Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 183(5): 1202-1218.e25, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33142117

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) tumors have a nutrient-poor, desmoplastic, and highly innervated tumor microenvironment. Although neurons can release stimulatory factors to accelerate PDAC tumorigenesis, the metabolic contribution of peripheral axons has not been explored. We found that peripheral axons release serine (Ser) to support the growth of exogenous Ser (exSer)-dependent PDAC cells during Ser/Gly (glycine) deprivation. Ser deprivation resulted in ribosomal stalling on two of the six Ser codons, TCC and TCT, and allowed the selective translation and secretion of nerve growth factor (NGF) by PDAC cells to promote tumor innervation. Consistent with this, exSer-dependent PDAC tumors grew slower and displayed enhanced innervation in mice on a Ser/Gly-free diet. Blockade of compensatory neuronal innervation using LOXO-101, a Trk-NGF inhibitor, further decreased PDAC tumor growth. Our data indicate that axonal-cancer metabolic crosstalk is a critical adaptation to support PDAC growth in nutrient poor environments.


Subject(s)
Neurons/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Protein Biosynthesis , Serine/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aged , Animals , Axons/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Codon/genetics , Female , Glycine/metabolism , Humans , Male , Mice , Middle Aged , Mitochondria/metabolism , Nerve Tissue/pathology , Oxygen Consumption , Pancreatic Neoplasms/pathology , Pyrazoles , Pyrimidines , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer/genetics , Rats
2.
Blood ; 143(23): 2433-2437, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38518102

ABSTRACT

ABSTRACT: Iron-mediated induction of bone morphogenetic protein (BMP)6 expression by liver endothelial cells is essential for iron homeostasis regulation. We used multiple dietary and genetic mouse cohorts to demonstrate a minor functional role for the metal-ion transporter ZIP8 in regulating BMP6 expression under high-iron conditions.


Subject(s)
Bone Morphogenetic Protein 6 , Cation Transport Proteins , Iron , Animals , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein 6/genetics , Mice , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Iron/metabolism , Endothelial Cells/metabolism , Mice, Knockout , Gene Expression Regulation , Liver/metabolism , Mice, Inbred C57BL , Homeostasis
3.
Nature ; 581(7806): 100-105, 2020 05.
Article in English | MEDLINE | ID: mdl-32376951

ABSTRACT

Immune evasion is a major obstacle for cancer treatment. Common mechanisms of evasion include impaired antigen presentation caused by mutations or loss of heterozygosity of the major histocompatibility complex class I (MHC-I), which has been implicated in resistance to immune checkpoint blockade (ICB) therapy1-3. However, in pancreatic ductal adenocarcinoma (PDAC), which is resistant to most therapies including ICB4, mutations that cause loss of MHC-I are rarely found5 despite the frequent downregulation of MHC-I expression6-8. Here we show that, in PDAC, MHC-I molecules are selectively targeted for lysosomal degradation by an autophagy-dependent mechanism that involves the autophagy cargo receptor NBR1. PDAC cells display reduced expression of MHC-I at the cell surface and instead demonstrate predominant localization within autophagosomes and lysosomes. Notably, inhibition of autophagy restores surface levels of MHC-I and leads to improved antigen presentation, enhanced anti-tumour T cell responses and reduced tumour growth in syngeneic host mice. Accordingly, the anti-tumour effects of autophagy inhibition are reversed by depleting CD8+ T cells or reducing surface expression of MHC-I. Inhibition of autophagy, either genetically or pharmacologically with chloroquine, synergizes with dual ICB therapy (anti-PD1 and anti-CTLA4 antibodies), and leads to an enhanced anti-tumour immune response. Our findings demonstrate a role for enhanced autophagy or lysosome function in immune evasion by selective targeting of MHC-I molecules for degradation, and provide a rationale for the combination of autophagy inhibition and dual ICB therapy as a therapeutic strategy against PDAC.


