.
Mol Carcinog ; 59(9): 1052-1063, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32562448

RESUMEN

Melanoma is an aggressive, deadly skin cancer derived from melanocytes, a neural crest cell derivative. Melanoma cells mirror the developmental program of neural crest cells in that they exhibit the same gene expression patterns and utilize similar cellular mechanisms, including increased cell proliferation, epithelial-mesenchymal transition, and migration. Here we studied the role of neural crest regulator PRDM1 in melanoma onset and progression. In development, Prdm1a functions to promote neural crest progenitor fate, and in melanoma, we found that PRDM1 has reduced copy number and is recurrently deleted in both zebrafish and humans. When examining expression of neural crest and melanocyte development genes, we show that sox10 progenitor expression is high in prdm1a-/- mutants, while more differentiated melanocyte markers are reduced, suggesting that normally Prdm1a is required for differentiation. Data mining of human melanoma datasets indicates that high PRDM1 expression in human melanoma is correlated with better patient survival and decreased PRDM1 expression is common in metastatic tumors. When one copy of prdm1a is lost in the zebrafish melanoma model Tg(mitfa:BRAFV600E );p53-/- ;prdm1a+/- , melanoma onset occurs more quickly, and the tumors that form have a larger area with increased expression of sox10. These data demonstrate a novel role for PRDM1 as a tumor suppressor in melanoma.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Melanocitos/patología , Melanoma/patología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Animales , Diferenciación Celular , Células Cultivadas , Progresión de la Enfermedad , Humanos , Melanocitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Pronóstico , Tasa de Supervivencia , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
14.
Cells ; 9(5)2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32455885

RESUMEN

Melanoma is the deadliest form of skin cancer and one of few cancers with a growing incidence. A thorough understanding of its pathogenesis is fundamental to developing new strategies to combat mortality and morbidity. Zebrafish-due in large part to their tractable genetics, conserved pathways, and optical properties-have emerged as an excellent system to model melanoma. Zebrafish have been used to study melanoma from a single tumor initiating cell, through metastasis, remission, and finally into relapse. In this review, we examine seminal zebrafish studies that have advanced our understanding of melanoma.


Asunto(s)
Melanoma/patología , Pez Cebra/fisiología , Animales , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Melanocitos/patología , Melanoma/genética , Cresta Neural/patología
15.
Elife ; 82019 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-31868592

RESUMEN

Preventing terminal differentiation is important in the development and progression of many cancers including melanoma. Recent identification of the BMP ligand GDF6 as a novel melanoma oncogene showed GDF6-activated BMP signaling suppresses differentiation of melanoma cells. Previous studies have identified roles for GDF6 orthologs during early embryonic and neural crest development, but have not identified direct regulation of melanocyte development by GDF6. Here, we investigate the BMP ligand gdf6a, a zebrafish ortholog of human GDF6, during the development of melanocytes from the neural crest. We establish that the loss of gdf6a or inhibition of BMP signaling during neural crest development disrupts normal pigment cell development, leading to an increase in the number of melanocytes and a corresponding decrease in iridophores, another neural crest-derived pigment cell type in zebrafish. This shift occurs as pigment cells arise from the neural crest and depends on mitfa, an ortholog of MITF, a key regulator of melanocyte development that is also targeted by oncogenic BMP signaling. Together, these results indicate that the oncogenic role ligand-dependent BMP signaling plays in suppressing differentiation in melanoma is a reiteration of its physiological roles during melanocyte development.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Factor 6 de Diferenciación de Crecimiento/genética , Melanocitos/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Proteínas de Pez Cebra/genética , Animales , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ligandos , Melanocitos/patología , Neoplasias/genética , Neoplasias/patología , Cresta Neural/crecimiento & desarrollo , Cresta Neural/metabolismo , Pigmentación/genética , Transducción de Señal/genética , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
16.
J Clin Invest ; 128(1): 294-308, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29202482

RESUMEN

Oncogenomic studies indicate that copy number variation (CNV) alters genes involved in tumor progression; however, identification of specific driver genes affected by CNV has been difficult, as these rearrangements are often contained in large chromosomal intervals among several bystander genes. Here, we addressed this problem and identified a CNV-targeted oncogene by performing comparative oncogenomics of human and zebrafish melanomas. We determined that the gene encoding growth differentiation factor 6 (GDF6), which is the ligand for the BMP family, is recurrently amplified and transcriptionally upregulated in melanoma. GDF6-induced BMP signaling maintained a trunk neural crest gene signature in melanomas. Additionally, GDF6 repressed the melanocyte differentiation gene MITF and the proapoptotic factor SOX9, thereby preventing differentiation, inhibiting cell death, and promoting tumor growth. GDF6 was specifically expressed in melanomas but not melanocytes. Moreover, GDF6 expression levels in melanomas were inversely correlated with patient survival. Our study has identified a fundamental role for GDF6 and BMP signaling in governing an embryonic cell gene signature to promote melanoma progression, thus providing potential opportunities for targeted therapy to treat GDF6-positive cancers.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Factor 6 de Diferenciación de Crecimiento/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Animales , Proteínas Morfogenéticas Óseas/genética , Línea Celular Tumoral , Femenino , Factor 6 de Diferenciación de Crecimiento/genética , Células HEK293 , Humanos , Ligandos , Melanoma/genética , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteínas de Neoplasias/genética
18.
Pigment Cell Melanoma Res ; 31(1): 115-119, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28646617

RESUMEN

Large-scale sequencing studies have revealed several genes that are recurrently mutated in melanomas. To annotate the melanoma genome, we have expressed tumor-associated variants of these genes in zebrafish and characterized their effects on melanocyte development and function. Here, we describe expression of tumor-associated variants of the recurrently mutated metabotropic glutamate receptor 3 (GRM3) gene. Unlike wild-type GRM3, tumor-associated GRM3 variants disrupted trafficking of melanosomes, causing their aggregation in the cell body. Melanosomes are trafficked in a cAMP-dependent manner, and drugs that directly or indirectly increased cAMP levels were able to suppress melanosome aggregation in mutant GRM3-expressing melanocytes. Our data show that oncogenic GRM3 variants dysregulate cAMP signaling, a heretofore unknown role for these oncogenes. cAMP signaling has been implicated in melanoma progression and drug resistance, and our data show that oncogenic properties of GRM3 could be mediated, at least in part, by alterations in cAMP signaling.


Asunto(s)
AMP Cíclico/metabolismo , Variación Genética , Melanocitos/patología , Melanoma/patología , Melanosomas/patología , Receptor del Glutamato Metabotropico 5/genética , Pez Cebra/metabolismo , Animales , Humanos , Melanocitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Melanosomas/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Transducción de Señal , Pez Cebra/crecimiento & desarrollo
19.
J Exp Med ; 214(12): 3519-3530, 2017 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-29066577

RESUMEN

Regulatory T (T reg) cells are a specialized sublineage of T lymphocytes that suppress autoreactive T cells. Functional studies of T reg cells in vitro have defined multiple suppression mechanisms, and studies of T reg-deficient humans and mice have made clear the important role that these cells play in preventing autoimmunity. However, many questions remain about how T reg cells act in vivo. Specifically, it is not clear which suppression mechanisms are most important, where T reg cells act, and how they get there. To begin to address these issues, we sought to identify T reg cells in zebrafish, a model system that provides unparalleled advantages in live-cell imaging and high-throughput genetic analyses. Using a FOXP3 orthologue as a marker, we identified CD4-enriched, mature T lymphocytes with properties of T reg cells. Zebrafish mutant for foxp3a displayed excess T lymphocytes, splenomegaly, and a profound inflammatory phenotype that was suppressed by genetic ablation of lymphocytes. This study identifies T reg-like cells in zebrafish, providing both a model to study the normal functions of these cells in vivo and mutants to explore the consequences of their loss.


Asunto(s)
Linfocitos T Reguladores/inmunología , Pez Cebra/inmunología , Animales , Secuencia de Bases , Enfermedad Crónica , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Hematopoyesis , Inflamación/patología , Linfocitos/metabolismo , Mutación/genética , Filogenia , Esplenomegalia/patología , Análisis de Supervivencia , Timocitos/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo
20.
J Exp Med ; 214(10): 2875-2887, 2017 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-28878000

RESUMEN

Recent advances in single-cell, transcriptomic profiling have provided unprecedented access to investigate cell heterogeneity during tissue and organ development. In this study, we used massively parallel, single-cell RNA sequencing to define cell heterogeneity within the zebrafish kidney marrow, constructing a comprehensive molecular atlas of definitive hematopoiesis and functionally distinct renal cells found in adult zebrafish. Because our method analyzed blood and kidney cells in an unbiased manner, our approach was useful in characterizing immune-cell deficiencies within DNA-protein kinase catalytic subunit (prkdc), interleukin-2 receptor γ a (il2rga), and double-homozygous-mutant fish, identifying blood cell losses in T, B, and natural killer cells within specific genetic mutants. Our analysis also uncovered novel cell types, including two classes of natural killer immune cells, classically defined and erythroid-primed hematopoietic stem and progenitor cells, mucin-secreting kidney cells, and kidney stem/progenitor cells. In total, our work provides the first, comprehensive, single-cell, transcriptomic analysis of kidney and marrow cells in the adult zebrafish.


Asunto(s)
Hematopoyesis Extramedular/genética , Riñón/citología , ARN/genética , Pez Cebra/anatomía & histología , Animales , Animales Modificados Genéticamente , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Perfilación de la Expresión Génica , Hematopoyesis Extramedular/fisiología , Células Madre Hematopoyéticas , Riñón/metabolismo , Análisis de Secuencia de ARN , Pez Cebra/genética , Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...