Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.046
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 187(12): 3072-3089.e20, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38781967

RESUMO

Tissue folds are structural motifs critical to organ function. In the intestine, bending of a flat epithelium into a periodic pattern of folds gives rise to villi, finger-like protrusions that enable nutrient absorption. However, the molecular and mechanical processes driving villus morphogenesis remain unclear. Here, we identify an active mechanical mechanism that simultaneously patterns and folds the intestinal epithelium to initiate villus formation. At the cellular level, we find that PDGFRA+ subepithelial mesenchymal cells generate myosin II-dependent forces sufficient to produce patterned curvature in neighboring tissue interfaces. This symmetry-breaking process requires altered cell and extracellular matrix interactions that are enabled by matrix metalloproteinase-mediated tissue fluidization. Computational models, together with in vitro and in vivo experiments, revealed that these cellular features manifest at the tissue level as differences in interfacial tensions that promote mesenchymal aggregation and interface bending through a process analogous to the active dewetting of a thin liquid film.


Assuntos
Matriz Extracelular , Mucosa Intestinal , Animais , Camundongos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/citologia , Matriz Extracelular/metabolismo , Miosina Tipo II/metabolismo , Mesoderma/metabolismo , Mesoderma/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Morfogênese , Metaloproteinases da Matriz/metabolismo
2.
Cell ; 186(4): 877-891.e14, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36708705

RESUMO

We introduce BacDrop, a highly scalable technology for bacterial single-cell RNA sequencing that has overcome many challenges hindering the development of scRNA-seq in bacteria. BacDrop can be applied to thousands to millions of cells from both gram-negative and gram-positive species. It features universal ribosomal RNA depletion and combinatorial barcodes that enable multiplexing and massively parallel sequencing. We applied BacDrop to study Klebsiella pneumoniae clinical isolates and to elucidate their heterogeneous responses to antibiotic stress. In an unperturbed population presumed to be homogeneous, we found within-population heterogeneity largely driven by the expression of mobile genetic elements that promote the evolution of antibiotic resistance. Under antibiotic perturbation, BacDrop revealed transcriptionally distinct subpopulations associated with different phenotypic outcomes including antibiotic persistence. BacDrop thus can capture cellular states that cannot be detected by bulk RNA-seq, which will unlock new microbiological insights into bacterial responses to perturbations and larger bacterial communities such as the microbiome.


Assuntos
Perfilação da Expressão Gênica , Análise da Expressão Gênica de Célula Única , Análise de Sequência de RNA , RNA-Seq , Bactérias/genética , Análise de Célula Única
3.
Cell ; 186(7): 1398-1416.e23, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36944331

RESUMO

CD3δ SCID is a devastating inborn error of immunity caused by mutations in CD3D, encoding the invariant CD3δ chain of the CD3/TCR complex necessary for normal thymopoiesis. We demonstrate an adenine base editing (ABE) strategy to restore CD3δ in autologous hematopoietic stem and progenitor cells (HSPCs). Delivery of mRNA encoding a laboratory-evolved ABE and guide RNA into a CD3δ SCID patient's HSPCs resulted in a 71.2% ± 7.85% (n = 3) correction of the pathogenic mutation. Edited HSPCs differentiated in artificial thymic organoids produced mature T cells exhibiting diverse TCR repertoires and TCR-dependent functions. Edited human HSPCs transplanted into immunodeficient mice showed 88% reversion of the CD3D defect in human CD34+ cells isolated from mouse bone marrow after 16 weeks, indicating correction of long-term repopulating HSCs. These findings demonstrate the preclinical efficacy of ABE in HSPCs for the treatment of CD3δ SCID, providing a foundation for the development of a one-time treatment for CD3δ SCID patients.


Assuntos
Imunodeficiência Combinada Severa , Linfócitos T , Humanos , Animais , Camundongos , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/terapia , Edição de Genes , Camundongos SCID , Complexo CD3 , Receptores de Antígenos de Linfócitos T/genética
4.
Cell ; 185(11): 1905-1923.e25, 2022 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-35523183

RESUMO

Tumor evolution is driven by the progressive acquisition of genetic and epigenetic alterations that enable uncontrolled growth and expansion to neighboring and distal tissues. The study of phylogenetic relationships between cancer cells provides key insights into these processes. Here, we introduced an evolving lineage-tracing system with a single-cell RNA-seq readout into a mouse model of Kras;Trp53(KP)-driven lung adenocarcinoma and tracked tumor evolution from single-transformed cells to metastatic tumors at unprecedented resolution. We found that the loss of the initial, stable alveolar-type2-like state was accompanied by a transient increase in plasticity. This was followed by the adoption of distinct transcriptional programs that enable rapid expansion and, ultimately, clonal sweep of stable subclones capable of metastasizing. Finally, tumors develop through stereotypical evolutionary trajectories, and perturbing additional tumor suppressors accelerates progression by creating novel trajectories. Our study elucidates the hierarchical nature of tumor evolution and, more broadly, enables in-depth studies of tumor progression.


Assuntos
Neoplasias , Animais , Genes ras , Camundongos , Neoplasias/genética , Filogenia , Sequenciamento do Exoma
5.
Nat Immunol ; 25(2): 330-342, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38172260

RESUMO

Antibody-secreting plasma cells (PCs) are generated in secondary lymphoid organs but are reported to reside in an emerging range of anatomical sites. Analysis of the transcriptome of different tissue-resident (Tr)PC populations revealed that they each have their own transcriptional signature indicative of functional adaptation to the host tissue environment. In contrast to expectation, all TrPCs were extremely long-lived, regardless of their organ of residence, with longevity influenced by intrinsic factors like the immunoglobulin isotype. Analysis at single-cell resolution revealed that the bone marrow is unique in housing a compendium of PCs generated all over the body that retain aspects of the transcriptional program indicative of their tissue of origin. This study reveals that extreme longevity is an intrinsic property of TrPCs whose transcriptome is imprinted by signals received both at the site of induction and within the tissue of residence.


Assuntos
Medula Óssea , Plasmócitos , Células da Medula Óssea
6.
Nat Immunol ; 25(6): 1033-1045, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38745085

RESUMO

The etiology and effect of age-related immune dysfunction in cancer is not completely understood. Here we show that limited priming of CD8+ T cells in the aged tumor microenvironment (TME) outweighs cell-intrinsic defects in limiting tumor control. Increased tumor growth in aging is associated with reduced CD8+ T cell infiltration and function. Transfer of T cells from young mice does not restore tumor control in aged mice owing to rapid induction of T cell dysfunction. Cell-extrinsic signals in the aged TME drive a tumor-infiltrating age-associated dysfunctional (TTAD) cell state that is functionally, transcriptionally and epigenetically distinct from canonical T cell exhaustion. Altered natural killer cell-dendritic cell-CD8+ T cell cross-talk in aged tumors impairs T cell priming by conventional type 1 dendritic cells and promotes TTAD cell formation. Aged mice are thereby unable to benefit from therapeutic tumor vaccination. Critically, myeloid-targeted therapy to reinvigorate conventional type 1 dendritic cells can improve tumor control and restore CD8+ T cell immunity in aging.


Assuntos
Envelhecimento , Linfócitos T CD8-Positivos , Células Dendríticas , Microambiente Tumoral , Animais , Microambiente Tumoral/imunologia , Linfócitos T CD8-Positivos/imunologia , Camundongos , Células Dendríticas/imunologia , Envelhecimento/imunologia , Camundongos Endogâmicos C57BL , Células Matadoras Naturais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Humanos , Neoplasias/imunologia , Linhagem Celular Tumoral , Feminino , Ativação Linfocitária/imunologia
7.
Nat Immunol ; 25(4): 644-658, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38503922

RESUMO

The organization of immune cells in human tumors is not well understood. Immunogenic tumors harbor spatially localized multicellular 'immunity hubs' defined by expression of the T cell-attracting chemokines CXCL10/CXCL11 and abundant T cells. Here, we examined immunity hubs in human pre-immunotherapy lung cancer specimens and found an association with beneficial response to PD-1 blockade. Critically, we discovered the stem-immunity hub, a subtype of immunity hub strongly associated with favorable PD-1-blockade outcome. This hub is distinct from mature tertiary lymphoid structures and is enriched for stem-like TCF7+PD-1+CD8+ T cells, activated CCR7+LAMP3+ dendritic cells and CCL19+ fibroblasts as well as chemokines that organize these cells. Within the stem-immunity hub, we find preferential interactions between CXCL10+ macrophages and TCF7-CD8+ T cells as well as between mature regulatory dendritic cells and TCF7+CD4+ and regulatory T cells. These results provide a picture of the spatial organization of the human intratumoral immune response and its relevance to patient immunotherapy outcomes.


Assuntos
Neoplasias Pulmonares , Humanos , Linfócitos T CD8-Positivos , Receptor de Morte Celular Programada 1 , Quimiocinas/metabolismo , Imunoterapia/métodos , Microambiente Tumoral
8.
Cell ; 184(18): 4734-4752.e20, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34450029

RESUMO

Immune responses to cancer are highly variable, with mismatch repair-deficient (MMRd) tumors exhibiting more anti-tumor immunity than mismatch repair-proficient (MMRp) tumors. To understand the rules governing these varied responses, we transcriptionally profiled 371,223 cells from colorectal tumors and adjacent normal tissues of 28 MMRp and 34 MMRd individuals. Analysis of 88 cell subsets and their 204 associated gene expression programs revealed extensive transcriptional and spatial remodeling across tumors. To discover hubs of interacting malignant and immune cells, we identified expression programs in different cell types that co-varied across tumors from affected individuals and used spatial profiling to localize coordinated programs. We discovered a myeloid cell-attracting hub at the tumor-luminal interface associated with tissue damage and an MMRd-enriched immune hub within the tumor, with activated T cells together with malignant and myeloid cells expressing T cell-attracting chemokines. By identifying interacting cellular programs, we reveal the logic underlying spatially organized immune-malignant cell networks.


Assuntos
Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Proteínas Morfogenéticas Ósseas/metabolismo , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Compartimento Celular , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Estudos de Coortes , Neoplasias Colorretais/genética , Reparo de Erro de Pareamento de DNA/genética , Células Endoteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade , Inflamação/patologia , Monócitos/patologia , Células Mieloides/patologia , Neutrófilos/patologia , Células Estromais/metabolismo , Linfócitos T/metabolismo , Transcrição Gênica
9.
Cell ; 182(6): 1606-1622.e23, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32888429

RESUMO

The enteric nervous system (ENS) coordinates diverse functions in the intestine but has eluded comprehensive molecular characterization because of the rarity and diversity of cells. Here we develop two methods to profile the ENS of adult mice and humans at single-cell resolution: RAISIN RNA-seq for profiling intact nuclei with ribosome-bound mRNA and MIRACL-seq for label-free enrichment of rare cell types by droplet-based profiling. The 1,187,535 nuclei in our mouse atlas include 5,068 neurons from the ileum and colon, revealing extraordinary neuron diversity. We highlight circadian expression changes in enteric neurons, show that disease-related genes are dysregulated with aging, and identify differences between the ileum and proximal/distal colon. In humans, we profile 436,202 nuclei, recovering 1,445 neurons, and identify conserved and species-specific transcriptional programs and putative neuro-epithelial, neuro-stromal, and neuro-immune interactions. The human ENS expresses risk genes for neuropathic, inflammatory, and extra-intestinal diseases, suggesting neuronal contributions to disease.


Assuntos
Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Neurônios/metabolismo , Corpos de Nissl/metabolismo , RNA Mensageiro/metabolismo , Análise de Célula Única/métodos , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Relógios Circadianos/genética , Colo/citologia , Colo/metabolismo , Retículo Endoplasmático Rugoso/genética , Retículo Endoplasmático Rugoso/metabolismo , Retículo Endoplasmático Rugoso/ultraestrutura , Células Epiteliais/metabolismo , Feminino , Predisposição Genética para Doença/genética , Humanos , Íleo/citologia , Íleo/metabolismo , Inflamação/genética , Inflamação/metabolismo , Enteropatias/genética , Enteropatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Corpos de Nissl/genética , Corpos de Nissl/ultraestrutura , RNA Mensageiro/genética , RNA-Seq , Ribossomos/metabolismo , Ribossomos/ultraestrutura , Células Estromais/metabolismo
10.
Cell ; 178(3): 714-730.e22, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31348891

RESUMO

Genome-wide association studies (GWAS) have revealed risk alleles for ulcerative colitis (UC). To understand their cell type specificities and pathways of action, we generate an atlas of 366,650 cells from the colon mucosa of 18 UC patients and 12 healthy individuals, revealing 51 epithelial, stromal, and immune cell subsets, including BEST4+ enterocytes, microfold-like cells, and IL13RA2+IL11+ inflammatory fibroblasts, which we associate with resistance to anti-TNF treatment. Inflammatory fibroblasts, inflammatory monocytes, microfold-like cells, and T cells that co-express CD8 and IL-17 expand with disease, forming intercellular interaction hubs. Many UC risk genes are cell type specific and co-regulated within relatively few gene modules, suggesting convergence onto limited sets of cell types and pathways. Using this observation, we nominate and infer functions for specific risk genes across GWAS loci. Our work provides a framework for interrogating complex human diseases and mapping risk variants to cell types and pathways.


Assuntos
Colite Ulcerativa/patologia , Colo/metabolismo , Adulto , Idoso , Anticorpos Monoclonais/uso terapêutico , Bestrofinas/metabolismo , Antígenos CD8/metabolismo , Estudos de Casos e Controles , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/metabolismo , Colo/patologia , Enterócitos/citologia , Enterócitos/metabolismo , Feminino , Loci Gênicos , Estudo de Associação Genômica Ampla , Humanos , Interleucina-17/metabolismo , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Linfócitos T/citologia , Linfócitos T/metabolismo , Trombospondinas/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Adulto Jovem
11.
Immunity ; 57(8): 1731-1733, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39142274

RESUMO

Response to immune checkpoint blockade is increased in obesity-related cancers, but the mechanisms remain unclear. In a recent issue of Nature, Bader et al. report that obesity in mice induces macrophage PD-1 upregulation to promote tumor growth while potentiating immunotherapy responses.


Assuntos
Macrófagos , Neoplasias , Obesidade , Obesidade/imunologia , Animais , Neoplasias/imunologia , Neoplasias/etiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Humanos , Camundongos , Receptor de Morte Celular Programada 1/metabolismo , Imunoterapia/métodos , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Paradoxo da Obesidade
12.
Cell ; 175(5): 1177-1179, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30445036

RESUMO

In Drosophila, well-delineated circuits control circadian rhythms, but the electrophysiological patterns that occur within these circuits are not well understood. In this issue, Tabuchi et al. clarify the temporal coding within a circuit, linking patterns of neural activity to sleep behavior.


Assuntos
Proteínas de Drosophila , Animais , Ritmo Circadiano , Drosophila , Plasticidade Neuronal , Sono
13.
Cell ; 175(4): 921-933.e14, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30388452

RESUMO

Contact-dependent growth inhibition (CDI) entails receptor-mediated delivery of CdiA-derived toxins into Gram-negative target bacteria. Using electron cryotomography, we show that each CdiA effector protein forms a filament extending ∼33 nm from the cell surface. Remarkably, the extracellular filament represents only the N-terminal half of the effector. A programmed secretion arrest sequesters the C-terminal half of CdiA, including the toxin domain, in the periplasm prior to target-cell recognition. Upon binding receptor, CdiA secretion resumes, and the periplasmic FHA-2 domain is transferred to the target-cell outer membrane. The C-terminal toxin region of CdiA then penetrates into the target-cell periplasm, where it is cleaved for subsequent translocation into the cytoplasm. Our findings suggest that the FHA-2 domain assembles into a transmembrane conduit for toxin transport into the periplasm of target bacteria. We propose that receptor-triggered secretion ensures that FHA-2 export is closely coordinated with integration into the target-cell outer membrane. VIDEO ABSTRACT.


Assuntos
Antibiose , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana/metabolismo , Sistemas de Secreção Tipo V/metabolismo , Extensões da Superfície Celular/metabolismo , Extensões da Superfície Celular/ultraestrutura , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Membrana/química , Domínios Proteicos , Receptores de Superfície Celular/metabolismo
14.
Cell ; 173(1): 117-129.e14, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29570992

