Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.722
Filter
1.
J Mol Biol ; 436(1): 168372, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37979908

ABSTRACT

Brca1 mouse models were first reported in the mid-1990's shortly after cloning the human gene. Since then, many mouse models with a range of mutations have been generated, some mimic patient mutations, others are designed to probe specific protein domains and functions. In this review, we discuss early and recent studies using engineered Brca1 mouse alleles, and their implications for understanding Brca1 protein function in the context of DNA repair, tumorigenesis, and anti-cancer therapeutics.


Subject(s)
BRCA1 Protein , Neoplasms, Experimental , Animals , Humans , Mice , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , DNA Repair , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics
2.
Proc Natl Acad Sci U S A ; 119(29): e2202015119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858326

ABSTRACT

Epigenetic dysregulation is a universal feature of cancer that results in altered patterns of gene expression that drive malignancy. Brain tumors exhibit subtype-specific epigenetic alterations; however, the molecular mechanisms responsible for these diverse epigenetic states remain unclear. Here, we show that the developmental transcription factor Sox9 differentially regulates epigenomic states in high-grade glioma (HGG) and ependymoma (EPN). Using our autochthonous mouse models, we found that Sox9 suppresses HGG growth and expands associated H3K27ac states, while promoting ZFTA-RELA (ZRFUS) EPN growth and diminishing H3K27ac states. These contrasting roles for Sox9 correspond with protein interactions with histone deacetylating complexes in HGG and an association with the ZRFUS oncofusion in EPN. Mechanistic studies revealed extensive Sox9 and ZRFUS promoter co-occupancy, indicating functional synergy in promoting EPN tumorigenesis. Together, our studies demonstrate how epigenomic states are differentially regulated in distinct subtypes of brain tumors, while revealing divergent roles for Sox9 in HGG and EPN tumorigenesis.


Subject(s)
Brain Neoplasms , Ependymoma , Epigenesis, Genetic , SOX9 Transcription Factor , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carcinogenesis/genetics , Ependymoma/genetics , Ependymoma/pathology , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/physiology
4.
J Clin Invest ; 132(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35104810

ABSTRACT

PD-1 signaling on T cells is the major pathway that limits T cell immunity, but the efficacy of anti-PD-1 therapy has been limited to a small proportion of patients with advanced cancers. We fortuitously observed that anti-PD-1 therapy depends on IL-2 signaling, which raises the possibility that a lack of IL-2 limits anti-PD-1-induced effector T cell expansion. To selectively deliver IL-2 to PD-1+CD8+ tumor-infiltrating lymphocytes (TILs), we engineered a low-affinity IL-2 paired with anti-PD-1 (PD-1-laIL-2), which reduced affinity to peripheral Treg cells but enhanced avidity to PD-1+CD8+ TILs. PD-1-laIL-2 exerted better tumor control and lower toxicity than single or mixed treatments. Mechanistically, PD-1-laIL-2 could effectively expand dysfunctional and tumor-specific CD8+ T cells. Furthermore, we discovered that presumably dysfunctional PD-1+TIM3+ TILs are the dominant tumor-specific T cells responding to PD-1-laIL-2. Collectively, these results highlight that PD-1-laIL-2 can target and reactivate tumor-specific TILs for tumor regression as a unique strategy with stronger efficacy and lower toxicity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular/drug effects , Interleukin-2/pharmacology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Immunity, Cellular/genetics , Interleukin-2/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Programmed Cell Death 1 Receptor/genetics
5.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35110400

ABSTRACT

Oncogenic imbalance of DNA methylation is well recognized in cancer development. The ten-eleven translocation (TET) family of dioxygenases, which facilitates DNA demethylation, is frequently dysregulated in cancers. How such dysregulation contributes to tumorigenesis remains poorly understood, especially in solid tumors which present infrequent mutational incidence of TET genes. Here, we identify loss-of-function mutations of TET in 7.4% of human lung adenocarcinoma (LUAD), which frequently co-occur with oncogenic KRAS mutations, and this co-occurrence is predictive of poor survival in LUAD patients. Using an autochthonous mouse model of KrasG12D -driven LUAD, we show that individual or combinational loss of Tet genes markedly promotes tumor development. In this Kras-mutant and Tet-deficient model, the premalignant lung epithelium undergoes neoplastic reprogramming of DNA methylation and transcription, with a particular impact on Wnt signaling. Among the Wnt-associated components that undergo reprogramming, multiple canonical Wnt antagonizing genes present impaired expression arising from elevated DNA methylation, triggering aberrant activation of Wnt signaling. These impairments can be largely reversed upon the restoration of TET activity. Correspondingly, genetic depletion of ß-catenin, the transcriptional effector of Wnt signaling, substantially reverts the malignant progression of Tet-deficient LUAD. These findings reveal TET enzymes as critical epigenetic barriers against lung tumorigenesis and highlight the therapeutic vulnerability of TET-mutant lung cancer through targeting Wnt signaling.


Subject(s)
Adenocarcinoma of Lung/metabolism , DNA Methylation , DNA, Neoplasm/metabolism , Lung Neoplasms/metabolism , Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins/deficiency , Wnt Signaling Pathway , Adenocarcinoma of Lung/genetics , Animals , DNA, Neoplasm/genetics , Humans , Lung Neoplasms/genetics , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Proto-Oncogene Proteins/metabolism
6.
J Clin Invest ; 132(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35166238

ABSTRACT

Extracellular proteolysis is frequently dysregulated in disease and can generate proteoforms with unique neoepitopes not found in healthy tissue. Here, we demonstrate that Abs that selectively recognize a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) could enable more effective and safer treatments for solid tumors. CDCP1 is highly overexpressed in RAS-driven cancers, and its ectodomain is cleaved by extracellular proteases. Biochemical, biophysical, and structural characterization revealed that the 2 cleaved fragments of CDCP1 remain tightly associated with minimal proteolysis-induced conformational change. Using differential phage display, we generated recombinant Abs that are exquisitely selective to cleaved CDCP1 with no detectable binding to the uncleaved form. These Abs potently targeted cleaved CDCP1-expressing cancer cells as an Ab-drug conjugate, an Ab-radionuclide conjugate, and a bispecific T cell engager. In a syngeneic pancreatic tumor model, these cleaved-specific Abs showed tumor-specific localization and antitumor activity with superior safety profiles compared with a pan-CDCP1 approach. Targeting proteolytic neoepitopes could provide an orthogonal "AND" gate for improving the therapeutic index.


Subject(s)
Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Epitopes/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Pancreatic Neoplasms/immunology , Proteolysis , Animals , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Epitopes/genetics , Humans , Male , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Pancreatic Neoplasms/genetics
7.
Bioengineered ; 13(2): 3183-3193, 2022 02.
Article in English | MEDLINE | ID: mdl-35068336

ABSTRACT

Dysregulation of calcium-activated nucleotides 1 (CANT1) has been observed in different organs. Thus, its biological function in cancer has increasingly attracted researchers. The current work aims to study the CANT1 role in lung cancer and understand the underlying pathological mechanisms. High amplification of CANT1 was observed in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) tissues compared to normal tissues. The high-CANT1 patients showed a dismal prognosis in comparison with the low-CANT1 patients. Highly expressed CANT1 was significantly associated with the N stage of LUSC patients. Ectopic expression of CANT1 conspicuously increased the proliferation and viability of A549 cells. Conversely, CANT1 depletion resulted in adverse effects in H1299 cells. CANT1 depletion also resulted in the retardation of tumor growth in vivo. Mechanically, we found that CANT1 could elevate NF-ĸB (nuclear factor-k-gene binding) transcriptional activity in a concentration-dependent manner. This regulatory relationship was also established by the Western blot technique. Inhibiting NF-ĸB can significantly blunt the increased NF-κ-B Inhibitor-α (IκBα) expression caused by CANT1 overexpression in A549 cells. In conclusion, highly amplified CANT1 promotes the proliferation and viability of lung cancer cells. We also elucidate a new signaling axis of CANT1-NF-ĸB in lung cancer. This approach might be a promising strategy for lung cancer treatment.


Subject(s)
Lung Neoplasms/metabolism , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Nucleotidases/metabolism , Signal Transduction , A549 Cells , Animals , Humans , Lung Neoplasms/genetics , Male , Mice , Mice, Nude , NF-kappa B/genetics , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Nucleotidases/genetics
8.
Toxicol Pathol ; 50(2): 232-234, 2022 02.
Article in English | MEDLINE | ID: mdl-34747286

ABSTRACT

Non-glandular squamous cell carcinoma (NGSCC) is an extremely rare tumor in Tg.raH2 mice. There have been 5 NGSCC in 1615 control male mice (0.31%) and 2 NGSCC in 1560 control female mice (0.13%) on 26-week carcinogenicity studies, with a range of 0 to 1 of per group per sex in each study without statistical significance in 52 male and 51 female studies conducted in Tg.rasH2 mice. Every case of NGSCC was accompanied by profound granulocytosis.


Subject(s)
Carcinoma, Squamous Cell , Neoplasms, Experimental , Paraneoplastic Syndromes , Animals , Carcinogenicity Tests , Female , Genes, ras , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics
9.
Am J Pathol ; 192(2): 353-360, 2022 02.
Article in English | MEDLINE | ID: mdl-34774516

ABSTRACT

Although the uterine cervix responds to the female sex hormone change, the role of progesterone in cervical cancer is poorly understood. It has been shown that medroxyprogesterone acetate (MPA) regresses cervical cancer in the transgenic mouse model expressing human papillomavirus type 16 E6 and E7 oncogenes. As MPA interacts most strongly with progesterone receptor (PR), we reasoned that PR would contribute to MPA-induced regression of cervical cancer. We also hypothesized that estrogen influences the therapeutic activity of MPA because it promotes cervical cancer growth in the same mouse model. The present study showed that the deletion of Pgr in the cervical cancer cells ablated the MPA's therapeutic effect in the human papillomavirus transgenic mouse model. Additionally, estrogen attenuated cancer regression by MPA in the same model system. These observations indicate that MPA can effectively regress cervical cancer only when cancer cells express PR and estrogen levels are low. These results suggest that, if translatable, MPA should be administered when estrogen levels are low in patients with PR-positive cervical cancer.


Subject(s)
Epithelial Cells , Estrogens/metabolism , Neoplasm Proteins , Neoplasms, Experimental , Progestins/pharmacology , Receptors, Progesterone , Uterine Cervical Neoplasms , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Deletion , Male , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
10.
JCI Insight ; 7(2)2022 01 25.
Article in English | MEDLINE | ID: mdl-34905507

ABSTRACT

Programmed death-1 homolog (PD-1H) is a coinhibitory molecule that negatively regulates T cell-mediated immune responses. In this study, we determined whether ablation of T cell-associated PD-1H could enhance adoptive T cell therapy in experimental tumor models. The expression of PD-1H is upregulated in activated and tumor-infiltrating CD8+ T cells. Activated CD8+ T cells from PD-1H-deficient (PD-1H-KO) mice exhibited increased cell proliferation, cytokine production, and antitumor activity in vitro. Adoptive transfer of PD-1H-KO CD8+ T cells resulted in the regression of established syngeneic mouse tumors. Similar results were obtained when PD-1H was ablated in T cells by CRISPR/Cas9-mediated gene silencing. Furthermore, ablation of PD-1H in CAR-T cells significantly improved their antitumor activity against human xenografts in vivo. Our results indicate that T cell-associated PD-1H could suppress immunity in the tumor microenvironment and that targeting PD-1H may improve T cell adoptive immunotherapy.


Subject(s)
Adoptive Transfer/methods , CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Membrane Proteins , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Cell Proliferation , Cytokines/biosynthesis , Gene Silencing , Gene Targeting/methods , Humans , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Mice , Neoplasms, Experimental/genetics , Xenograft Model Antitumor Assays
11.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34825915

ABSTRACT

Targeted therapies represent attractive combination partners with immune checkpoint blockade (ICB) to increase the population of patients who benefit or to interdict the emergence of resistance. We demonstrate that targeting WEE1 up-regulates immune signaling through the double-stranded RNA (dsRNA) viral defense pathway with subsequent responsiveness to immune checkpoint blockade even in cGAS/STING-deficient tumors, which is a typical phenotype across multiple cancer types. WEE1 inhibition increases endogenous retroviral elements (ERVs) expression by relieving SETDB1/H3K9me3 repression through down-regulating FOXM1. ERVs trigger dsRNA stress and interferon response, increasing recruitment of anti-tumor T cells with concurrent PD-L1 elevation in multiple tumor models. Furthermore, combining WEE1 inhibition and PD-L1 blockade induced striking tumor regression in a CD8+ T cell-dependent manner. A WEE1 inhibition-induced viral defense signature provides a potentially informative biomarker for patient selection for combination therapy with WEE1 and ICB. WEE1 inhibition stimulates anti-tumor immunity and enhances sensitivity to ICB, providing a rationale for the combination of WEE1 inhibitors and ICB in clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Endogenous Retroviruses/genetics , Neoplasms, Experimental/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Double-Stranded/genetics , Signal Transduction/drug effects , A549 Cells , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Endogenous Retroviruses/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/pharmacology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacology , RNA, Double-Stranded/metabolism , Signal Transduction/genetics , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Burden/immunology
12.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34969858

ABSTRACT

Brain metastases are a leading cause of death in patients with breast cancer. The lack of clinical trials and the presence of the blood-brain barrier limit therapeutic options. Furthermore, overexpression of the human epidermal growth factor receptor 2 (HER2) increases the incidence of breast cancer brain metastases (BCBM). HER2-targeting agents, such as the monoclonal antibodies trastuzumab and pertuzumab, improved outcomes in patients with breast cancer and extracranial metastases. However, continued BCBM progression in breast cancer patients highlighted the need for novel and effective targeted therapies against intracranial metastases. In this study, we engineered the highly migratory and brain tumor tropic human neural stem cells (NSCs) LM008 to continuously secrete high amounts of functional, stable, full-length antibodies against HER2 (anti-HER2Ab) without compromising the stemness of LM008 cells. The secreted anti-HER2Ab impaired tumor cell proliferation in vitro in HER2+ BCBM cells by inhibiting the PI3K-Akt signaling pathway and resulted in a significant benefit when injected in intracranial xenograft models. In addition, dual HER2 blockade using anti-HER2Ab LM008 NSCs and the tyrosine kinase inhibitor tucatinib significantly improved the survival of mice in a clinically relevant model of multiple HER2+ BCBM. These findings provide compelling evidence for the use of HER2Ab-secreting LM008 NSCs in combination with tucatinib as a promising therapeutic regimen for patients with HER2+ BCBM.


Subject(s)
Antineoplastic Agents, Immunological/metabolism , Brain Neoplasms , Neoplasms, Experimental , Neural Stem Cells , Oxazoles/pharmacology , Pyridines/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2 , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays
13.
Front Immunol ; 12: 770080, 2021.
Article in English | MEDLINE | ID: mdl-34925340

ABSTRACT

Enhancer of Zeste Homolog 2 (EZH2) inhibitors (EZH2i) are approved to treat certain cancer types. Previous studies have suggested the potential to combine EZH2i with immune checkpoint blockade targeting coinhibitory receptors like PD-(L)1 and CTLA-4, but whether it can also enhance the activity of agents targeting costimulatory receptors is not known. Here, we explore the combination between EZH2i and an agonist antibody targeting the T cell costimulatory receptor 4-1BB (α4-1BB). Our data show that EZH2i compromise the efficacy of α4-1BB in both CT26 colon carcinoma and in an in vivo protein immunization model. We link this to reduced effector survival and increased BIM expression in CD8+ T cells upon EZH2i treatment. These data support the requirement of EZH2 function in 4-1BB-mediated CD8+ T cell expansion and effector programming and emphasize the consideration that must be given when combining such antitumoral therapies.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neoplasms, Experimental/prevention & control , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Animals , Antibodies, Monoclonal/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Enhancer of Zeste Homolog 2 Protein/immunology , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Burden/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
14.
PLoS One ; 16(12): e0259301, 2021.
Article in English | MEDLINE | ID: mdl-34855754

ABSTRACT

Systemic interleukin-12 (IL12) anti-tumor therapy is highly potent but has had limited utility in the clinic due to severe toxicity. Here, we present two IL12-expressing vector platforms, both of which can overcome the deficiencies of previous systemic IL12 therapies: 1) an integrating lentiviral vector, and 2) a self-replicating messenger RNA formulated with polyethyleneimine. Intratumoral administration of either IL12 vector platform resulted in recruitment of immune cells, including effector T cells and dendritic cells, and the complete remission of established tumors in multiple murine models. Furthermore, concurrent intratumoral administration of the synthetic TLR4 agonist glucopyranosyl lipid A formulated in a stable emulsion (GLA-SE) induced systemic memory T cell responses that mediated complete protection against tumor rechallenge in all survivor mice (8/8 rechallenged mice), whereas only 2/6 total rechallenged mice treated with intratrumoral IL12 monotherapy rejected the rechallenge. Taken together, expression of vectorized IL12 in combination with a TLR4 agonist represents a varied approach to broaden the applicability of intratumoral immune therapies of solid tumors.


Subject(s)
Glucosides/pharmacology , Immunologic Memory/drug effects , Interleukin-12/genetics , Lipid A/pharmacology , Neoplasms, Experimental/immunology , Toll-Like Receptor 4/agonists , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Gene Expression Regulation , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacology , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunologic Memory/genetics , Immunotherapy/methods , Interferon-gamma/blood , Interleukin-12/blood , Interleukin-12/immunology , Lentivirus/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology
15.
PLoS Genet ; 17(11): e1009893, 2021 11.
Article in English | MEDLINE | ID: mdl-34780467

ABSTRACT

Identifying a common oncogenesis pathway among tumors with different oncogenic mutations is critical for developing anti-cancer strategies. Here, we performed transcriptome analyses on two different models of Drosophila malignant tumors caused by Ras activation with cell polarity defects (RasV12/scrib-/-) or by microRNA bantam overexpression with endocytic defects (bantam/rab5-/-), followed by an RNAi screen for genes commonly essential for tumor growth and malignancy. We identified that Juvenile hormone Inducible-21 (JhI-21), a Drosophila homolog of the L-amino acid transporter 1 (LAT1), is upregulated in these malignant tumors with different oncogenic mutations and knocking down of JhI-21 strongly blocked their growth and invasion. JhI-21 expression was induced by simultaneous activation of c-Jun N-terminal kinase (JNK) and Yorkie (Yki) in these tumors and thereby contributed to tumor growth and progression by activating the mTOR-S6 pathway. Pharmacological inhibition of LAT1 activity in Drosophila larvae significantly suppressed growth of RasV12/scrib-/- tumors. Intriguingly, LAT1 inhibitory drugs did not suppress growth of bantam/rab5-/- tumors and overexpression of bantam rendered RasV12/scrib-/- tumors unresponsive to LAT1 inhibitors. Further analyses with RNA sequencing of bantam-expressing clones followed by an RNAi screen suggested that bantam induces drug resistance against LAT1 inhibitors via downregulation of the TMEM135-like gene CG31157. Our observations unveil an evolutionarily conserved role of LAT1 induction in driving Drosophila tumor malignancy and provide a powerful genetic model for studying cancer progression and drug resistance.


Subject(s)
Amino Acid Transport Systems/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Drosophila Proteins/genetics , Drug Resistance, Neoplasm , MAP Kinase Kinase 4/metabolism , YAP-Signaling Proteins/metabolism , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/genetics , Animals , Drosophila , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , MAP Kinase Kinase 4/genetics , MicroRNAs/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , RNA Interference , Signal Transduction , Up-Regulation , YAP-Signaling Proteins/genetics
16.
Mol Biol Cell ; 32(22): ae4, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34747628

ABSTRACT

Today's cell biology could be considered a fusion of disciplines that blends advanced genetics, molecular biology, biochemistry, and engineering to answer fundamental as well as medically relevant scientific questions. Accordingly, our understanding of diseases is greatly aided by an existing vast knowledge base of fundamental cell biology. Gunter Blobel captured this concept when he said, "the tremendous acquisition of basic knowledge will allow a much more rational treatment of cancer, viral infection, degenerative disease and mental disease." In other words, without cell biology can we truly understand, prevent, or effectively treat a disease?


Subject(s)
Cell Biology , Lysosomes/pathology , Neoplasms/pathology , Animals , Autophagy/genetics , Humans , Mice , Neoplasms/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology
17.
Int J Mol Sci ; 22(19)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34638774

ABSTRACT

Targeted immunotherapies have greatly changed treatment of patients with B cell malignancies. To further enhance immunotherapies, research increasingly focuses on the tumor microenvironment (TME), which differs considerably by organ site. However, immunocompetent mouse models of disease to study immunotherapies targeting human molecules within organ-specific TME are surprisingly rare. We developed a myc-driven, primary murine lymphoma model expressing a human-mouse chimeric CD22 (h/mCD22). Stable engraftment of three distinct h/mCD22+ lymphoma was established after subcutaneous and systemic injection. However, only systemic lymphoma showed immune infiltration that reflected human disease. In this model, myeloid cells supported lymphoma growth and showed a phenotype of myeloid-derived suppressor cells. The human CD22-targeted immunotoxin Moxetumomab was highly active against h/mCD22+ lymphoma and similarly reduced infiltration of bone marrow and spleen of all three models up to 90-fold while efficacy against lymphoma in lymph nodes varied substantially, highlighting relevance of organ-specific TME. As in human aggressive lymphoma, anti-PD-L1 as monotherapy was not efficient. However, anti-PD-L1 enhanced efficacy of Moxetumomab suggesting potential for future clinical application. The novel model system of h/mCD22+ lymphoma provides a unique platform to test targeted immunotherapies and may be amenable for other human B cell targets such as CD19 and CD20.


Subject(s)
Immunotherapy , Lymphoma , Neoplasm Proteins , Neoplasms, Experimental , Sialic Acid Binding Ig-like Lectin 2 , Tumor Microenvironment , Animals , Humans , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Organ Specificity/genetics , Organ Specificity/immunology , Sialic Acid Binding Ig-like Lectin 2/genetics , Sialic Acid Binding Ig-like Lectin 2/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
18.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34638879

ABSTRACT

Colorectal cancer (CRC) is one of the most frequently diagnosed cancers in humans. At early stages CRC is treated by surgery and at advanced stages combined with chemotherapy. We examined here the potential effect of glucosylceramide synthase (GCS)-inhibition on CRC biology. GCS is the rate-limiting enzyme in the glycosphingolipid (GSL)-biosynthesis pathway and overexpressed in many human tumors. We suppressed GSL-biosynthesis using the GCS inhibitor Genz-123346 (Genz), NB-DNJ (Miglustat) or by genetic targeting of the GCS-encoding gene UDP-glucose-ceramide-glucosyltransferase- (UGCG). GCS-inhibition or GSL-depletion led to a marked arrest of the cell cycle in Lovo cells. UGCG silencing strongly also inhibited tumor spheroid growth in Lovo cells and moderately in HCT116 cells. MS/MS analysis demonstrated markedly elevated levels of sphingomyelin (SM) and phosphatidylcholine (PC) that occurred in a Genz-concentration dependent manner. Ultrastructural analysis of Genz-treated cells indicated multi-lamellar lipid storage in vesicular compartments. In mice, Genz lowered the incidence of experimentally induced colorectal tumors and in particular the growth of colorectal adenomas. These results highlight the potential for GCS-based inhibition in the treatment of CRC.


Subject(s)
Cell Cycle/drug effects , Colonic Neoplasms , Dioxanes/pharmacology , Glycosphingolipids , Pyrrolidines/pharmacology , Spheroids, Cellular , Animals , Colonic Neoplasms/chemically induced , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Glycosphingolipids/biosynthesis , Glycosphingolipids/genetics , HCT116 Cells , Humans , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
19.
Sci Rep ; 11(1): 21087, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702924

ABSTRACT

Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer in the clinic. Further discovery of novel drugs or therapeutic protocols that enhance efficacy requires reliable animal models that recapitulate human immune responses to ICI treatment in vivo. In this study, we utilized an immunodeficient NOG mouse substrain deficient for mouse FcγR genes, NOG-FcγR-/- mice, to evaluate the anti-cancer effects of nivolumab, an anti-programmed cell death-1 (PD-1) antibody. After reconstitution of human immune systems by human hematopoietic stem cell transplantation (huNOG-FcγR-/- mice), four different programmed death-ligand 1 (PD-L1)-positive human cancer cell lines were tested. Among them, the growth of three cell lines was strongly suppressed by nivolumab in huNOG-FcγR-/- mice, but not in conventional huNOG mice. Accordingly, immunohistochemistry demonstrated the enhanced infiltration of human T cells into tumor parenchyma in only nivolumab-treated huNOG-FcγR-/- mice. Consistently, the number of human T cells was increased in the spleen in huNOG-FcγR-/- mice by nivolumab but not in huNOG mice. Furthermore, human PD-L1 expression was strongly induced in the spleen of huNOG-FcγR-/- mice. Collectively, our results suggest that the anti-cancer effects of anti-PD-1 antibodies can be detected more clearly in NOG-FcγR-/- mice than in NOG mice.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Proteins , Neoplasms, Experimental , Nivolumab/pharmacology , Programmed Cell Death 1 Receptor , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Hematopoietic Stem Cell Transplantation , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology
20.
J Clin Invest ; 131(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34491908

ABSTRACT

Initiation of T cell receptor (TCR) signaling involves the activation of the tyrosine kinase LCK; however, it is currently unclear how LCK is recruited and activated. Here, we have identified the membrane protein CD146 as an essential member of the TCR network for LCK activation. CD146 deficiency in T cells substantially impaired thymocyte development and peripheral activation, both of which depend on TCR signaling. CD146 was found to directly interact with the SH3 domain of coreceptor-free LCK via its cytoplasmic domain. Interestingly, we found CD146 to be present in both monomeric and dimeric forms in T cells, with the dimerized form increasing after TCR ligation. Increased dimerized CD146 recruited LCK and promoted LCK autophosphorylation. In tumor models, CD146 deficiency dramatically impaired the antitumor response of T cells. Together, our data reveal an LCK activation mechanism for TCR initiation. We also underscore a rational intervention based on CD146 for tumor immunotherapy.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Neoplasms, Experimental/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , CD146 Antigen/genetics , CD146 Antigen/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...