Subject(s)
Adenocarcinoma/immunology , Autophagy/immunology , Carcinoma, Pancreatic Ductal/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Pancreatic Neoplasms/immunology , Tumor Escape/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Autophagy/drug effects , Autophagy/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/immunology , Cell Line, Tumor , Chloroquine/pharmacology , Female , Histocompatibility Antigens Class I/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Escape/drug effects
4.
J Biol Chem ; 299(5): 104691, 2023 05.
Article in English | MEDLINE | ID: mdl-37037306

ABSTRACT

Mitophagy is a cargo-specific autophagic process that recycles damaged mitochondria to promote mitochondrial turnover. PTEN-induced putative kinase 1 (PINK1) mediates the canonical mitophagic pathway. However, the role of PINK1 in diseases where mitophagy has been purported to play a role, such as colorectal cancer, is unclear. Our results here demonstrate that higher PINK1 expression is positively correlated with decreased colon cancer survival, and mitophagy is required for colon cancer growth. We show that doxycycline-inducible knockdown (KD) of PINK1 in a panel of colon cancer cell lines inhibited proliferation, whereas disruption of other mitophagy receptors did not impact cell growth. We observed that PINK KD led to a decrease in mitochondrial respiration, membrane hyperpolarization, accumulation of mitochondrial DNA, and depletion of antioxidant glutathione. In addition, mitochondria are important hubs for the utilization of iron and synthesizing iron-dependent cofactors such as heme and iron sulfur clusters. We observed an increase in the iron storage protein ferritin and a decreased labile iron pool in the PINK1 KD cells, but total cellular iron or markers of iron starvation/overload were not affected. Finally, cellular iron storage and the labile iron pool are maintained via autophagic degradation of ferritin (ferritinophagy). We found overexpressing nuclear receptor coactivator 4, a key adaptor for ferritinophagy, rescued cell growth and the labile iron pool in PINK1 KD cells. These results indicate that PINK1 integrates mitophagy and ferritinophagy to regulate intracellular iron availability and is essential for maintaining intracellular iron homeostasis to support survival and growth in colorectal cancer cells.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Mitophagy , Protein Kinases , Humans , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Ferritins , Iron/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
Blood ; 139(16): 2547-2552, 2022 04 21.
Article in English | MEDLINE | ID: mdl-34990508

ABSTRACT

Intestinal iron absorption is activated during increased systemic demand for iron. The best-studied example is iron deficiency anemia, which increases intestinal iron absorption. Interestingly, the intestinal response to anemia is very similar to that of iron overload disorders, as both the conditions activate a transcriptional program that leads to a hyperabsorption of iron via the transcription factor hypoxia-inducible factor 2α (HIF2α). However, pathways for selective targeting of intestine-mediated iron overload remain unknown. Nuclear receptor coactivator 4 (NCOA4) is a critical cargo receptor for autophagic breakdown of ferritin and the subsequent release of iron, in a process termed ferritinophagy. Our work demonstrates that NCOA4-mediated intestinal ferritinophagy is integrated into systemic iron demand via HIF2α. To demonstrate the importance of the intestinal HIF2α/ferritinophagy axis in systemic iron homeostasis, whole-body and intestine-specific NCOA4-/- mouse lines were generated and assessed. The analyses revealed that the intestinal and systemic response to iron deficiency was not altered after disruption of intestinal NCOA4. However, in a mouse model of hemochromatosis, ablation of intestinal NCOA4 was protective against iron overload. Therefore, NCOA4 can be selectively targeted for the management of iron overload disorders without disrupting the physiological processes involved in the response to systemic iron deficiency.


Subject(s)
Anemia , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hemochromatosis , Iron Overload , Animals , Enterocytes/metabolism , Hemochromatosis/genetics , Iron/metabolism , Mice , Nuclear Receptor Coactivators/genetics , Transcription Factors/metabolism
6.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33531365

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest forms of cancer and is highly refractory to current therapies. We had previously shown that PDAC can utilize its high levels of basal autophagy to support its metabolism and maintain tumor growth. Consistent with the importance of autophagy in PDAC, autophagy inhibition significantly enhances response of PDAC patients to chemotherapy in two randomized clinical trials. However, the specific metabolite(s) that autophagy provides to support PDAC growth is not yet known. In this study, we demonstrate that under nutrient-replete conditions, loss of autophagy in PDAC leads to a relatively restricted impairment of amino acid pools, with cysteine levels showing a significant drop. Additionally, we made the striking discovery that autophagy is critical for the proper membrane localization of the cystine transporter SLC7A11. Mechanistically, autophagy impairment results in the loss of SLC7A11 on the plasma membrane and increases its localization at the lysosome in an mTORC2-dependent manner. Our results demonstrate a critical link between autophagy and cysteine metabolism and provide mechanistic insights into how targeting autophagy can cause metabolic dysregulation in PDAC.


Subject(s)
Adenocarcinoma/genetics , Amino Acid Transport System y+/genetics , Carcinoma, Pancreatic Ductal/genetics , Cell Proliferation/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Autophagy/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Homeostasis/genetics , Humans , Mice , Reactive Oxygen Species/metabolism
7.
J Appl Clin Med Phys ; 24(7): e13965, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36924220

ABSTRACT

PURPOSE: The role of biliary stents in image-guided localization for pancreatic cancer has been inconclusive. To date, stent accuracy has been largely evaluated against implanted fiducials on cone beam computed tomography. We aim to use magnetic resonance (MR) soft tissue as a direct reference to examine the geometric and dosimetric impacts of stent-based localization on the newly available MR linear accelerator. METHODS: Thirty pancreatic cancer patients (132 fractions) treated on our MR linear accelerator were identified to have a biliary stent. In our standard adaptive workflow, patients were set up to the target using soft tissue for image registration and structures were re-contoured on daily MR images. The original plan was then projected on treatment anatomy and dose predicted, followed by plan re-optimization and treatment delivery. These online predicted plans were soft tissue-based and served as reference plans. Retrospective image registration to the stent was performed offline to simulate stent-based localization and the magnitude of shifts was taken as the geometric accuracy of stent localization. New predicted plans were generated based on stent-alignment for dosimetric comparison. RESULTS: Shifts were within 3 mm for 90% of the cases (mean = 1.5 mm); however, larger shifts up to 7.2 mm were observed. Average PTV coverage dropped by 1.1% with a maximum drop of 26.8%. The mean increase in V35Gy was 0.15, 0.05, 0.02, and 0.02 cc for duodenum, stomach, small bowel and large bowel, respectively. Stent alignment was significantly worse for all metrics except for small bowel (p = 0.07). CONCLUSIONS: Overall discrepancy between stent- and soft tissue-alignment was modest; however, large discrepancies were observed for select cases. While PTV coverage loss may be compensated for by using a larger margin, the increase in dose to gastrointestinal organs at risk may limit the role of biliary stents in image-guided localization.


Subject(s)
Pancreatic Neoplasms , Radiosurgery , Radiotherapy, Image-Guided , Humans , Radiosurgery/methods , Retrospective Studies , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/radiotherapy , Pancreatic Neoplasms/surgery , Stents , Magnetic Resonance Spectroscopy , Radiotherapy Planning, Computer-Assisted/methods , Radiotherapy Dosage , Radiotherapy, Image-Guided/methods , Pancreatic Neoplasms
8.
Nat Chem Biol ; 16(6): 635-643, 2020 06.
Article in English | MEDLINE | ID: mdl-32251410

ABSTRACT

Doublecortin like kinase 1 (DCLK1) is an understudied kinase that is upregulated in a wide range of cancers, including pancreatic ductal adenocarcinoma (PDAC). However, little is known about its potential as a therapeutic target. We used chemoproteomic profiling and structure-based design to develop a selective, in vivo-compatible chemical probe of the DCLK1 kinase domain, DCLK1-IN-1. We demonstrate activity of DCLK1-IN-1 against clinically relevant patient-derived PDAC organoid models and use a combination of RNA-sequencing, proteomics and phosphoproteomics analysis to reveal that DCLK1 inhibition modulates proteins and pathways associated with cell motility in this context. DCLK1-IN-1 will serve as a versatile tool to investigate DCLK1 biology and establish its role in cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Movement , Doublecortin Protein , Doublecortin-Like Kinases , Drug Screening Assays, Antitumor , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Molecular Docking Simulation , Molecular Structure , Protein Kinase Inhibitors/pharmacokinetics , Proteomics , Rats , Structure-Activity Relationship , Zebrafish , Pancreatic Neoplasms
9.
Cell Commun Signal ; 19(1): 116, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34801048