RESUMO

Angiogenesis, the formation of new blood vessels by endothelial cells (ECs), is an adaptive response to oxygen/nutrient deprivation orchestrated by vascular endothelial growth factor (VEGF) upon ischemia or exercise. Hypoxia is the best-understood trigger of VEGF expression via the transcription factor HIF1α. Nutrient deprivation is inseparable from hypoxia during ischemia, yet its role in angiogenesis is poorly characterized. Here, we identified sulfur amino acid restriction as a proangiogenic trigger, promoting increased VEGF expression, migration and sprouting in ECs in vitro, and increased capillary density in mouse skeletal muscle in vivo via the GCN2/ATF4 amino acid starvation response pathway independent of hypoxia or HIF1α. We also identified a requirement for cystathionine-γ-lyase in VEGF-dependent angiogenesis via increased hydrogen sulfide (H2S) production. H2S mediated its proangiogenic effects in part by inhibiting mitochondrial electron transport and oxidative phosphorylation, resulting in increased glucose uptake and glycolytic ATP production.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Aminoácidos Sulfúricos/deficiência , Sulfeto de Hidrogênio/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator 4 Ativador da Transcrição/antagonistas & inibidores , Fator 4 Ativador da Transcrição/genética , Aminoácidos Sulfúricos/metabolismo , Animais , Cistationina gama-Liase/metabolismo , Modelos Animais de Doenças , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Isquemia/metabolismo , Isquemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Condicionamento Físico Animal , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
15.
Cell ; 168(1-2): 150-158.e10, 2017 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-28041849

RESUMO

Bacterial CRISPR-Cas systems utilize sequence-specific RNA-guided nucleases to defend against bacteriophage infection. As a countermeasure, numerous phages are known that produce proteins to block the function of class 1 CRISPR-Cas systems. However, currently no proteins are known to inhibit the widely used class 2 CRISPR-Cas9 system. To find these inhibitors, we searched cas9-containing bacterial genomes for the co-existence of a CRISPR spacer and its target, a potential indicator for CRISPR inhibition. This analysis led to the discovery of four unique type II-A CRISPR-Cas9 inhibitor proteins encoded by Listeria monocytogenes prophages. More than half of L. monocytogenes strains with cas9 contain at least one prophage-encoded inhibitor, suggesting widespread CRISPR-Cas9 inactivation. Two of these inhibitors also blocked the widely used Streptococcus pyogenes Cas9 when assayed in Escherichia coli and human cells. These natural Cas9-specific "anti-CRISPRs" present tools that can be used to regulate the genome engineering activities of CRISPR-Cas9.


Assuntos
Bacteriófagos/metabolismo , Sistemas CRISPR-Cas , Endonucleases/antagonistas & inibidores , Engenharia Genética , Listeria monocytogenes/enzimologia , Proteínas de Bactérias/antagonistas & inibidores , Proteína 9 Associada à CRISPR , Escherichia coli , Células HEK293 , Humanos , Listeria monocytogenes/imunologia , Listeria monocytogenes/virologia , Prófagos
16.
Mol Cell ; 84(11): 2036-2052.e7, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38688279

RESUMO

Alterations of bases in DNA constitute a major source of genomic instability. It is believed that base alterations trigger base excision repair (BER), generating DNA repair intermediates interfering with DNA replication. Here, we show that genomic uracil, a common type of base alteration, induces DNA replication stress (RS) without being processed by BER. In the absence of uracil DNA glycosylase (UNG), genomic uracil accumulates to high levels, DNA replication forks slow down, and PrimPol-mediated repriming is enhanced, generating single-stranded gaps in nascent DNA. ATR inhibition in UNG-deficient cells blocks the repair of uracil-induced gaps, increasing replication fork collapse and cell death. Notably, a subset of cancer cells upregulates UNG2 to suppress genomic uracil and limit RS, and these cancer cells are hypersensitive to co-treatment with ATR inhibitors and drugs increasing genomic uracil. These results reveal unprocessed genomic uracil as an unexpected source of RS and a targetable vulnerability of cancer cells.


Assuntos
Reparo do DNA , Replicação do DNA , Instabilidade Genômica , Uracila-DNA Glicosidase , Uracila , Humanos , Uracila/metabolismo , Uracila-DNA Glicosidase/metabolismo , Uracila-DNA Glicosidase/genética , Reparo do DNA/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/genética , Dano ao DNA , Linhagem Celular Tumoral , Neoplasias/genética , Neoplasias/patologia , Neoplasias/metabolismo
17.
Mol Cell ; 2024 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-39357514

RESUMO

Inter-kingdom communication through small molecules is essential to the coexistence of organisms in an ecosystem. In soil communities, the plant root is a nexus of interactions for a remarkable number of fungi and is a source of small-molecule plant hormones that shape fungal compositions. Although hormone signaling pathways are established in plants, how fungi perceive and respond to molecules is unclear because many plant-associated fungi are recalcitrant to experimentation. Here, we develop an approach using the model fungus, Saccharomyces cerevisiae, to elucidate mechanisms of fungal response to plant hormones. Two plant hormones, strigolactone and methyl jasmonate, produce unique transcript profiles in yeast, affecting phosphate and sugar metabolism, respectively. Genetic analysis in combination with structural studies suggests that SLs require the high-affinity transporter Pho84 to modulate phosphate homeostasis. The ability to study small-molecule plant hormones in a tractable genetic system should have utility in understanding fungal-plant interactions.

18.
CA Cancer J Clin ; 74(4): 383-396, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38703384

RESUMO

Cancer mortality rates have declined during the last 28 years, but that process is not equitably shared. Disparities in cancer outcomes by race, ethnicity, socioeconomic status, sexual orientation and gender identity, and geographic location persist across the cancer care continuum. Consequently, community outreach and engagement (COE) efforts within National Cancer Institute-Designated Cancer Center (NCI-DCC) catchment areas have intensified during the last 10 years as has the emphasis on COE and catchment areas in NCI's Cancer Center Support Grant applications. This review article attempts to provide a historic perspective of COE within NCI-DCCs. Improving COE has long been an important initiative for the NCI, but it was not until 2012 and 2016 that NCI-DCCs were required to define their catchment areas rigorously and to provide specific descriptions of COE interventions, respectively. NCI-DCCs had previously lacked adequate focus on the inclusion of historically marginalized patients in cancer innovation efforts. Integrating COE efforts throughout the research and operational aspects of the cancer centers, at both the patient and community levels, will expand the footprint of COE efforts within NCI-DCCs. Achieving this change requires sustained commitment by the centers to adjust their activities and improve access and outcomes for historically marginalized communities.


Assuntos
Institutos de Câncer , Relações Comunidade-Instituição , National Cancer Institute (U.S.) , Neoplasias , Humanos , Estados Unidos/epidemiologia , Neoplasias/terapia , Neoplasias/epidemiologia , Institutos de Câncer/organização & administração , Disparidades em Assistência à Saúde
19.
Genes Dev ; 37(5-6): 171-190, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36859339

RESUMO

Both the presence of an abnormal complement of chromosomes (aneuploidy) and an increased frequency of chromosome missegregation (chromosomal instability) are hallmarks of cancer. Analyses of cancer genome data have identified certain aneuploidy patterns in tumors; however, the bases behind their selection are largely unexplored. By establishing time-resolved long-term adaptation protocols, we found that human cells adapt to persistent spindle assembly checkpoint (SAC) inhibition by acquiring specific chromosome arm gains and losses. Independently adapted populations converge on complex karyotypes, which over time are refined to contain ever smaller chromosomal changes. Of note, the frequencies of chromosome arm gains in adapted cells correlate with those detected in cancers, suggesting that our cellular adaptation approach recapitulates selective traits that dictate the selection of aneuploidies frequently observed across many cancer types. We further engineered specific aneuploidies to determine the genetic basis behind the observed karyotype patterns. These experiments demonstrated that the adapted and engineered aneuploid cell lines limit CIN by extending mitotic duration. Heterozygous deletions of key SAC and APC/C genes recapitulated the rescue phenotypes of the monosomic chromosomes. We conclude that aneuploidy-induced gene dosage imbalances of individual mitotic regulators are sufficient for altering mitotic timing to reduce CIN.


Assuntos
Pontos de Checagem da Fase M do Ciclo Celular , Neoplasias , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Aneuploidia , Neoplasias/genética , Instabilidade Cromossômica/genética , Cariótipo , Fuso Acromático/genética , Mitose
20.
Cell ; 161(7): 1492-3, 2015 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-26091029

RESUMO

XBP1 is part of the ER stress response, and when activated in cancer cells, it fosters tumor growth. In this issue of Cell, Cubillos-Ruiz et al. demonstrate that XBP1 in tumor-infiltrating dendritic cells blunts anti-tumor immunity. These findings further imply XBP1 as a relevant target for cancer therapy.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células Dendríticas/patologia , Estresse do Retículo Endoplasmático , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Fatores de Transcrição/metabolismo , Animais , Feminino , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa