Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Publication year range
1.
Genes Dev ; 37(3-4): 86-102, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36732025

ABSTRACT

Glioblastomas (GBMs) are heterogeneous, treatment-resistant tumors driven by populations of cancer stem cells (CSCs). However, few molecular mechanisms critical for CSC population maintenance have been exploited for therapeutic development. We developed a spatially resolved loss-of-function screen in GBM patient-derived organoids to identify essential epigenetic regulators in the SOX2-enriched, therapy-resistant niche and identified WDR5 as indispensable for this population. WDR5 is a component of the WRAD complex, which promotes SET1 family-mediated Lys4 methylation of histone H3 (H3K4me), associated with positive regulation of transcription. In GBM CSCs, WDR5 inhibitors blocked WRAD complex assembly and reduced H3K4 trimethylation and expression of genes involved in CSC-relevant oncogenic pathways. H3K4me3 peaks lost with WDR5 inhibitor treatment occurred disproportionally on POU transcription factor motifs, including the POU5F1(OCT4)::SOX2 motif. Use of a SOX2/OCT4 reporter demonstrated that WDR5 inhibitor treatment diminished cells with high reporter activity. Furthermore, WDR5 inhibitor treatment and WDR5 knockdown altered the stem cell state, disrupting CSC in vitro growth and self-renewal, as well as in vivo tumor growth. These findings highlight the role of WDR5 and the WRAD complex in maintaining the CSC state and provide a rationale for therapeutic development of WDR5 inhibitors for GBM and other advanced cancers.


Subject(s)
Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Histone-Lysine N-Methyltransferase/metabolism , Transcription Factors , Neoplastic Stem Cells/pathology , Intracellular Signaling Peptides and Proteins/genetics
2.
Cell ; 149(5): 979-93, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22608084

ABSTRACT

All cancers carry somatic mutations. The patterns of mutation in cancer genomes reflect the DNA damage and repair processes to which cancer cells and their precursors have been exposed. To explore these mechanisms further, we generated catalogs of somatic mutation from 21 breast cancers and applied mathematical methods to extract mutational signatures of the underlying processes. Multiple distinct single- and double-nucleotide substitution signatures were discernible. Cancers with BRCA1 or BRCA2 mutations exhibited a characteristic combination of substitution mutation signatures and a distinctive profile of deletions. Complex relationships between somatic mutation prevalence and transcription were detected. A remarkable phenomenon of localized hypermutation, termed "kataegis," was observed. Regions of kataegis differed between cancers but usually colocalized with somatic rearrangements. Base substitutions in these regions were almost exclusively of cytosine at TpC dinucleotides. The mechanisms underlying most of these mutational signatures are unknown. However, a role for the APOBEC family of cytidine deaminases is proposed.


Subject(s)
Breast Neoplasms/genetics , DNA Mutational Analysis , Genome-Wide Association Study , Mutation , APOBEC-1 Deaminase , BRCA2 Protein/genetics , Cytidine Deaminase/metabolism , Female , Genes, BRCA1 , High-Throughput Nucleotide Sequencing , Humans
3.
J Biol Chem ; 299(11): 105299, 2023 11.
Article in English | MEDLINE | ID: mdl-37777156

ABSTRACT

Microbes living in the intestine can regulate key signaling processes in the central nervous system that directly impact brain health. This gut-brain signaling axis is partially mediated by microbe-host-dependent immune regulation, gut-innervating neuronal communication, and endocrine-like small molecule metabolites that originate from bacteria to ultimately cross the blood-brain barrier. Given the mounting evidence of gut-brain crosstalk, a new therapeutic approach of "psychobiotics" has emerged, whereby strategies designed to primarily modify the gut microbiome have been shown to improve mental health or slow neurodegenerative diseases. Diet is one of the most powerful determinants of gut microbiome community structure, and dietary habits are associated with brain health and disease. Recently, the metaorganismal (i.e., diet-microbe-host) trimethylamine N-oxide (TMAO) pathway has been linked to the development of several brain diseases including Alzheimer's, Parkinson's, and ischemic stroke. However, it is poorly understood how metaorganismal TMAO production influences brain function under normal physiological conditions. To address this, here we have reduced TMAO levels by inhibiting gut microbe-driven choline conversion to trimethylamine (TMA), and then performed comprehensive behavioral phenotyping in mice. Unexpectedly, we find that TMAO is particularly enriched in the murine olfactory bulb, and when TMAO production is blunted at the level of bacterial choline TMA lyase (CutC/D), olfactory perception is altered. Taken together, our studies demonstrate a previously underappreciated role for the TMAO pathway in olfactory-related behaviors.


Subject(s)
Olfactory Perception , Animals , Mice , Bacteria/metabolism , Choline/metabolism , Methylamines/metabolism , Female , Mice, Inbred C57BL
4.
Cell ; 138(2): 352-65, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19592082

ABSTRACT

Cyclins are regulatory subunits of cyclin-dependent kinases. Cyclin A, the first cyclin ever cloned, is thought to be an essential component of the cell-cycle engine. Mammalian cells encode two A-type cyclins, testis-specific cyclin A1 and ubiquitously expressed cyclin A2. Here, we tested the requirement for cyclin A function using conditional knockout mice lacking both A-type cyclins. We found that acute ablation of cyclin A in fibroblasts did not affect cell proliferation, but led to prolonged expression of another cyclin, cyclin E, across the cell cycle. However, combined ablation of all A- and E-type cyclins extinguished cell division. In contrast, cyclin A function was essential for cell-cycle progression of hematopoietic and embryonic stem cells. Expression of cyclin A is particularly high in these compartments, which might render stem cells dependent on cyclin A, whereas in fibroblasts cyclins A and E play redundant roles in cell proliferation.


Subject(s)
Cyclin A/metabolism , Embryo, Mammalian/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Cyclin A/genetics , Cyclin E/genetics , Cyclin E/metabolism , Mice , Mice, Knockout
5.
Cell Mol Life Sci ; 77(24): 5101-5119, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32506168

ABSTRACT

The metabolic complexity and flexibility commonly observed in brain tumors, especially glioblastoma, is fundamental for their development and progression. The ability of tumor cells to modify their genetic landscape and adapt metabolically, subverts therapeutic efficacy, and inevitably instigates therapeutic resistance. To overcome these challenges and develop effective therapeutic strategies targeting essential metabolic processes, it is necessary to identify the mechanisms underlying heterogeneity and define metabolic preferences and liabilities of malignant cells. In this review, we will discuss metabolic diversity in brain cancer and highlight the role of cancer stem cells in regulating metabolic heterogeneity. We will also highlight potential therapeutic modalities targeting metabolic vulnerabilities and examine how intercellular metabolic signaling can shape the tumor microenvironment.


Subject(s)
Brain Neoplasms/genetics , Genetic Heterogeneity , Glioblastoma/genetics , Metabolism/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Glycolysis/genetics , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/genetics , Tumor Microenvironment
6.
Breast Cancer Res Treat ; 181(3): 679-689, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32367460

ABSTRACT

PURPOSE: Discordance between HER2 expression in tumor tissue (tHER2) and HER2 status on circulating tumor cells (cHER2) has been reported. It remains largely underexplored whether patients with tHER2-/cHER2+ can benefit from anti-HER2 targeted therapies. METHODS: cHER2 status was determined in 105 advanced-stage patients with tHER2- breast tumors. Association between cHER2 status and progression-free survival (PFS) was analyzed by univariate and multivariate Cox models and survival differences were compared by Kaplan-Meier method. RESULTS: Compared to the patients with low-risk cHER2 (cHER2+ < 2), those with high-risk cHER2 (cHER2+ ≥ 2) had shorter survival time and an increased risk for disease progression (hazard ratio [HR] 2.16, 95% confidence interval [CI] 1.20-3.88, P = 0.010). Among the patients with high-risk cHER2, those who received anti-HER2 targeted therapies had improved PFS compared with those who did not (HR 0.30, 95% CI 0.10-0.92, P = 0.035). In comparison, anti-HER2 targeted therapy did not affect PFS among those with low-risk cHER2 (HR 0.70, 95% CI 0.36-1.38, P = 0.306). Similar results were obtained after adjusting covariates. A longitudinal analysis of 67 patients with cHER2 detected during follow-ups found that those whose cHER2 status changed from high-risk at baseline to low-risk at first follow-up exhibited a significantly improved survival compared to those whose cHER2 remained high-risk (median PFS: 11.7 weeks vs. 2.0 weeks, log-rank P = 0.001). CONCLUSION: In advanced-stage breast cancer patients with tHER2- tumors, cHER2 status has the potential to guide the use of anti-HER2 targeted therapy in patients with high-risk cHER2.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Receptor, ErbB-2/metabolism , Breast Neoplasms/blood , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Humans , Middle Aged , Neoplasm Staging , Neoplastic Cells, Circulating/metabolism , Receptor, ErbB-2/genetics , Survival Rate
7.
J Pathol ; 244(3): 260-264, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29282720

ABSTRACT

Glioblastoma (GBM) cancer stem cells (CSCs) are insidious. They extensively infiltrate brain tissue, resist radiotherapy and chemotherapy, and are thought to represent the ultimate drivers of disease progression. New research has identified CD109, a GPI-anchored protein, on a population of perivascular CSCs. Investigation of primary human tumour tissue suggests a role for CD109-expressing CSCs in the progression from low-grade to high-grade glioma, and animal modelling reveals a critical role for CD109 in the maintenance of the GBM CSC phenotype. Furthermore, CD109-expressing CSCs appear to drive the proliferation of adjacent non-stem tumour cells (NSTCs) in a rare example of CSC-NSTC cooperative interaction. With this Commentary, we highlight the newly revealed biology of CD109, and offer a synthesis of the published information on glioma CSCs in a variety of anatomical growth zones. We also discuss the landscape of interacting cells within GBM tumours, emphasizing the few reported examples of pro-tumourigenic, interactive tumour cell partnerships, as well as a variety of tumour cell-non-transformed neural cell interactions. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Colorectal Neoplasms , Glioblastoma , Glioma , Adult , Animals , Antigens, CD/genetics , Bone Morphogenetic Proteins , GPI-Linked Proteins , Humans , Neoplasm Proteins , Neoplastic Stem Cells , Prognosis , United Kingdom
8.
Breast Cancer Res ; 19(1): 23, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28264701

ABSTRACT

BACKGROUND: The bone-targeting agent zoledronic acid (ZOL) increases breast cancer survival in subsets of patients, but the underlying reasons for this protective effect are unknown. ZOL modulates the activity of osteoclasts and osteoblasts, which form hematopoietic stem cell niches, and therefore may affect hematopoietic cells that play a role in breast cancer progression. METHOD: Immunocompetent and immunocompromised strains of mice commonly used for breast cancer research were injected with a single, clinically relevant dose of ZOL (100 µg/kg) or vehicle control. The effects of ZOL on the bone marrow microenvironment (bone volume, bone cell number/activity, extracellular matrix composition) were established at various time points following treatment, using micro-computed tomography (µCT) analysis, histomorphometry, ELISA and immunofluorescence. The effects on peripheral blood and bone marrow hematopoietic progenitor populations were assessed using a HEMAVET® hematology analyzer and multicolor flow cytometry, respectively. Tumor support function of bone marrow cells was determined using an in vivo functional assay developed in our laboratory. RESULTS: Using multiple mouse strains, we observed transient changes in numbers of hematopoietic stem cells, myeloid-biased progenitor cells, and lymphoid-biased cells concurrent with changes to hematopoietic stem cell niches following ZOL administration. Importantly, bone marrow cells from mice treated with a single, clinically relevant dose of ZOL inhibited breast tumor outgrowth in vivo. The ZOL-induced tumor suppressive function of the bone marrow persisted beyond the time point at which numbers of hematopoietic progenitor cells had returned to baseline. CONCLUSIONS: These findings provide novel evidence that alterations to the bone marrow play a role in the anti-tumor activity of ZOL and suggest possibilities for capitalizing on the beneficial effects of ZOL in reducing breast cancer development and progression.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Breast Neoplasms/blood , Breast Neoplasms/metabolism , Diphosphonates/pharmacology , Hematopoiesis/drug effects , Imidazoles/pharmacology , Animals , Bone Marrow/diagnostic imaging , Bone Marrow/metabolism , Bone Marrow/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Bone and Bones/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Colony-Forming Units Assay , Disease Models, Animal , Extracellular Matrix , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Leukocyte Count , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , X-Ray Microtomography , Zoledronic Acid
9.
Stem Cells ; 34(8): 2026-39, 2016 08.
Article in English | MEDLINE | ID: mdl-27145382

ABSTRACT

Shifting the balance away from tumor-mediated immune suppression toward tumor immune rejection is the conceptual foundation for a variety of immunotherapy efforts currently being tested. These efforts largely focus on activating antitumor immune responses but are confounded by multiple immune cell populations, including myeloid-derived suppressor cells (MDSCs), which serve to suppress immune system function. We have identified immune-suppressive MDSCs in the brains of GBM patients and found that they were in close proximity to self-renewing cancer stem cells (CSCs). MDSCs were selectively depleted using 5-flurouracil (5-FU) in a low-dose administration paradigm, which resulted in prolonged survival in a syngeneic mouse model of glioma. In coculture studies, patient-derived CSCs but not nonstem tumor cells selectively drove MDSC-mediated immune suppression. A cytokine screen revealed that CSCs secreted multiple factors that promoted this activity, including macrophage migration inhibitory factor (MIF), which was produced at high levels by CSCs. Addition of MIF increased production of the immune-suppressive enzyme arginase-1 in MDSCs in a CXCR2-dependent manner, whereas blocking MIF reduced arginase-1 production. Similarly to 5-FU, targeting tumor-derived MIF conferred a survival advantage to tumor-bearing animals and increased the cytotoxic T cell response within the tumor. Importantly, tumor cell proliferation, survival, and self-renewal were not impacted by MIF reduction, demonstrating that MIF is primarily an indirect promoter of GBM progression, working to suppress immune rejection by activating and protecting immune suppressive MDSCs within the GBM tumor microenvironment. Stem Cells 2016;34:2026-2039.


Subject(s)
Brain Neoplasms/immunology , Glioblastoma/immunology , Immune Evasion , Macrophage Migration-Inhibitory Factors/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Neoplastic Stem Cells/metabolism , Animals , Arginase/metabolism , Brain Neoplasms/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Female , Glioblastoma/pathology , Humans , Immune Evasion/drug effects , Mice, Inbred C57BL , Mice, Nude , Myeloid-Derived Suppressor Cells/drug effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Tumor Microenvironment/drug effects
11.
Nature ; 462(7276): 1005-10, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20033038

ABSTRACT

Multiple somatic rearrangements are often found in cancer genomes; however, the underlying processes of rearrangement and their contribution to cancer development are poorly characterized. Here we use a paired-end sequencing strategy to identify somatic rearrangements in breast cancer genomes. There are more rearrangements in some breast cancers than previously appreciated. Rearrangements are more frequent over gene footprints and most are intrachromosomal. Multiple rearrangement architectures are present, but tandem duplications are particularly common in some cancers, perhaps reflecting a specific defect in DNA maintenance. Short overlapping sequences at most rearrangement junctions indicate that these have been mediated by non-homologous end-joining DNA repair, although varying sequence patterns indicate that multiple processes of this type are operative. Several expressed in-frame fusion genes were identified but none was recurrent. The study provides a new perspective on cancer genomes, highlighting the diversity of somatic rearrangements and their potential contribution to cancer development.


Subject(s)
Breast Neoplasms/genetics , Chromosome Aberrations , Gene Rearrangement/genetics , Genome, Human/genetics , Cell Line, Tumor , Cells, Cultured , DNA Breaks , Female , Genomic Library , Humans , Sequence Analysis, DNA
12.
J Neurosci ; 33(39): 15603-17, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-24068827

ABSTRACT

Glioblastoma (GBM) remains the most pervasive and lethal of all brain malignancies. One factor that contributes to this poor prognosis is the highly invasive character of the tumor. GBM is characterized by microscopic infiltration of tumor cells throughout the brain, whereas non-neural metastases, as well as select lower grade gliomas, develop as self-contained and clearly delineated lesions. Illustrated by rodent xenograft tumor models as well as pathological human patient specimens, we present evidence that one fundamental switch between these two distinct pathologies--invasion and noninvasion--is mediated through the tumor extracellular matrix. Specifically, noninvasive lesions are associated with a rich matrix containing substantial amounts of glycosylated chondroitin sulfate proteoglycans (CSPGs), whereas glycosylated CSPGs are essentially absent from diffusely infiltrating tumors. CSPGs, acting as central organizers of the tumor microenvironment, dramatically influence resident reactive astrocytes, inducing their exodus from the tumor mass and the resultant encapsulation of noninvasive lesions. Additionally, CSPGs induce activation of tumor-associated microglia. We demonstrate that the astrogliotic capsule can directly inhibit tumor invasion, and its absence from GBM presents an environment favorable to diffuse infiltration. We also identify the leukocyte common antigen-related phosphatase receptor (PTPRF) as a putative intermediary between extracellular glycosylated CSPGs and noninvasive tumor cells. In all, we present CSPGs as critical regulators of brain tumor histopathology and help to clarify the role of the tumor microenvironment in brain tumor invasion.


Subject(s)
Brain Neoplasms/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Glioma/metabolism , Tumor Microenvironment , Adult , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cells, Cultured , Child , Chondroitin Sulfate Proteoglycans/genetics , Female , Glioma/pathology , Glycosylation , Humans , Male , Mice , Microglia/metabolism , Microglia/pathology , Middle Aged , Neoplasm Invasiveness , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Xenograft Model Antitumor Assays
13.
J Neurooncol ; 117(1): 15-24, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24510433

ABSTRACT

Glioblastoma (GBM) is the most common malignant adult brain tumor and carries a poor prognosis due to primary and acquired resistance. While many cellular features of GBM have been documented, it is unclear if cells within these tumors extend a primary cilium, an organelle whose associated signaling pathways may regulate proliferation, migration, and survival of neural precursor and tumor cells. Using immunohistochemical and electron microscopy (EM) techniques, we screened human GBM tumor biopsies and primary cell lines for cilia. Immunocytochemical staining of five primary GBM cell lines revealed that between 8 and 25 % of the cells in each line possessed gamma tubulin-positive basal bodies from which extended acetylated, alpha-tubulin-positive axonemes. EM analyses confirmed the presence of cilia at the cell surface and revealed that their axonemes contained organized networks of microtubules, a structural feature consistent with our detection of IFT88 and Arl13b, two trafficked cilia proteins, along the lengths of the axonemes. Notably, cilia were detected in each of 23 tumor biopsies (22 primary and 1 recurrent) examined. These cilia were distributed across the tumor landscape including regions proximal to the vasculature and within necrotic areas. Moreover, ciliated cells within these tumors co-stained with Ki67, a marker for actively dividing cells, and ZEB1, a transcription factor that is upregulated in GBM and linked to tumor initiation, invasion, and chemoresistance. Collectively, our data show that subpopulations of cells within human GBM tumors are ciliated. In view of mounting evidence supporting roles of primary cilia in tumor initiation and propagation, it is likely that further study of the effects of cilia on GBM tumor cell function will improve our understanding of GBM pathogenesis and may provide new directions for GBM treatment strategies.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/ultrastructure , Cilia/ultrastructure , Glioblastoma/metabolism , Glioblastoma/ultrastructure , ADP-Ribosylation Factors/metabolism , Aged, 80 and over , Axoneme/metabolism , Axoneme/ultrastructure , Basal Bodies/metabolism , Basal Bodies/ultrastructure , Cell Line, Tumor , Cilia/metabolism , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Microscopy, Electron , Middle Aged , Transcription Factors/metabolism , Tubulin/metabolism , Tumor Suppressor Proteins/metabolism , Zinc Finger E-box-Binding Homeobox 1
14.
PLoS One ; 19(6): e0304564, 2024.
Article in English | MEDLINE | ID: mdl-38900724

ABSTRACT

Early optimism saw possibilities for social media to renew democratic discourse, marked by hopes for individuals from diverse backgrounds to find opportunities to learn from and interact with others different from themselves. This optimism quickly waned as social media seemed to breed ideological homophily marked by "filter bubbles" or "echo chambers." A typical response to the sense of fragmentation has been to encourage exposure to more cross-partisan sources of information. But do outlets that reach across partisan lines in fact generate more civil discourse? And does the civility of discourse hosted by such outlets vary depending on the political context in which they operate? To answer these questions, we identified bubble reachers, users who distribute content that reaches other users with diverse political opinions in recent presidential elections in Brazil, where populism has deep roots in the political culture, and Canada, where the political culture is comparatively moderate. Given that background, this research studies unexplored properties of content shared by bubble reachers, specifically the quality of conversations and comments it generates. We examine how ideologically neutral bubble reachers differ from ideologically partisan accounts in the level of uncivil discourse they provoke, and explore how this varies in the context of the two countries considered. Our results suggest that while ideologically neutral bubble reachers support less uncivil discourse in Canada, the opposite relationship holds in Brazil. Even non-political content by ideologically neutral bubble reachers elicits a considerable amount of uncivil discourse in Brazil. This indicates that bubble reaching and incivility are moderated by the national political context. Our results complicate the simple hypothesis of a universal impact of neutral bubble reachers across contexts.


Subject(s)
Politics , Social Media , Humans , Brazil , Canada , Public Opinion , Communication
15.
J Clin Invest ; 134(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38299594

ABSTRACT

A major challenge in treating patients with glioblastoma is the inability to eliminate highly invasive cells with chemotherapy, radiation, or surgical resection. As cancer cells face the issue of replicating or invading neighboring tissue, they rewire their metabolism in a concerted effort to support necessary cellular processes and account for altered nutrient abundance. In this issue of the JCI, Garcia et al. compared an innovative 3D hydrogel-based invasion device to regional patient biopsies through a comprehensive multiomics-based approach paired with a CRISPR knockout screen. Their findings elucidate a role for cystathionine γ-lyase (CTH), an enzyme in the transsulfuration pathway, as a means of regulating the cellular response to oxidative stress. CTH-mediated conversion of cystathionine to cysteine was necessary for regulating reactive oxygen species to support invasion. Meanwhile, inhibition of CTH suppressed the invasive glioblastoma phenotype. However, inhibiting CTH resulted in a larger overall tumor mass. These findings suggest that targeting the transsulfuration pathway may serve as a means of redirecting glioblastoma to proliferate or invade.


Subject(s)
Glioblastoma , Humans , Glioblastoma/genetics , Glioblastoma/pathology , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Cystathionine/metabolism , Oxidative Stress , Reactive Oxygen Species
16.
Neuro Oncol ; 26(5): 785-795, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38394444

ABSTRACT

Over the past 2 decades, the cancer stem cell (CSC) hypothesis has provided insight into many malignant tumors, including glioblastoma (GBM). Cancer stem cells have been identified in patient-derived tumors and in some mouse models, allowing for a deeper understanding of cellular and molecular mechanisms underlying GBM growth and therapeutic resistance. The CSC hypothesis has been the cornerstone of cellular heterogeneity, providing a conceptual and technical framework to explain this longstanding phenotype in GBM. This hypothesis has evolved to fit recent insights into how cellular plasticity drives tumor growth to suggest that CSCs do not represent a distinct population but rather a cellular state with substantial plasticity that can be achieved by non-CSCs under specific conditions. This has further been reinforced by advances in genomics, including single-cell approaches, that have used the CSC hypothesis to identify multiple putative CSC states with unique properties, including specific developmental and metabolic programs. In this review, we provide a historical perspective on the CSC hypothesis and its recent evolution, with a focus on key functional phenotypes, and provide an update on the definition for its use in future genomic studies.


Subject(s)
Brain Neoplasms , Glioblastoma , Neoplastic Stem Cells , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/genetics , Humans , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Animals , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics
17.
Res Sq ; 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38585839

ABSTRACT

Many cancers, including glioblastoma (GBM), have a male-biased sex difference in incidence and outcome. The underlying reasons for this sex bias are unclear but likely involve differences in tumor cell state and immune response. This effect is further amplified by sex hormones, including androgens, which have been shown to inhibit anti-tumor T cell immunity. Here, we show that androgens drive anti-tumor immunity in brain tumors, in contrast to its effect in other tumor types. Upon castration, tumor growth was accelerated with attenuated T cell function in GBM and brain tumor models, but the opposite was observed when tumors were located outside the brain. Activity of the hypothalamus-pituitary-adrenal gland (HPA) axis was increased in castrated mice, particularly in those with brain tumors. Blockade of glucocorticoid receptors reversed the accelerated tumor growth in castrated mice, indicating that the effect of castration was mediated by elevated glucocorticoid signaling. Furthermore, this mechanism was not GBM specific, but brain specific, as hyperactivation of the HPA axis was observed with intracranial implantation of non-GBM tumors in the brain. Together, our findings establish that brain tumors drive distinct endocrine-mediated mechanisms in the androgen-deprived setting and highlight the importance of organ-specific effects on anti-tumor immunity.

18.
Neurooncol Adv ; 6(1): vdad154, 2024.
Article in English | MEDLINE | ID: mdl-38239626

ABSTRACT

Background: Glioblastoma (GBM) displays alterations in iron that drive proliferation and tumor growth. Iron regulation is complex and involves many regulatory mechanisms, including the homeostatic iron regulator (HFE) gene, which encodes the homeostatic iron regulatory protein. While HFE is upregulated in GBM and correlates with poor survival outcomes, the function of HFE in GBM remains unclear. Methods: We interrogated the impact of cell-intrinsic Hfe expression on proliferation and survival of intracranially implanted animals through genetic gain- and loss-of-function approaches in syngeneic mouse glioma models, along with in vivo immune assessments. We also determined the expression of iron-associated genes and their relationship to survival in GBM using public data sets and used transcriptional profiling to identify differentially expressed pathways in control compared to Hfe-knockdown cells. Results: Overexpression of Hfe accelerated GBM proliferation and reduced animal survival, whereas suppression of Hfe induced apoptotic cell death and extended survival, which was more pronounced in females and associated with attenuation of natural killer cells and CD8+ T cell activity. Analysis of iron gene signatures in Hfe-knockdown cells revealed alterations in the expression of several iron-associated genes, suggesting global disruption of intracellular iron homeostasis. Further analysis of differentially expressed pathways revealed oxidative stress as the top pathway upregulated following Hfe loss. Hfe knockdown indeed resulted in enhanced 55Fe uptake and generation of reactive oxygen species. Conclusions: These findings reveal an essential function for HFE in GBM cell growth and survival, as well as a sex-specific interaction with the immune response.

19.
Hematol Oncol Stem Cell Ther ; 16(3): 186-196, 2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37023220

ABSTRACT

BACKGROUND: The PARP inhibitors (PARPis) olaparib and talazoparib are currently approved for the treatment of deleterious germline BRCA1/2-mutated (gBRCA+) metastatic breast cancer (MBC). These approvals were based on improvements in progression-free survival (PFS) observed in two randomized controlled trials (RCTs). Other PARPis, such as veliparib and niraparib, have also been studied. We conducted this meta-analysis of RCTs to assess the PFS and overall survival (OS) benefits of PARPis in gBRCA + MBC. METHODS: We performed a systematic search for RCTs using the Cochrane Library, PubMed, Embase, and Web of Science databases up to March 2021. Only phase II and III RCTs evaluating PFS and OS for PARPis alone or in combination with chemotherapy (CT) and comparing the findings with standard CT were included in this meta-analysis. Pooled analysis of the hazard ratio (HR) was performed with RevMan v5.4 using a random effects method. RESULTS: Five RCTs with a total of 1563 BRCA-mutated MBC patients were included in this meta-analysis. Temozolomide was used in the treatment arm in the BROCADE trial. Since temozolomide has limited effects on breast cancer, this arm was excluded from our meta-analysis. A statistically significant increase in PFS was observed in the PARPi group compared to the standard CT group (HR, 0.64; 95% CI, 0.56-0.74; P < 0.00001). However, the differences in OS did not reach statistical significance (HR, 0.89; 95% CI, 0.77-1.02; P = 0.09). Moreover, differences were not observed in the adverse event profile between the two groups (odds ratio, 1.18; 95% CI, 0.84-1.64; P = 0.33). CONCLUSION: The results of our meta-analysis confirm the previously reported PFS benefit of PARPis over standard CT. PARPis lead to superior PFS in gBRCA + MBC when used alone or in combination with standard CT. The OS benefit is similar between PARPis and standard CT. Ongoing trials are evaluating the benefits of PARPis in early stage gBRCA + BC.


Subject(s)
Breast Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Female , Humans , BRCA1 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Temozolomide/therapeutic use , BRCA2 Protein/metabolism
20.
J Neurotrauma ; 40(23-24): 2500-2521, 2023 12.
Article in English | MEDLINE | ID: mdl-37606910

ABSTRACT

Spinal cord injuries (SCI), for which there are limited effective treatments, result in enduring paralysis and hypoesthesia, in part because of the inhibitory microenvironment that develops and limits regeneration/sprouting, especially during chronic stages. Recently, we discovered that targeted enzymatic removal of the inhibitory chondroitin sulfate proteoglycan (CSPG) component of the extracellular and perineuronal net (PNN) matrix via Chondroitinase ABC (ChABC) rapidly restored robust respiratory function to the previously paralyzed hemi-diaphragm after remarkably long times post-injury (up to 1.5 years) following a cervical level 2 lateral hemi-transection. Importantly, ChABC treatment at cervical level 4 in this chronic model also elicited improvements in gross upper arm function. In the present study, we focused on arm and hand function, seeking to highlight and optimize crude as well as fine motor control of the forearm and digits at lengthy chronic stages post-injury. However, instead of using ChABC, we utilized a novel and more clinically relevant systemic combinatorial treatment strategy designed to simultaneously reduce and overcome inhibitory CSPGs. Following a 3-month upper cervical spinal hemi-lesion using adult female Sprague Dawley rats, we show that the combined treatment had a profound effect on functional recovery of the chronically paralyzed forelimb and paw, as well as on precision movements of the digits. The regenerative and immune system related events that we describe deepen our basic understanding of the crucial role of CSPG-mediated inhibition via the PTPσ receptor in constraining functional synaptic plasticity at lengthy time points following SCI, hopefully leading to clinically relevant translational benefits.


Subject(s)
Chondroitin Sulfate Proteoglycans , Spinal Cord Injuries , Animals , Female , Rats , Chondroitin ABC Lyase/pharmacology , Chondroitin Sulfate Proteoglycans/pharmacology , Nerve Regeneration/physiology , Rats, Sprague-Dawley , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Spinal Cord , Forelimb
SELECTION OF CITATIONS
SEARCH DETAIL