ABSTRACT

BACKGROUND: Wolfram syndrome (WFS) is a rare autosomal recessive syndrome in which diabetes mellitus and neurodegenerative disorders occur as a result of Wolframin deficiency and increased ER stress. In addition, WFS1 deficiency leads to calcium homeostasis disturbances and can change mitochondrial dynamics. The aim of this study was to evaluate protein levels and changes in gene transcription on human WFS cell model under experimental ER stress. METHODS: We performed transcriptomic and proteomic analysis on WFS human cell model-skin fibroblasts reprogrammed into induced pluripotent stem (iPS) cells and then into neural stem cells (NSC) with subsequent ER stress induction using tunicamycin (TM). Results were cross-referenced with publicly available RNA sequencing data in hippocampi and hypothalami of mice with WFS1 deficiency. RESULTS: Proteomic analysis identified specific signal pathways that differ in NSC WFS cells from healthy ones. Next, detailed analysis of the proteins involved in the mitochondrial function showed the down-regulation of subunits of the respiratory chain complexes in NSC WFS cells, as well as the up-regulation of proteins involved in Krebs cycle and glycolysis when compared to the control cells. Based on pathway enrichment analysis we concluded that in samples from mice hippocampi the mitochondrial protein import machinery and OXPHOS were significantly down-regulated. CONCLUSIONS: Our results show the functional and morphological secondary mitochondrial damage in patients with WFS. Video Abstract.


Subject(s)
Wolfram Syndrome
10.
Adv Exp Med Biol ; 1301: 41-57, 2021.
Article in English | MEDLINE | ID: mdl-34370287

ABSTRACT

Nuclear receptor coactivator 4 (NCOA4) is a selective cargo receptor that mediates the autophagic degradation of ferritin, the cytosolic iron storage complex, in a process known as ferritinophagy. NCOA4-mediated ferritinophagy is required to maintain intracellular and systemic iron homeostasis and thereby iron-dependent physiologic processes such as erythropoiesis. Given this role of ferritinophagy in regulating iron homeostasis, modulating NCOA4-mediated ferritinophagic flux alters sensitivity to ferroptosis, a non-apoptotic iron-dependent form of cell death triggered by peroxidation of polyunsaturated fatty acids (PUFAs). A role for ferroptosis has been established in the pathophysiology of cancer and neurodegeneration; however, the importance of ferritinophagy in these pathologies remains largely unknown. Here, we review the available evidence on biochemical regulation of NCOA4-mediated ferritinophagy and its role in modulating sensitivity to innate and induced ferroptosis in neurodegenerative diseases and cancer. Finally, we evaluate the potential of modulating ferritinophagy in combination with ferroptosis inducers as a therapeutic strategy.


Subject(s)
Ferroptosis , Nuclear Receptor Coactivators , Autophagy , Ferritins/genetics , Humans , Iron/metabolism , Nuclear Receptor Coactivators/genetics , Nuclear Receptor Coactivators/metabolism
11.
Ann Surg Oncol ; 27(4): 1191-1200, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31802297

ABSTRACT

BACKGROUND: Neoadjuvant therapy has shown value in various cancer types. The role of neoadjuvant therapy in pancreatic ductal adenocarcinoma (PDAC), however, remains unknown. The aim of the present work is to evaluate the effect of neoadjuvant therapy on the survival of patients with borderline-resectable PDAC. PATIENTS AND METHODS: Between 2004 and 2015, 7730 patients with resectable PDAC and 1980 patients with borderline-resectable PDAC were identified from the National Cancer Database (NCDB). Survival was compared between resectable and borderline-resectable patients. Survival and pathologic characteristics were also compared within borderline-resectable patients who received neoadjuvant therapy and those who received adjuvant therapy alone. Kaplan-Meier method and Cox proportional-hazard models were used for analysis. RESULTS: Median overall survival (mOS) of all patients with resectable PDAC was similar to that of patients with borderline-resectable disease treated with neoadjuvant therapy (26.5 versus 25.7 months, p = 0.78). Patients with borderline-resectable disease treated with neoadjuvant therapy had improved mOS compared with borderline-resectable patients treated with adjuvant therapy alone (25.7 versus 19.6 months, p < 0.0001). When comparing patients with borderline-resectable disease who received neoadjuvant therapy versus those who received adjuvant therapy alone, the former less often had node-positive pancreatic cancer (40.6% versus 76.3%, p < 0.001) and margin-positive resections (17.8% versus 44.4%, p < 0.001). CONCLUSION: Neoadjuvant therapy is associated with enhanced survival in patients with borderline-resectable pancreatic cancer, which may be attributed to tumor downstaging.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Neoadjuvant Therapy/mortality , Pancreatectomy/mortality , Pancreatic Neoplasms/therapy , Aged , Carcinoma, Pancreatic Ductal/pathology , Combined Modality Therapy , Databases, Factual , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/pathology , Retrospective Studies , Survival Analysis , Treatment Outcome , United States/epidemiology
12.
Nat Chem Biol ; 14(7): 706-714, 2018 07.
Article in English | MEDLINE | ID: mdl-29892083

ABSTRACT

Heterobifunctional small-molecule degraders that induce protein degradation through ligase-mediated ubiquitination have shown considerable promise as a new pharmacological modality. However, we currently lack a detailed understanding of the molecular basis for target recruitment and selectivity, which is critically required to enable rational design of degraders. Here we utilize a comprehensive characterization of the ligand-dependent CRBN-BRD4 interaction to demonstrate that binding between proteins that have not evolved to interact is plastic. Multiple X-ray crystal structures show that plasticity results in several distinct low-energy binding conformations that are selectively bound by ligands. We demonstrate that computational protein-protein docking can reveal the underlying interprotein contacts and inform the design of a BRD4 selective degrader that can discriminate between highly homologous BET bromodomains. Our findings that plastic interprotein contacts confer selectivity for ligand-induced protein dimerization provide a conceptual framework for the development of heterobifunctional ligands.


Subject(s)
Acetamides/pharmacology , Nuclear Proteins/metabolism , Peptide Hydrolases/metabolism , Thalidomide/pharmacology , Thiophenes/pharmacology , Transcription Factors/metabolism , Acetamides/chemistry , Adaptor Proteins, Signal Transducing , Binding Sites/drug effects , Cell Cycle Proteins , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Ligands , Models, Molecular , Molecular Conformation , Nuclear Proteins/chemistry , Peptide Hydrolases/chemistry , Thalidomide/chemistry , Thiophenes/chemistry , Transcription Factors/chemistry , Ubiquitin-Protein Ligases
13.
Nature ; 509(7498): 105-9, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24695223

ABSTRACT

Autophagy, the process by which proteins and organelles are sequestered in double-membrane structures called autophagosomes and delivered to lysosomes for degradation, is critical in diseases such as cancer and neurodegeneration. Much of our understanding of this process has emerged from analysis of bulk cytoplasmic autophagy, but our understanding of how specific cargo, including organelles, proteins or intracellular pathogens, are targeted for selective autophagy is limited. Here we use quantitative proteomics to identify a cohort of novel and known autophagosome-enriched proteins in human cells, including cargo receptors. Like known cargo receptors, nuclear receptor coactivator 4 (NCOA4) was highly enriched in autophagosomes, and associated with ATG8 proteins that recruit cargo-receptor complexes into autophagosomes. Unbiased identification of NCOA4-associated proteins revealed ferritin heavy and light chains, components of an iron-filled cage structure that protects cells from reactive iron species but is degraded via autophagy to release iron through an unknown mechanism. We found that delivery of ferritin to lysosomes required NCOA4, and an inability of NCOA4-deficient cells to degrade ferritin led to decreased bioavailable intracellular iron. This work identifies NCOA4 as a selective cargo receptor for autophagic turnover of ferritin (ferritinophagy), which is critical for iron homeostasis, and provides a resource for further dissection of autophagosomal cargo-receptor connectivity.


Subject(s)
Autophagy , Ferritins/metabolism , Nuclear Receptor Coactivators/metabolism , Phagosomes/metabolism , Proteomics , Adaptor Proteins, Signal Transducing/metabolism , Autophagy-Related Protein 8 Family , Biological Availability , Ferritins/chemistry , Homeostasis , Humans , Iron/metabolism , Lysosomes/metabolism , Microfilament Proteins/metabolism , Nuclear Receptor Coactivators/deficiency , Nuclear Receptor Coactivators/genetics , Protein Binding , Protein Transport , Substrate Specificity
14.
Anal Chem ; 91(8): 5106-5115, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30779550

ABSTRACT

MHC-I peptides are intracellular-cleaved peptides, usually 8-11 amino acids in length, which are presented on the cell surface and facilitate CD8+ T cell responses. Despite the appreciation of CD8+ T-cell antitumor immune responses toward improvement in patient outcomes, the MHC-I peptide ligands that facilitate the response are poorly described. Along these same lines, although many therapies have been recognized for their ability to reinvigorate antitumor CD8+ T-cell responses, whether these therapies alter the MHC-I peptide repertoire has not been fully assessed due to the lack of quantitative strategies. We develop a multiplexing platform for screening therapy-induced MHC-I ligands by employing tandem mass tags (TMTs). We applied this approach to measuring responses to doxorubicin, which is known to promote antitumor CD8+ T-cell responses during its therapeutic administration in cancer patients. Using both in vitro and in vivo systems, we show successful relative quantitation of MHC-I ligands using TMT-based multiplexing and demonstrate that doxorubicin induces MHC-I peptide ligands that are largely derived from mitotic progression and cell-cycle proteins. This high-throughput MHC-I ligand discovery approach may enable further explorations to understand how small molecules and other therapies alter MHC-I ligand presentation that may be harnessed for CD8+ T-cell-based immunotherapies.


Subject(s)
Antibiotics, Antineoplastic/analysis , Colonic Neoplasms/therapy , Doxorubicin/analysis , Histocompatibility Antigens Class I/analysis , Lymphoma/therapy , Animals , Antibiotics, Antineoplastic/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/immunology , Doxorubicin/pharmacology , Drug Discovery , HCT116 Cells , Histocompatibility Antigens Class I/immunology , Humans , Immunotherapy , Ligands , Lymphoma/immunology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured
15.
Haematologica ; 104(7): 1342-1354, 2019 07.
Article in English | MEDLINE | ID: mdl-30630985

ABSTRACT

Ncoa4 mediates autophagic degradation of ferritin, the cytosolic iron storage complex, to maintain intracellular iron homeostasis. Recent evidence also supports a role for Ncoa4 in systemic iron homeostasis and erythropoiesis. However, the specific contribution and temporal importance of Ncoa4-mediated ferritinophagy in regulating systemic iron homeostasis and erythropoiesis is unclear. Here, we show that Ncoa4 has a critical role in basal systemic iron homeostasis and both cell autonomous and non-autonomous roles in murine erythropoiesis. Using an inducible murine model of Ncoa4 knockout, acute systemic disruption of Ncoa4 impaired systemic iron homeostasis leading to tissue ferritin and iron accumulation, a decrease in serum iron, and anemia. Mice acutely depleted of Ncoa4 engaged the Hif2a-erythropoietin system to compensate for anemia. Mice with targeted deletion of Ncoa4 specifically in the erythroid compartment developed a pronounced anemia in the immediate postnatal stage, a mild hypochromic microcytic anemia at adult stages, and were more sensitive to hemolysis with higher requirements for the Hif2a-erythropoietin axis and extramedullary erythropoiesis during recovery. These studies demonstrate the importance of Ncoa4-mediated ferritinophagy as a regulator of systemic iron homeostasis and define the relative cell autonomous and non-autonomous contributions of Ncoa4 in supporting erythropoiesis in vivo.


Subject(s)
Anemia/pathology , Erythropoiesis , Homeostasis , Iron/metabolism , Nuclear Receptor Coactivators/physiology , Anemia/metabolism , Animals , Autophagy , Female , Hemolysis , Humans , K562 Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Coactivators/metabolism
16.
Trends Biochem Sci ; 39(2): 91-100, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24388967

ABSTRACT

Oncogenic KRAS mutation is the signature genetic event in the progression and growth of pancreatic ductal adenocarcinoma (PDAC), an almost universally fatal disease. Although it has been appreciated for some time that nearly 95% of PDAC harbor mutationally activated KRAS, to date no effective treatments that target this mutant protein have reached the clinic. A number of studies have shown that oncogenic KRAS plays a central role in controlling tumor metabolism by orchestrating multiple metabolic changes including stimulation of glucose uptake, differential channeling of glucose intermediates, reprogrammed glutamine metabolism, increased autophagy, and macropinocytosis. We review these recent findings and address how they may be applied to develop new PDAC treatments.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glutamine/metabolism , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins/metabolism , ras Proteins/metabolism , Autophagy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pinocytosis , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Signal Transduction , ras Proteins/genetics
17.
World J Surg ; 42(12): 4097-4106, 2018 12.
Article in English | MEDLINE | ID: mdl-29971463

ABSTRACT

BACKGROUND: Suitability is a patient-centered metric defined as how appropriately health information is targeted to specific populations to increase knowledge. However, suitability is most commonly evaluated exclusively by healthcare professionals without collaboration from intended audiences. Suitability (as rated by intended audiences), accuracy and readability have not been evaluated on websites discussing pancreatic cancer. METHODS: Ten healthy volunteers evaluated fifty pancreatic cancer websites using the suitability assessment of materials (SAM instrument) for the materials' overall suitability. Readability and accuracy were correlated. RESULTS: Ten recruited volunteers (ages 23-63, 50% female) found websites to be on average "adequate" or "superior" in suitability. Surgery, radiotherapy and nonprofit websites had higher suitability scores as compared to counterparts (p ≤ 0.03). There was no correlation between readability and accuracy levels and suitability scores (p ≥ 0.3). Presence of visual aids was associated with better suitability scores after controlling for website quality (p ≤ 0.01). CONCLUSION: Suitability of websites discussing pancreatic cancer treatments as rated by lay audiences differed based on therapy type and website affiliation, and was independent of readability level and accuracy of information. Nonprofit affiliation websites focusing on surgery or radiotherapy were most suitable. Online information should be assessed for suitability by target populations, in addition to readability level and accuracy, to ensure information reaches the intended audience.


Subject(s)
Comprehension , Internet , Pancreatic Neoplasms/therapy , Adult , Female , Humans , Male , Middle Aged
18.
Cancer ; 123(21): 4158-4167, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28708929

ABSTRACT

BACKGROUND: The role of conventional radiotherapy in the management of pancreatic cancer has yet to be elucidated. Over the past decade, stereotactic body radiotherapy (SBRT) has emerged as a novel therapeutic option in pancreatic cancer care. This study evaluated the survival impact of SBRT on patients with unresected pancreatic cancer. METHODS: The National Cancer Data Base was queried for unresected patients who received chemotherapy for nonmetastatic pancreatic adenocarcinoma between 2004 and 2012. Four treatment groups were identified: chemotherapy alone, chemotherapy combined with external-beam radiotherapy (EBRT), chemotherapy combined with intensity-modulated radiotherapy (IMRT), and chemotherapy combined with SBRT. Propensity score models predicting the odds of receiving SBRT were created to control for potential selection bias, and patients were matched by propensity scores. The survival analysis was performed with the Kaplan-Meier method. RESULTS: A total of 14,331 patients met the inclusion criteria. Chemotherapy alone was delivered to 5464 patients (38.1%); 6418 (44.8%), 322 (2.3%), and 2127 (14.8%) received chemotherapy along with EBRT, IMRT, and SBRT, respectively. The unadjusted median survival before matching was 9.9, 10.9, 12.0, and 13.9 months for patients treated with chemotherapy, EBRT, IMRT, and SBRT, respectively. In separate matched analyses, SBRT remained superior to chemotherapy alone (log-rank P < .0001) and EBRT (log-rank P = .0180). After matching, survival did not differ between patients receiving IMRT and patients receiving SBRT (log-rank P = .0492). CONCLUSIONS: SBRT is associated with a significantly better outcome than chemotherapy alone or in conjunction with traditional EBRT. These results support the idea that SBRT is a promising treatment approach for patients with unresected pancreatic cancer. Cancer 2017;123:4158-4167. © 2017 American Cancer Society.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/radiotherapy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/radiotherapy , Radiosurgery/mortality , Adenocarcinoma/therapy , Aged , Antineoplastic Agents/therapeutic use , Chemoradiotherapy/methods , Chemoradiotherapy/statistics & numerical data , Databases, Factual , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Pancreatic Neoplasms/therapy , Propensity Score , Radiosurgery/methods , Radiosurgery/statistics & numerical data , Radiotherapy, Intensity-Modulated/statistics & numerical data , Retrospective Studies , Selection Bias
20.
Methods Mol Biol ; 2845: 177-189, 2024.
Article in English | MEDLINE | ID: mdl-39115666

ABSTRACT

Ferritinophagy is a selective form of autophagy in which ferritin, the primary intracellular iron storage protein complex, is targeted by NCOA4 (Nuclear receptor coactivator 4) to the lysosome for degradation. NCOA4-mediated ferritinophagy plays a crucial role in cellular iron metabolism, influencing iron homeostasis, heme synthesis, mitochondrial respiratory function, and ferroptosis, an iron-dependent form of cell death. Targeting ferritinophagy has emerged as a potential anticancer therapeutic strategy. In this context, we provide a flowchart of the procedures and accompanying protocols for monitoring ferritinophagic flux.


Subject(s)
Autophagy , Ferritins , Nuclear Receptor Coactivators , Nuclear Receptor Coactivators/metabolism , Nuclear Receptor Coactivators/genetics , Ferritins/metabolism , Humans , Iron/metabolism , Lysosomes/metabolism , Animals
SELECTION OF CITATIONS
SEARCH DETAIL