Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
J Immunother Cancer ; 10(4)2022 04.
Article in English | MEDLINE | ID: mdl-35483745

ABSTRACT

BACKGROUND: Anti-GD2 monoclonal antibody immunotherapy has significantly improved the overall survival rate for high-risk neuroblastoma patients. However, 40% of patients fail to respond or develop resistance to treatment, and the molecular mechanisms by which this occurs remain poorly understood. Tumor-derived small extracellular vesicles (sEVs) have emerged as critical regulators in modulating the response to immunotherapy. In this study, we investigated the role of neuroblastoma-derived sEVs in promoting resistance to the anti-GD2 monoclonal antibody dinutuximab. Moreover, to determine whether pharmacologic inhibition of sEV secretion sensitizes tumors to dinutuximab treatment, we combined dinutuximab with tipifarnib, a farnesyltransferase inhibitor that inhibits sEV secretion. METHODS: We investigated the role of neuroblastoma-derived sEVs in modulating the response to dinutuximab by utilizing the syngeneic 9464D-GD2 mouse model. The effect of neuroblastoma-derived sEVs in modulating the tumor microenvironment (TME) and host immune system were evaluated by RNA-sequencing and flow cytometry. Importantly, we used this mouse model to investigate the efficacy of tipifarnib in sensitizing neuroblastoma tumors to dinutuximab. The effect of tipifarnib on both the TME and host immune system were assessed by flow cytometry. RESULTS: We demonstrated that neuroblastoma-derived sEVs significantly attenuated the efficacy of dinutuximab in vivo and modulated tumor immune cell infiltration upon dinutuximab treatment to create an immunosuppressive TME that contains more tumor-associated macrophages and fewer tumor-infiltrating NK cells. In addition, we demonstrated that neuroblastoma-derived sEVs suppress splenic NK cell maturation in vivo and dinutuximab-induced NK cell-mediated antibody-dependent cellular cytotoxicity in vitro. Importantly, tipifarnib drastically enhanced the efficacy of dinutuximab-mediated inhibition of tumor growth and prevented the immunosuppressive effects of neuroblastoma-derived sEVs in vivo. CONCLUSIONS: These preclinical findings uncover a novel mechanism by which neuroblastoma-derived sEVs modulate the immune system to promote resistance to dinutuximab and suggest that tipifarnib-mediated inhibition of sEV secretion may serve as a viable treatment strategy to enhance the antitumor efficacy of anti-GD2 immunotherapy in high-risk neuroblastoma patients.


Subject(s)
Antineoplastic Agents , Extracellular Vesicles , Neuroblastoma , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Immunologic Factors/therapeutic use , Immunotherapy , Mice , Neuroblastoma/pathology , Quinolones , Tumor Microenvironment
3.
Children (Basel) ; 9(2)2022 Feb 06.
Article in English | MEDLINE | ID: mdl-35204936

ABSTRACT

BACKGROUND: Enlarged lymph nodes are a common complaint in a Pediatrician's office. Diagnosis of reactive lymphadenopathy secondary to infectious, inflammatory, immune dysregulation calls for clinical investigation, including a thorough history, physical exam, imaging, and less often, a biopsy of the lymph node. Here we discuss a rare presentation of extensive generalized, chronic, waxing, and waning lymphadenopathy diagnosed as Progressive Transformation of Germinal Centers (PTGC) and the course of illness over eight years follow up period. DISCUSSION: Progressive Transformation of Germinal Centers (PTGC) is considered a benign condition, but extensive recurrent generalized lymphadenopathy in a very young child has not been reported before. This case demonstrates the importance of long-term follow-up and tailoring the diagnostic work-up and management based on new signs and symptoms. Here we focus on the clinical considerations and management of complex presentation of a common clinical finding.

4.
In Vivo ; 36(1): 49-56, 2022.
Article in English | MEDLINE | ID: mdl-34972699

ABSTRACT

BACKGROUND/AIM: Neuroblastoma is clinically and molecularly heterogeneous, with poor outcomes despite multimodal treatment strategies. The primary tumor site is an independent predictor of survival; adrenal tumors have the worst outcomes, while posterior mediastinum tumors carry a more favorable prognosis. MATERIALS AND METHODS: To elucidate the role of the primary tumor microenvironment in mediating survival outcomes, we developed a mouse model for the study of extra-adrenal neuroblastoma by injecting luciferase-tagged cells into either the subpleural space of the posterior chest or the adrenal gland. RESULTS: Solid tumors developed in the thoracic cavity at the same rate and efficiency as the adrenal as early as one week post-surgery. The survival rate following surgery was equivalent, though the physiological tolerance for large tumors was lower in the thoracic group. CONCLUSION: This novel mouse model of survivable extra-adrenal neuroblastoma will enable future investigations of the distinct tumor microenvironments between the adrenal gland and posterior mediastinum.


Subject(s)
Adrenal Gland Neoplasms , Mediastinal Neoplasms , Neuroblastoma , Adrenal Gland Neoplasms/surgery , Animals , Mediastinal Neoplasms/surgery , Mice , Models, Anatomic , Neuroblastoma/surgery , Prognosis , Tumor Microenvironment
5.
Cancer Res ; 81(2): 452-463, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33115808

ABSTRACT

Although neoadjuvant chemotherapy is a standard component of breast cancer treatment, recent evidence suggests that chemotherapeutic drugs can promote metastasis through poorly defined mechanisms. Here we utilize xenograft mouse models of triple-negative breast cancer to explore the importance of chemotherapy-induced tumor-derived small extracellular vesicles (sEV) in metastasis. Doxorubicin (DXR) enhanced tumor cell sEV secretion to accelerate pulmonary metastasis by priming the premetastatic niche. Proteomic analysis and CRISPR/Cas9 gene editing identified the inflammatory glycoprotein PTX3 enriched in DXR-elicited sEV as a critical regulator of chemotherapy-induced metastasis. Both genetic inhibition of sEV secretion from primary tumors and pharmacologic inhibition of sEV uptake in secondary organs suppressed metastasis following chemotherapy. Taken together, this research uncovers a mechanism of chemotherapy-mediated metastasis by which drug-induced upregulation of sEV secretion and PTX3 protein cargo primes the premetastatic niche and suggests that inhibition of either sEV uptake in secondary organs or secretion from primary tumor cells may be promising therapeutic strategies to suppress metastasis. SIGNIFICANCE: These findings show that chemotherapy-induced small extracellular vesicles accelerate breast cancer metastasis, and targeted inhibition of tumor-derived vesicles may be a promising therapeutic strategy to improve the efficacy of chemotherapy treatment.


Subject(s)
Breast Neoplasms/drug therapy , C-Reactive Protein/metabolism , Doxorubicin/adverse effects , Extracellular Vesicles/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Serum Amyloid P-Component/metabolism , Animals , Antibiotics, Antineoplastic/adverse effects , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , C-Reactive Protein/genetics , Cell Movement , Cell Proliferation , Extracellular Vesicles/drug effects , Female , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Serum Amyloid P-Component/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cell Death Differ ; 28(2): 657-670, 2021 02.
Article in English | MEDLINE | ID: mdl-32807832

ABSTRACT

Autophagosomal membranes can serve as activation platforms for intracellular death-inducing signaling complexes (iDISCs) to initiate Caspase-8-dependent apoptosis. In this study, we explore the impact of ESCRT-III-dependent phagophore closure on iDISC assemblies and cell death in osteosarcoma and neuroblastoma cells. Inhibition of phagophore closure by conditional depletion of CHMP2A, an ESCRT-III component, stabilizes iDISCs on immature autophagosomal membranes and induces Caspase-8-dependent cell death. Importantly, suppression of the iDISC formation via deletion of ATG7, an E1 enzyme for ubiquitin-like autophagy-related proteins, blocks Caspase-8 activation and cell death following CHMP2A depletion. Although DR5 expression and TRAIL-induced apoptosis are enhanced in CHMP2A-depleted cells, the canonical extrinsic pathway of apoptosis is not responsible for the initiation of cell death by CHMP2A depletion. Furthermore, the loss of CHMP2A impairs neuroblastoma tumor growth associated with decreased autophagy and increased apoptosis in vivo. Together, these findings indicate that inhibition of the ESCRT-III-dependent autophagosome sealing process triggers noncanonical Caspase-8 activation and apoptosis, which may open new avenues for therapeutic targeting of autophagy in cancer.


Subject(s)
Autophagy , Caspase 8/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Osteosarcoma/metabolism , Signal Transduction , Animals , Apoptosis , Autophagosomes/metabolism , Cell Line, Tumor , Endosomal Sorting Complexes Required for Transport/genetics , Female , Humans , Male , Mice , Neuroblastoma/metabolism , Xenograft Model Antitumor Assays
7.
Front Cell Dev Biol ; 8: 469, 2020.
Article in English | MEDLINE | ID: mdl-32637411

ABSTRACT

PURPOSE: To investigate the role of protein misfolding in retinal pigment epithelial (RPE) cell dysfunction, the effects of R345W-Fibulin-3 expression on RPE cell phenotype were studied. METHODS: Primary RPE cells were cultured to confluence on Transwells and infected with lentivirus constructs to express wild-type (WT)- or R345W-Fibulin-3. Barrier function was assessed by evaluating zonula occludens-1 (ZO-1) distribution and trans-epithelial electrical resistance (TER). Polarized secretion of vascular endothelial growth factor (VEGF), was measured by Enzyme-linked immunosorbent assay (ELISA). Differentiation status was assessed by qPCR of genes known to be preferentially expressed in terminally differentiated RPE cells, and conversion to an epithelial-mesenchymal transition (EMT) phenotype was assessed by a migration assay. RESULTS: Compared to RPE cells expressing WT-Fibulin-3, ZO-1 distribution was disrupted and TER values were significantly lower in RPE cells expressing R345W-Fibulin-3. In cells expressing mutant Fibulin-3, VEGF secretion was attenuated basally but not in the apical direction, whereas Fibulin-3 secretion was reduced in both the apical and basal directions. Retinal pigment epithelial signature genes were downregulated and multiple genes associated with EMT were upregulated in the mutant group. Migration assays revealed a faster recovery rate in ARPE-19 cells overexpressing R345W-Fibulin-3 compared to WT. CONCLUSIONS: The results suggest that expression of R345W-Fibulin-3 promotes EMT in RPE cells.

8.
Mol Cancer Ther ; 17(11): 2365-2376, 2018 11.
Article in English | MEDLINE | ID: mdl-30166400

ABSTRACT

Neuroblastoma is the most common extracranial solid malignancy in the pediatric population, accounting for over 9% of all cancer-related deaths in children. Autophagy is a cell self-protective mechanism that promotes tumor cell growth and survival, making it an attractive target for treating cancer. However, the role of autophagy in neuroblastoma tumor growth and metastasis is largely undefined. Here we demonstrate that targeted inhibition of an essential autophagy kinase, unc-51 like autophagy kinase 1 (ULK1), with a recently developed small-molecule inhibitor of ULK1, SBI-0206965, significantly reduces cell growth and promotes apoptosis in SK-N-AS, SH-SY5Y, and SK-N-DZ neuroblastoma cell lines. Furthermore, inhibition of ULK1 by a dominant-negative mutant of ULK1 (dnULK1K46N) significantly reduces growth and metastatic disease and prolongs survival of mice bearing SK-N-AS xenograft tumors. We also show that SBI-0206965 sensitizes SK-N-AS cells to TRAIL treatment, but not to mTOR inhibitors (INK128, Torin1) or topoisomerase inhibitors (doxorubicin, topotecan). Collectively, these findings demonstrate that ULK1 is a viable drug target and suggest that inhibitors of ULK1 may provide a novel therapeutic option for the treatment of neuroblastoma. Mol Cancer Ther; 17(11); 2365-76. ©2018 AACR.


Subject(s)
Apoptosis , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Neuroblastoma/enzymology , Neuroblastoma/pathology , Animals , Apoptosis/drug effects , Autophagy-Related Protein-1 Homolog/metabolism , Benzamides/chemistry , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Pyrimidines/chemistry , Pyrimidines/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Topoisomerase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
9.
Nat Commun ; 9(1): 2855, 2018 07 20.
Article in English | MEDLINE | ID: mdl-30030437

ABSTRACT

The mechanism of phagophore closure remains unclear due to technical limitations in distinguishing unclosed and closed autophagosomal membranes. Here, we report the HaloTag-LC3 autophagosome completion assay that specifically detects phagophores, nascent autophagosomes, and mature autophagic structures. Using this assay, we identify the endosomal sorting complexes required for transport (ESCRT)-III component CHMP2A as a critical regulator of phagophore closure. During autophagy, CHMP2A translocates to the phagophore and regulates the separation of the inner and outer autophagosomal membranes to form double-membrane autophagosomes. Consistently, inhibition of the AAA-ATPase VPS4 activity impairs autophagosome completion. The ESCRT-mediated membrane abscission appears to be a critical step in forming functional autolysosomes by preventing mislocalization of lysosome-associated membrane glycoprotein 1 to the inner autophagosomal membrane. Collectively, our work reveals a function for the ESCRT machinery in the final step of autophagosome formation and provides a useful tool for quantitative analysis of autophagosome biogenesis and maturation.


Subject(s)
Autophagy , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Gene Expression Regulation , Lysosomes/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Carrier Proteins , Cell Membrane/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , RNA, Small Interfering/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
10.
Autophagy ; 14(7): 1110-1128, 2018.
Article in English | MEDLINE | ID: mdl-29863947

ABSTRACT

Macroautophagy/autophagy is a fundamental cellular degradation mechanism that maintains cell homeostasis, regulates cell signaling, and promotes cell survival. Its role in promoting tumor cell survival in stress conditions is well characterized, and makes autophagy an attractive target for cancer therapy. Emerging research indicates that autophagy also influences cancer metastasis, which is the primary cause of cancer-associated mortality. However, data demonstrate that the regulatory role of autophagy in metastasis is multifaceted, and includes both metastasis-suppressing and -promoting functions. The metastasis-suppressing functions of autophagy, in particular, have important implications for autophagy-based treatments, as inhibition of autophagy may increase the risk of metastasis. In this review, we discuss the mechanisms and context underlying the role of autophagy in metastasis, which include autophagy-mediated regulation of focal adhesion dynamics, integrin signaling and trafficking, Rho GTPase-mediated cytoskeleton remodeling, anoikis resistance, extracellular matrix remodeling, epithelial-to-mesenchymal transition signaling, and tumor-stromal cell interactions. Through this, we aim to clarify the context-dependent nature of autophagy-mediated metastasis and provide direction for further research investigating the role of autophagy in cancer metastasis.


Subject(s)
Autophagy , Neoplasm Metastasis/pathology , Animals , Epithelial-Mesenchymal Transition , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Humans , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...