Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Cell ; 145(4): 571-83, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21565614

ABSTRACT

The molecular basis for p53-mediated tumor suppression remains unclear. Here, to elucidate mechanisms of p53 tumor suppression, we use knockin mice expressing an allelic series of p53 transcriptional activation mutants. Microarray analysis reveals that one mutant, p53(25,26), is severely compromised for transactivation of most p53 target genes, and, moreover, p53(25,26) cannot induce G(1)-arrest or apoptosis in response to acute DNA damage. Surprisingly, p53(25,26) retains robust activity in senescence and tumor suppression, indicating that efficient transactivation of the majority of known p53 targets is dispensable for these pathways. In contrast, the transactivation-dead p53(25,26,53,54) mutant cannot induce senescence or inhibit tumorigenesis, like p53 nullizygosity. Thus, p53 transactivation is essential for tumor suppression but, intriguingly, in association with a small set of novel p53 target genes. Together, our studies distinguish the p53 transcriptional programs involved in acute DNA-damage responses and tumor suppression-a critical goal for designing therapeutics that block p53-dependent side effects of chemotherapy without compromising p53 tumor suppression.


Subject(s)
DNA Repair , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Cycle , Cellular Senescence , DNA Damage , Gene Knock-In Techniques , Humans , Mice , Mutation , Neoplasms/metabolism , Protein Structure, Tertiary , Transcriptional Activation , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
2.
EMBO J ; 39(14): e104036, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32484965

ABSTRACT

Mechanistic understanding of how ionizing radiation induces type I interferon signaling and how to amplify this signaling module should help to maximize the efficacy of radiotherapy. In the current study, we report that inhibitors of the DNA damage response kinase ATR can significantly potentiate ionizing radiation-induced innate immune responses. Using a series of mammalian knockout cell lines, we demonstrate that, surprisingly, both the cGAS/STING-dependent DNA-sensing pathway and the MAVS-dependent RNA-sensing pathway are responsible for type I interferon signaling induced by ionizing radiation in the presence or absence of ATR inhibitors. The relative contributions of these two pathways in type I interferon signaling depend on cell type and/or genetic background. We propose that DNA damage-elicited double-strand DNA breaks releases DNA fragments, which may either activate the cGAS/STING-dependent pathway or-especially in the case of AT-rich DNA sequences-be transcribed and initiate MAVS-dependent RNA sensing and signaling. Together, our results suggest the involvement of two distinct pathways in type I interferon signaling upon DNA damage. Moreover, radiation plus ATR inhibition may be a promising new combination therapy against cancer.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/immunology , DNA Breaks, Double-Stranded/radiation effects , Interferon Type I/immunology , Radiation, Ionizing , Signal Transduction/radiation effects , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line, Tumor , Humans , Interferon Type I/genetics , Signal Transduction/genetics , Signal Transduction/immunology
3.
Proc Natl Acad Sci U S A ; 115(24): 6279-6284, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29794221

ABSTRACT

Xerostomia (dry mouth) is the most common side effect of radiation therapy in patients with head and neck cancer and causes difficulty speaking and swallowing. Since aldehyde dehydrogenase 3A1 (ALDH3A1) is highly expressed in mouse salivary stem/progenitor cells (SSPCs), we sought to determine the role of ALDH3A1 in SSPCs using genetic loss-of-function and pharmacologic gain-of-function studies. Using DarkZone dye to measure intracellular aldehydes, we observed higher aldehyde accumulation in irradiated Aldh3a1-/- adult murine salisphere cells and in situ in whole murine embryonic salivary glands enriched in SSPCs compared with wild-type glands. To identify a safe ALDH3A1 activator for potential clinical testing, we screened a traditional Chinese medicine library and isolated d-limonene, commonly used as a food-flavoring agent, as a single constituent activator. ALDH3A1 activation by d-limonene significantly reduced aldehyde accumulation in SSPCs and whole embryonic glands, increased sphere-forming ability, decreased apoptosis, and improved submandibular gland structure and function in vivo after radiation. A phase 0 study in patients with salivary gland tumors showed effective delivery of d-limonene into human salivary glands following daily oral dosing. Given its safety and bioavailability, d-limonene may be a good clinical candidate for mitigating xerostomia in patients with head and neck cancer receiving radiation therapy.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Aldehydes/metabolism , Cyclohexenes/pharmacology , Radiotherapy/adverse effects , Salivary Glands/metabolism , Terpenes/pharmacology , Xerostomia/metabolism , Animals , Apoptosis/drug effects , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Limonene , Medicine, Chinese Traditional/methods , Mice , Mice, Inbred C57BL , Protective Agents/pharmacology , Salivary Glands/drug effects , Salivary Glands/radiation effects , Stem Cells/drug effects , Stem Cells/metabolism , Submandibular Gland/drug effects , Submandibular Gland/metabolism , Xerostomia/drug therapy
4.
Genes Dev ; 27(9): 1016-31, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23651856

ABSTRACT

The mechanisms by which the p53 tumor suppressor acts remain incompletely understood. To gain new insights into p53 biology, we used high-throughput sequencing to analyze global p53 transcriptional networks in primary mouse embryo fibroblasts in response to DNA damage. Chromatin immunoprecipitation sequencing reveals 4785 p53-bound sites in the genome located near 3193 genes involved in diverse biological processes. RNA sequencing analysis shows that only a subset of p53-bound genes is transcriptionally regulated, yielding a list of 432 p53-bound and regulated genes. Interestingly, we identify a host of autophagy genes as direct p53 target genes. While the autophagy program is regulated predominantly by p53, the p53 family members p63 and p73 contribute to activation of this autophagy gene network. Induction of autophagy genes in response to p53 activation is associated with enhanced autophagy in diverse settings and depends on p53 transcriptional activity. While p53-induced autophagy does not affect cell cycle arrest in response to DNA damage, it is important for both robust p53-dependent apoptosis triggered by DNA damage and transformation suppression by p53. Together, our data highlight an intimate connection between p53 and autophagy through a vast transcriptional network and indicate that autophagy contributes to p53-dependent apoptosis and cancer suppression.


Subject(s)
Autophagy/genetics , DNA Damage/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Animals , Cell Cycle Checkpoints/genetics , Cell Survival/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Genome-Wide Association Study , Mice , Protein Binding , Sequence Analysis, RNA
5.
Proc Natl Acad Sci U S A ; 112(14): 4441-6, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25792458

ABSTRACT

Scaffold proteins are critical hubs within cells that have the ability to modulate upstream signaling molecules and their downstream effectors to fine-tune biological responses. Although they can serve as focal points for association of signaling molecules and downstream pathways that regulate tumorigenesis, little is known about how the tumor microenvironment affects the expression and activity of scaffold proteins. This study demonstrates that hypoxia, a common element of solid tumors harboring low oxygen levels, regulates expression of a specific variant of the scaffold protein AKAP12 (A-kinase anchor protein 12), AKAP12v2, in metastatic melanoma. In turn, through a kinome-wide phosphoproteomic and MS study, we demonstrate that this scaffolding protein regulates a shift in protein kinase A (PKA)-mediated phosphorylation events under hypoxia, causing alterations in tumor cell invasion and migration in vitro, as well as metastasis in an in vivo orthotopic model of melanoma. Mechanistically, the shift in AKAP12-dependent PKA-mediated phosphorylations under hypoxia is due to changes in AKAP12 localization vs. structural differences between its two variants. Importantly, our work defines a mechanism through which a scaffold protein can be regulated by the tumor microenvironment and further explains how a tumor cell can coordinate many critical signaling pathways that are essential for tumor growth through one individual scaffolding protein.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/pathology , Skin Neoplasms/metabolism , A Kinase Anchor Proteins/genetics , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Humans , Melanoma/metabolism , Mice , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Oxygen/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proteomics , Signal Transduction
6.
Wound Repair Regen ; 25(6): 964-971, 2017 11.
Article in English | MEDLINE | ID: mdl-29316036

ABSTRACT

Wound healing is characterized by the production of large amounts of protein necessary to replace lost cellular mass and extracellular matrix. The unfolded protein response (UPR) is an important adaptive cellular response to increased protein synthesis. One of the main components of the UPR is IRE1, an endoplasmic reticulum transmembrane protein with endonuclease activity that produces the activated form of the transcription factor XBP1. Using luciferase reporter mice for Xbp1 splicing, we showed that IRE1 was up-regulated during excisional wound healing at the time in wound healing consistent with that of the proliferative phase, when the majority of protein synthesis for cellular proliferation and matrix deposition occurs. Furthermore, using a small molecule inhibitor of IRE1 we demonstrated that inhibition of IRE1 led to decreased scar formation in treated mice. Results were recapitulated in a hypertrophic scar mouse model. These data help provide a cellular pathway to target in the treatment of hypertrophic scarring and keloid disorders.


Subject(s)
Cicatrix, Hypertrophic/metabolism , Cicatrix/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , X-Box Binding Protein 1/metabolism , Animals , Cell Proliferation , Extracellular Matrix/metabolism , Membrane Proteins/antagonists & inhibitors , Mice , Protein Serine-Threonine Kinases/antagonists & inhibitors , Unfolded Protein Response , Up-Regulation , Wound Healing
7.
J Chem Inf Model ; 57(4): 875-882, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28257191

ABSTRACT

We describe a new library generation method, Machine-based Identification of Molecules Inside Characterized Space (MIMICS), that generates sets of molecules inspired by a text-based input. MIMICS-generated libraries were found to preserve distributions of properties while simultaneously increasing structural diversity. Newly identified MIMICS-generated compounds were found to be bioactive as inhibitors of specific components of the unfolded protein response (UPR) and the VEGFR2 pathway in cell-based assays, thus confirming the applicability of this methodology toward drug design applications. Wider application of MIMICS could facilitate the efficient utilization of chemical space.


Subject(s)
Drug Discovery/methods , Neural Networks, Computer , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
8.
Semin Cancer Biol ; 33: 48-56, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25986851

ABSTRACT

Accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER) leads to ER stress, which is characteristic of cells with high level of secretory activity and implicated in a variety of disease conditions. In response to ER stress, the cell elicits an adaptive process called the unfolded protein response (UPR) to support cellular homeostasis and survival. However, prolonged and unsolvable ER stress also induces apoptosis. As the most conserved signaling branch of the UPR, the IRE1α-XBP1 pathway plays important roles in both physiological and pathological settings and its activity has profound effects on disease progression and prognosis. Recently, modulating this pathway with small molecule compounds has been demonstrated as a promising approach for disease therapy. In this review, we summarize a list of current investigational compounds targeting this pathway and their therapeutic features for treating human diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Unfolded Protein Response , Aldehydes/chemistry , Aldehydes/metabolism , Animals , Apoptosis , Cell Survival , Coumarins/chemistry , Disease Models, Animal , Disease Progression , Drug Design , Drug Discovery , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Homeostasis , Humans , Mice , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Prognosis , Protein Folding , Quercetin/chemistry , Regulatory Factor X Transcription Factors , Signal Transduction , Sulfonamides , Thiophenes , Toyocamycin/chemistry , X-Box Binding Protein 1
9.
Adv Exp Med Biol ; 899: 253-68, 2016.
Article in English | MEDLINE | ID: mdl-27325272

ABSTRACT

In silico drug discovery refers to a combination of computational techniques that augment our ability to discover drug compounds from compound libraries. Many such techniques exist, including virtual high-throughput screening (vHTS), high-throughput screening (HTS), and mechanisms for data storage and querying. However, presently these tools are often used independent of one another. In this chapter, we describe a new multimodal in silico technique for the hit identification and lead generation phases of traditional drug discovery. Our technique leverages the benefits of three independent methods-virtual high-throughput screening, high-throughput screening, and structural fingerprint analysis-by using a fourth technique called topological data analysis (TDA). We describe how a compound library can be independently tested with vHTS, HTS, and fingerprint analysis, and how the results can be transformed into a topological data analysis network to identify compounds from a diverse group of structural families. This process of using TDA or similar clustering methods to identify drug leads is advantageous because it provides a mechanism for choosing structurally diverse compounds while maintaining the unique advantages of already established techniques such as vHTS and HTS.


Subject(s)
Drug Discovery/methods , Statistics as Topic , High-Throughput Screening Assays , Small Molecule Libraries/analysis , User-Computer Interface
10.
Proc Natl Acad Sci U S A ; 108(41): 17123-8, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21969549

ABSTRACT

Over half of all human cancers, of a wide variety of types, sustain mutations in the p53 tumor suppressor gene. Although p53 limits tumorigenesis through the induction of apoptosis or cell cycle arrest, its molecular mechanism of action in tumor suppression has been elusive. The best-characterized p53 activity in vitro is as a transcriptional activator, but the identification of numerous additional p53 biochemical activities in vitro has made it unclear which mechanism accounts for tumor suppression. Here, we assess the importance of transcriptional activation for p53 tumor suppression function in vivo in several tissues, using a knock-in mouse strain expressing a p53 mutant compromised for transcriptional activation, p53(25,26). p53(25,26) is severely impaired for the transactivation of numerous classical p53 target genes, including p21, Noxa, and Puma, but it retains the ability to activate a small subset of p53 target genes, including Bax. Surprisingly, p53(25,26) can nonetheless suppress tumor growth in cancers derived from the epithelial, mesenchymal, central nervous system, and lymphoid lineages. Therefore, full transactivation of most p53 target genes is dispensable for p53 tumor suppressor function in a range of tissue types. In contrast, a transcriptional activation mutant that is completely defective for transactivation, p53(25,26,53,54), fails to suppress tumor development. These findings demonstrate that transcriptional activation is indeed broadly critical for p53 tumor suppressor function, although this requirement reflects the limited transcriptional activity observed with p53(25,26) rather than robust transactivation of a full complement of p53 target genes.


Subject(s)
Genes, p53 , Neoplasms/genetics , Neoplasms/prevention & control , Animals , Cell Lineage/genetics , Gene Knock-In Techniques , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/prevention & control , Medulloblastoma/genetics , Medulloblastoma/prevention & control , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Transcriptional Activation
11.
Nat Commun ; 15(1): 4114, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750057

ABSTRACT

Cellular sensitivity to ferroptosis is primarily regulated by mechanisms mediating lipid hydroperoxide detoxification. We show that inositol-requiring enzyme 1 (IRE1α), an endoplasmic reticulum (ER) resident protein critical for the unfolded protein response (UPR), also determines cellular sensitivity to ferroptosis. Cancer and normal cells depleted of IRE1α gain resistance to ferroptosis, while enhanced IRE1α expression promotes sensitivity to ferroptosis. Mechanistically, IRE1α's endoribonuclease activity cleaves and down-regulates the mRNA of key glutathione biosynthesis regulators glutamate-cysteine ligase catalytic subunit (GCLC) and solute carrier family 7 member 11 (SLC7A11). This activity of IRE1α is independent of its role in regulating the UPR and is evolutionarily conserved. Genetic deficiency and pharmacological inhibition of IRE1α have similar effects in inhibiting ferroptosis and reducing renal ischemia-reperfusion injury in mice. Our findings reveal a previously unidentified role of IRE1α to regulate ferroptosis and suggests inhibition of IRE1α as a promising therapeutic strategy to mitigate ferroptosis-associated pathological conditions.


Subject(s)
Amino Acid Transport System y+ , Endoribonucleases , Ferroptosis , Glutathione , Protein Serine-Threonine Kinases , Ferroptosis/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Animals , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mice , Glutathione/metabolism , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Glutamate-Cysteine Ligase/metabolism , Glutamate-Cysteine Ligase/genetics , Unfolded Protein Response , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Cell Line, Tumor , Mice, Inbred C57BL , Male , Mice, Knockout
12.
Nat Biomed Eng ; 8(5): 593-610, 2024 May.
Article in English | MEDLINE | ID: mdl-38641710

ABSTRACT

Intracellular DNA sensors regulate innate immunity and can provide a bridge to adaptive immunogenicity. However, the activation of the sensors in antigen-presenting cells (APCs) by natural agonists such as double-stranded DNAs or cyclic nucleotides is impeded by poor intracellular delivery, serum stability, enzymatic degradation and rapid systemic clearance. Here we show that the hydrophobicity, electrostatic charge and secondary conformation of helical polypeptides can be optimized to stimulate innate immune pathways via endoplasmic reticulum stress in APCs. One of the three polypeptides that we engineered activated two major intracellular DNA-sensing pathways (cGAS-STING (for cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes) and Toll-like receptor 9) preferentially in APCs by promoting the release of mitochondrial DNA, which led to the efficient priming of effector T cells. In syngeneic mouse models of locally advanced and metastatic breast cancers, the polypeptides led to potent DNA-sensor-mediated antitumour responses when intravenously given as monotherapy or with immune checkpoint inhibitors. The activation of multiple innate immune pathways via engineered cationic polypeptides may offer therapeutic advantages in the generation of antitumour immune responses.


Subject(s)
Antigen-Presenting Cells , Immunity, Innate , Peptides , Animals , Immunity, Innate/drug effects , Peptides/chemistry , Peptides/pharmacology , Mice , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/drug effects , Humans , Female , Cations/chemistry , Mice, Inbred C57BL , Cell Line, Tumor , Toll-Like Receptor 9/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/drug therapy , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/chemistry
13.
PLoS Genet ; 6(10): e1001168, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20975948

ABSTRACT

Dysregulated cell-cell adhesion plays a critical role in epithelial cancer development. Studies of human and mouse cancers have indicated that loss of adhesion complexes known as adherens junctions contributes to tumor progression and metastasis. In contrast, little is known regarding the role of the related cell-cell adhesion junction, the desmosome, during cancer development. Studies analyzing expression of desmosome components during human cancer progression have yielded conflicting results, and therefore genetic studies using knockout mice to examine the functional consequence of desmosome inactivation for tumorigenesis are essential for elucidating the role of desmosomes in cancer development. Here, we investigate the consequences of desmosome loss for carcinogenesis by analyzing conditional knockout mice lacking Perp, a p53/p63 regulated gene that encodes an important component of desmosomes. Analysis of Perp-deficient mice in a UVB-induced squamous cell skin carcinoma model reveals that Perp ablation promotes both tumor initiation and progression. Tumor development is associated with inactivation of both of Perp's known functions, in apoptosis and cell-cell adhesion. Interestingly, Perp-deficient tumors exhibit widespread downregulation of desmosomal constituents while adherens junctions remain intact, suggesting that desmosome loss is a specific event important for tumorigenesis rather than a reflection of a general change in differentiation status. Similarly, human squamous cell carcinomas display loss of PERP expression with retention of adherens junctions components, indicating that this is a relevant stage of human cancer development. Using gene expression profiling, we show further that Perp loss induces a set of inflammation-related genes that could stimulate tumorigenesis. Together, these studies suggest that Perp-deficiency promotes cancer by enhancing cell survival, desmosome loss, and inflammation, and they highlight a fundamental role for Perp and desmosomes in tumor suppression. An understanding of the factors affecting cancer progression is important for ultimately improving the diagnosis, prognostication, and treatment of cancer.


Subject(s)
Carcinoma, Squamous Cell/genetics , Desmosomes/metabolism , Membrane Proteins/genetics , Skin Neoplasms/genetics , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Cadherins/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Male , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Skin/immunology , Skin/pathology , Skin/radiation effects , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tissue Array Analysis , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays
14.
J Clin Invest ; 133(23)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37824211

ABSTRACT

An immunosuppressive microenvironment causes poor tumor T cell infiltration and is associated with reduced patient overall survival in colorectal cancer. How to improve treatment responses in these tumors is still a challenge. Using an integrated screening approach to identify cancer-specific vulnerabilities, we identified complement receptor C5aR1 as a druggable target, which when inhibited improved radiotherapy, even in tumors displaying immunosuppressive features and poor CD8+ T cell infiltration. While C5aR1 is well-known for its role in the immune compartment, we found that C5aR1 is also robustly expressed on malignant epithelial cells, highlighting potential tumor cell-specific functions. C5aR1 targeting resulted in increased NF-κB-dependent apoptosis specifically in tumors and not normal tissues, indicating that, in malignant cells, C5aR1 primarily regulated cell fate. Collectively, these data revealed that increased complement gene expression is part of the stress response mounted by irradiated tumors and that targeting C5aR1 could improve radiotherapy, even in tumors displaying immunosuppressive features.


Subject(s)
Complement C5a , Receptors, Complement , Humans , Complement C5a/genetics , Receptors, Complement/genetics
15.
Sci Adv ; 8(19): eabm6638, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35559673

ABSTRACT

Exploiting cancer vulnerabilities is critical for the discovery of anticancer drugs. However, tumor suppressors cannot be directly targeted because of their loss of function. To uncover specific vulnerabilities for cells with deficiency in any given tumor suppressor(s), we performed genome-scale CRISPR loss-of-function screens using a panel of isogenic knockout cells we generated for 12 common tumor suppressors. Here, we provide a comprehensive and comparative dataset for genetic interactions between the whole-genome protein-coding genes and a panel of tumor suppressor genes, which allows us to uncover known and new high-confidence synthetic lethal interactions. Mining this dataset, we uncover essential paralog gene pairs, which could be a common mechanism for interpreting synthetic lethality. Moreover, we propose that some tumor suppressors could be targeted to suppress proliferation of cells with deficiency in other tumor suppressors. This dataset provides valuable information that can be further exploited for targeted cancer therapy.


Subject(s)
Antineoplastic Agents , Neoplasms , CRISPR-Cas Systems , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Genes, Tumor Suppressor , Humans , Neoplasms/genetics , Synthetic Lethal Mutations
16.
J Exp Med ; 219(9)2022 09 05.
Article in English | MEDLINE | ID: mdl-35881112

ABSTRACT

Disease relapse and treatment-induced immunotoxicity pose significant clinical challenges for patients with hematological cancers. Here, we reveal distinctive requirements for neutralizing TNF receptor ligands APRIL and BAFF and their receptor activity in MM and DLBCL, impacting protein translation and production in MM cells and modulating the translation efficiency of the ATM interactor (ATMIN/ACSIZ). Therapeutically, we investigated the use of BCMA decoy receptor (sBCMA-Fc) as an inhibitor of APRIL and BAFF. While wild-type sBCMA-Fc effectively blocked APRIL signaling in MM, it lacked activity in DLBCL due to its weak BAFF binding. To expand the therapeutic utility of sBCMA-Fc, we engineered an affinity-enhanced mutant sBCMA-Fc fusion molecule (sBCMA-Fc V3) 4- and 500-fold stronger in binding to APRIL and BAFF, respectively. The mutant sBCMA-Fc V3 clone significantly enhanced antitumor activity against both MM and DLBCL. Importantly, we also demonstrated an adequate toxicity profile and on-target mechanism of action in nonhuman primate studies.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Multiple Myeloma , Animals , B-Cell Activating Factor/genetics , B-Cell Maturation Antigen/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Signal Transduction , Transmembrane Activator and CAML Interactor Protein , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
17.
Phys Med Biol ; 67(16)2022 08 05.
Article in English | MEDLINE | ID: mdl-35853442

ABSTRACT

Objective. Irradiation with ultra-high dose rates (>40 Gy s-1), also known as FLASH irradiation, has the potential to shift the paradigm of radiation therapy because of its reduced toxicity to normal tissues compared to that of conventional irradiations. The goal of this study was to (1) achieve FLASH irradiation conditions suitable for pre-clinicalin vitroandin vivobiology experiments using our synchrotron-based proton beamline and (2) commission the FLASH irradiation conditions achieved.Approach. To achieve these suitable FLASH conditions, we made a series of adaptations to our proton beamline, including modifying the spill length and size of accelerating cycles, repurposing the reference monitor for dose control, and expanding the field size with a custom double-scattering system. We performed the dosimetric commissioning with measurements using an Advanced Markus chamber and EBT-XD films as well as with Monte Carlo simulations.Main results. Through adaptations, we have successfully achieved FLASH irradiation conditions, with an average dose rate of up to 375 Gy s-1. The Advanced Markus chamber was shown to be appropriate for absolute dose calibration under our FLASH conditions with a recombination factor ranging from 1.002 to 1.006 because of the continuous nature of our synchrotron-based proton delivery within a spill. Additionally, the absolute dose measured using the Advanced Markus chamber and EBT-XD films agreed well, with average and maximum differences of 0.32% and 1.63%, respectively. We also performed a comprehensive temporal analysis for FLASH spills produced by our system, which helped us identify a unique relationship between the average dose rate and the dose in our FLASH irradiation.Significance.We have established a synchrotron-based proton FLASH irradiation platform with accurate and precise dosimetry that is suitable for pre-clinical biology experiments. The unique time structure of the FLASH irradiation produced by our synchrotron-based system may shed new light onto the mechanism behind the FLASH effect.


Subject(s)
Proton Therapy , Protons , Proton Therapy/methods , Radiometry , Radiotherapy Dosage , Synchrotrons
18.
PNAS Nexus ; 1(2): pgac056, 2022 May.
Article in English | MEDLINE | ID: mdl-35707206

ABSTRACT

Adult salivary stem/progenitor cells (SSPC) have an intrinsic property to self-renew in order to maintain tissue architecture and homeostasis. Adult salivary glands have been documented to harbor SSPC, which have been shown to play a vital role in the regeneration of the glandular structures postradiation damage. We have previously demonstrated that activation of aldehyde dehydrogenase 3A1 (ALDH3A1) after radiation reduced aldehyde accumulation in SSPC, leading to less apoptosis and improved salivary function. We subsequently found that sustained pharmacological ALDH3A1 activation is critical to enhance regeneration of murine submandibular gland after radiation damage. Further investigation shows that ALDH3A1 function is crucial for SSPC self-renewal and survival even in the absence of radiation stress. Salivary glands from Aldh3a1 -/- mice have fewer acinar structures than wildtype mice. ALDH3A1 deletion or pharmacological inhibition in SSPC leads to a decrease in mitochondrial DNA copy number, lower expression of mitochondrial specific genes and proteins, structural abnormalities, lower membrane potential, and reduced cellular respiration. Loss or inhibition of ALDH3A1 also elevates ROS levels, depletes glutathione pool, and accumulates ALDH3A1 substrate 4-hydroxynonenal (4-HNE, a lipid peroxidation product), leading to decreased survival of murine SSPC that can be rescued by treatment with 4-HNE specific carbonyl scavengers. Our data indicate that ALDH3A1 activity protects mitochondrial function and is important for the regeneration activity of SSPC. This knowledge will help to guide our translational strategy of applying ALDH3A1 activators in the clinic to prevent radiation-related hyposalivation in head and neck cancer patients.

19.
Clin Cancer Res ; 26(12): 2972-2985, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32098769

ABSTRACT

PURPOSE: To investigate how induced tumor heterogeneity influences immune responses to radiotherapy with different proportions of mixed immune-responsive and unresponsive tumor cells in a triple-negative breast cancer model. It is hypothesized that studying the immune environment of mixed tumors and responses to radiotherapy could nominate immune active therapies to enhance immune responses after radiotherapy. EXPERIMENTAL DESIGN: Evaluate efficacy and immune responses generated by radiotherapy in tumors with different proportions of immunologically responsive and unresponsive tumor cells. Then study the cellular responses and transcriptomic differences between the tumors to nominate immunotherapy combinations with radiotherapy and evaluate the combination. RESULTS: The addition of the responsive cells to unresponsive tumors led to a greater than expected therapeutic response to radiotherapy with both innate and adaptive immune components. There was a distinct change in myeloid cells, greater inflammatory macrophage activity, and enhanced antigen presentation with responsive cells after radiotherapy. Because differences in matrix components, cell adhesion biology, and innate immune signaling correlated with myeloid cell response and phenotype, we hypothesized that radiotherapy combined with CD40 agonist antibody would sensitize unresponsive tumors. The combination therapy resulted in improved innate and adaptive immune response. Importantly, CD40 treatment increased tumor response to radiotherapy and protected against metastatic spread in a metastatic model. CONCLUSIONS: These data combined with transcriptomics from human patients support radiotherapy and myeloid cell targeting for immunologically cold tumors. The established study model presents opportunities to investigate the complex overlapping biologic mechanisms that limit immunotherapy and to implement radiotherapy with different immunotherapy combinations.


Subject(s)
Breast Neoplasms/pathology , Immunotherapy/mortality , Radioimmunotherapy/mortality , Radiotherapy/mortality , Animals , Apoptosis , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Cell Proliferation , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Clin Cancer Res ; 25(10): 2969-2974, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30723144

ABSTRACT

The development of cancers and their response to radiation are intricately linked to the tumor microenvironment (TME) in which they reside. Tumor cells, immune cells, and stromal cells interact with each other and are influenced by the microbiome and metabolic state of the host, and these interactions are constantly evolving. Stromal cells not only secrete extracellular matrix and participate in wound contraction, but they also secrete fibroblast growth factors (FGF), which mediate macrophage differentiation. Tumor-associated macrophages migrate to hypoxic areas and secrete vascular endothelial growth factor (VEGF) to promote angiogenesis. The microbiome and its byproducts alter the metabolic milieu by shifting the balance between glucose utilization and fatty acid oxidation, and these changes subsequently influence the immune response in the TME. Not only does radiation exert cell-autonomous effects on tumor cells, but it influences both the tumor-promoting and tumor-suppressive components in the TME. To gain a deeper understanding of how the TME influences the response to radiation, the American Society for Radiation Oncology and the American Association of Cancer Research organized a scientific workshop on July 26-27, 2018, to discuss how the microbiome, the immune response, the metabolome, and the stroma all shift the balance between radiosensitivity and radioresistance. The proceedings from this workshop are discussed here and highlight recent discoveries in the field, as well as the most important areas for future research.


Subject(s)
Neoplasms/pathology , Neoplasms/radiotherapy , Radiation Oncology , Tumor Microenvironment/radiation effects , Disease Management , Humans , Immunomodulation/radiation effects , Neoplasms/immunology , Radiation Oncology/methods , Risk Factors , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/radiation effects , Treatment Outcome , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL