Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Proc Natl Acad Sci U S A ; 112(1): 268-72, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535386

ABSTRACT

Huntington's disease, the most common inherited neurodegenerative disease, is characterized by a dramatic loss of deep-layer cortical and striatal neurons, as well as morbidity in midlife. Human genetic studies led to the identification of the causative gene, huntingtin. Recent genomic advances have also led to the identification of hundreds of potential interacting partners for huntingtin protein and many hypotheses as to the molecular mechanisms whereby mutant huntingtin leads to cellular dysfunction and death. However, the multitude of possible interacting partners and cellular pathways affected by mutant huntingtin has complicated efforts to understand the etiology of this disease, and to date no curative therapeutic exists. To address the general problem of identifying the disease-phenotype contributing genes from a large number of correlative studies, here we develop a synthetic lethal screening methodology for the mammalian central nervous system, called SLIC, for synthetic lethal in the central nervous system. Applying SLIC to the study of Huntington's disease, we identify the age-regulated glutathione peroxidase 6 (Gpx6) gene as a modulator of mutant huntingtin toxicity and show that overexpression of Gpx6 can dramatically alleviate both behavioral and molecular phenotypes associated with a mouse model of Huntington's disease. SLIC can, in principle, be used in the study of any neurodegenerative disease for which a mouse model exists, promising to reveal modulators of neurodegenerative disease in an unbiased fashion, akin to screens in simpler model organisms.


Subject(s)
Central Nervous System/enzymology , Central Nervous System/pathology , Glutathione Peroxidase/metabolism , Huntington Disease/enzymology , Huntington Disease/pathology , Animals , Behavior, Animal , Central Nervous System/physiopathology , Disease Models, Animal , Gene Expression Regulation , Humans , Huntington Disease/genetics , Huntington Disease/physiopathology , Mice , Motor Activity , Neostriatum/metabolism , Neostriatum/pathology , Neostriatum/physiopathology
2.
Proc Natl Acad Sci U S A ; 111(12): 4578-83, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24599591

ABSTRACT

Levodopa treatment is the major pharmacotherapy for Parkinson's disease. However, almost all patients receiving levodopa eventually develop debilitating involuntary movements (dyskinesia). Although it is known that striatal spiny projection neurons (SPNs) are involved in the genesis of this movement disorder, the molecular basis of dyskinesia is not understood. In this study, we identify distinct cell-type-specific gene-expression changes that occur in subclasses of SPNs upon induction of a parkinsonian lesion followed by chronic levodopa treatment. We identify several hundred genes, the expression of which is correlated with levodopa dose, many of which are under the control of activator protein-1 and ERK signaling. Despite homeostatic adaptations involving several signaling modulators, activator protein-1-dependent gene expression remains highly dysregulated in direct pathway SPNs upon chronic levodopa treatment. We also discuss which molecular pathways are most likely to dampen abnormal dopaminoceptive signaling in spiny projection neurons, hence providing potential targets for antidyskinetic treatments in Parkinson's disease.


Subject(s)
Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/genetics , Levodopa/adverse effects , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine/metabolism , Gene Expression , Homeostasis , Mice
3.
bioRxiv ; 2023 Jul 17.
Article in English | MEDLINE | ID: mdl-37502974

ABSTRACT

Tumor mutations can influence the surrounding microenvironment leading to suppression of anti-tumor immune responses and thereby contributing to tumor progression and failure of cancer therapies. Here we use genetically engineered lung cancer mouse models and patient samples to dissect how LKB1 mutations accelerate tumor growth by reshaping the immune microenvironment. Comprehensive immune profiling of LKB1 -mutant vs wildtype tumors revealed dramatic changes in myeloid cells, specifically enrichment of Arg1 + interstitial macrophages and SiglecF Hi neutrophils. We discovered a novel mechanism whereby autocrine LIF signaling in Lkb1 -mutant tumors drives tumorigenesis by reprogramming myeloid cells in the immune microenvironment. Inhibiting LIF signaling in Lkb1 -mutant tumors, via gene targeting or with a neutralizing antibody, resulted in a striking reduction in Arg1 + interstitial macrophages and SiglecF Hi neutrophils, expansion of antigen specific T cells, and inhibition of tumor progression. Thus, targeting LIF signaling provides a new therapeutic approach to reverse the immunosuppressive microenvironment of LKB1 -mutant tumors.

4.
Nat Commun ; 14(1): 6764, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37938580

ABSTRACT

Approximately 30% of early-stage lung adenocarcinoma patients present with disease progression after successful surgical resection. Despite efforts of mapping the genetic landscape, there has been limited success in discovering predictive biomarkers of disease outcomes. Here we performed a systematic multi-omic assessment of 143 tumors and matched tumor-adjacent, histologically-normal lung tissue with long-term patient follow-up. Through histologic, mutational, and transcriptomic profiling of tumor and adjacent-normal tissue, we identified an inflammatory gene signature in tumor-adjacent tissue as the strongest clinical predictor of disease progression. Single-cell transcriptomic analysis demonstrated the progression-associated inflammatory signature was expressed in both immune and non-immune cells, and cell type-specific profiling in monocytes further improved outcome predictions. Additional analyses of tumor-adjacent transcriptomic data from The Cancer Genome Atlas validated the association of the inflammatory signature with worse outcomes across cancers. Collectively, our study suggests that molecular profiling of tumor-adjacent tissue can identify patients at high risk for disease progression.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Adenocarcinoma of Lung/genetics , Inflammation/genetics , Lung Neoplasms/genetics , Lung , Disease Progression
5.
Nat Commun ; 13(1): 4443, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35927254

ABSTRACT

A significant proportion of colorectal cancer (CRC) patients develop peritoneal metastases (PM) in the course of their disease. PMs are associated with a poor quality of life, significant morbidity and dismal disease outcome. To improve care for this patient group, a better understanding of the molecular characteristics of CRC-PM is required. Here we present a comprehensive molecular characterization of a cohort of 52 patients. This reveals that CRC-PM represent a distinct CRC molecular subtype, CMS4, but can be further divided in three separate categories, each presenting with unique features. We uncover that the CMS4-associated structural protein Moesin plays a key role in peritoneal dissemination. Finally, we define specific evolutionary features of CRC-PM which indicate that polyclonal metastatic seeding underlies these lesions. Together our results suggest that CRC-PM should be perceived as a distinct disease entity.


Subject(s)
Colorectal Neoplasms , Neoplasms, Second Primary , Peritoneal Neoplasms , Colorectal Neoplasms/pathology , Humans , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/secondary , Peritoneum/metabolism , Quality of Life
6.
Mol Neurodegener ; 15(1): 29, 2020 05 24.
Article in English | MEDLINE | ID: mdl-32448329

ABSTRACT

Huntington's disease (HD) is an incurable neurodegenerative disorder caused by CAG trinucleotide expansions in the huntingtin gene. Markers of both systemic and CNS immune activation and inflammation have been widely noted in HD and mouse models of HD. In particular, elevation of the pro-inflammatory cytokine interleukin-6 (IL-6) is the earliest reported marker of immune activation in HD, and this elevation has been suggested to contribute to HD pathogenesis. To test the hypothesis that IL-6 deficiency would be protective against the effects of mutant huntingtin, we generated R6/2 HD model mice that lacked IL-6. Contrary to our prediction, IL-6 deficiency exacerbated HD-model associated behavioral phenotypes. Single nuclear RNA Sequencing (snRNA-seq) analysis of striatal cell types revealed that IL-6 deficiency led to the dysregulation of various genes associated with synaptic function, as well as the BDNF receptor Ntrk2. These data suggest that IL-6 deficiency exacerbates the effects of mutant huntingtin through dysregulation of genes of known relevance to HD pathobiology in striatal neurons, and further suggest that modulation of IL-6 to a level that promotes proper regulation of genes associated with synaptic function may hold promise as an HD therapeutic target.


Subject(s)
Brain/metabolism , Huntington Disease/metabolism , Interleukin-6/deficiency , Phenotype , Animals , Brain/physiopathology , Corpus Striatum/metabolism , Disease Models, Animal , Huntington Disease/genetics , Interleukin-6/metabolism , Mice, Transgenic
7.
Neuron ; 106(1): 76-89.e8, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32004439

ABSTRACT

Unbiased in vivo genome-wide genetic screening is a powerful approach to elucidate new molecular mechanisms, but such screening has not been possible to perform in the mammalian central nervous system (CNS). Here, we report the results of the first genome-wide genetic screens in the CNS using both short hairpin RNA (shRNA) and CRISPR libraries. Our screens identify many classes of CNS neuronal essential genes and demonstrate that CNS neurons are particularly sensitive not only to perturbations to synaptic processes but also autophagy, proteostasis, mRNA processing, and mitochondrial function. These results reveal a molecular logic for the common implication of these pathways across multiple neurodegenerative diseases. To further identify disease-relevant genetic modifiers, we applied our screening approach to two mouse models of Huntington's disease (HD). Top mutant huntingtin toxicity modifier genes included several Nme genes and several genes involved in methylation-dependent chromatin silencing and dopamine signaling, results that reveal new HD therapeutic target pathways.


Subject(s)
Cell Survival/genetics , Huntingtin Protein/genetics , Huntington Disease/genetics , Neostriatum/metabolism , Neurons/metabolism , Animals , Behavior, Animal , CRISPR-Cas Systems , Gene Knockdown Techniques , Gene Library , Genes, Essential/genetics , Mice , Mice, Transgenic , NM23 Nucleoside Diphosphate Kinases/genetics , Nucleoside Diphosphate Kinase D/genetics , Protein Aggregates , RNA Interference , RNA, Guide, Kinetoplastida , RNA, Small Interfering , Receptors, Dopamine D2/genetics , Sequence Analysis, RNA
8.
Cell Rep ; 21(10): 2688-2695, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29212017

ABSTRACT

Alteration of corticostriatal glutamatergic function is an early pathophysiological change associated with Huntington's disease (HD). The factors that regulate the maintenance of corticostriatal glutamatergic synapses post-developmentally are not well understood. Recently, the striatum-enriched transcription factor Foxp2 was implicated in the development of these synapses. Here, we show that, in mice, overexpression of Foxp2 in the adult striatum of two models of HD leads to rescue of HD-associated behaviors, while knockdown of Foxp2 in wild-type mice leads to development of HD-associated behaviors. We note that Foxp2 encodes the longest polyglutamine repeat protein in the human reference genome, and we show that it can be sequestered into aggregates with polyglutamine-expanded mutant Huntingtin protein (mHTT). Foxp2 overexpression in HD model mice leads to altered expression of several genes associated with synaptic function, genes that present additional targets for normalization of corticostriatal dysfunction in HD.


Subject(s)
Corpus Striatum/metabolism , Forkhead Transcription Factors/metabolism , Huntington Disease/metabolism , Repressor Proteins/metabolism , Animals , Blotting, Western , Disease Models, Animal , Fluorescent Antibody Technique, Indirect , Forkhead Transcription Factors/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Huntington Disease/genetics , Male , Mice , Phenotype , Repressor Proteins/genetics
9.
Nat Protoc ; 9(6): 1282-91, 2014.
Article in English | MEDLINE | ID: mdl-24810037

ABSTRACT

Cellular diversity and architectural complexity create barriers to understanding the function of the mammalian CNS at a molecular level. To address this problem, we have recently developed a methodology that provides the ability to profile the entire translated mRNA complement of any genetically defined cell population. This methodology, which we termed translating ribosome affinity purification, or TRAP, combines cell type-specific transgene expression with affinity purification of translating ribosomes. TRAP can be used to study the cell type-specific mRNA profiles of any genetically defined cell type, and it has been used in organisms ranging from Drosophila melanogaster to mice and human cultured cells. Unlike other methodologies that rely on microdissection, cell panning or cell sorting, the TRAP methodology bypasses the need for tissue fixation or single-cell suspensions (and the potential artifacts that these treatments introduce) and reports on mRNAs in the entire cell body. This protocol provides a step-by-step guide to implement the TRAP methodology, which takes 2 d to complete once all materials are in hand.


Subject(s)
Gene Expression Profiling/methods , RNA, Messenger/isolation & purification , Ribosomes/genetics , Single-Cell Analysis/methods , Animals , Cells, Cultured , Humans , Mice , Polyribosomes/genetics , Polyribosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL