27 results on '"Amy-Jo Casbon"'
Search Results
2. 259 Discovery of synthetic signaling pathway receptors that increase the potency of CAR-T cells through optimized STAT activity
- Author
-
Lan Chen, Beatriz Millare, Jeremy Chen, Aaron Cooper, Natalie Bezman, Angela Boroughs, Suchismita Mohanty, David DeTomaso, Amy-Jo Casbon, Thomas J Gardner, Anzhi Yao, Ashley Cass, Alma Gomez, Manching Ku, Lionel Berthoin, Meng Lim, Azalea Ong, Vince Thomas, Nicholas Quant, Brian Hsu, Levi Gray-Rupp, and W Nicholas Haining
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. Therapeutically targeting type I interferon directly to XCR1+ dendritic cells reveals the role of cDC1s in anti-drug antibodies
- Author
-
Paul Noe, Joy H. Wang, Kyu Chung, Zhiyong Cheng, Jessica J. Field, Xiaomeng Shen, Stephanie C. Casey, Christa L. Cortesio, Cinthia V. Pastuskovas, Hyewon Phee, Kristin V. Tarbell, Jackson G. Egen, and Amy-Jo Casbon
- Subjects
immunogenicity ,interferon ,immunotherapy ,dendritic cells ,conventional type I DCs ,antibody IFN fusion ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Conventional type 1 dendritic cells (cDC1s) are superior in antigen cross-presentation and priming CD8+ T cell anti-tumor immunity and thus, are a target of high interest for cancer immunotherapy. Type I interferon (IFN) is a potent inducer of antigen cross-presentation, but, unfortunately, shows only modest results in the clinic given the short half-life and high toxicity of current type I IFN therapies, which limit IFN exposure in the tumor. CD8+ T cell immunity is dependent on IFN signaling in cDC1s and preclinical studies suggest targeting IFN directly to cDC1s may be sufficient to drive anti-tumor immunity. Here, we engineered an anti-XCR1 antibody (Ab) and IFN mutein (IFNmut) fusion protein (XCR1Ab-IFNmut) to determine whether systemic delivery could drive selective and sustained type I IFN signaling in cDC1s leading to anti-tumor activity and, in parallel, reduced systemic toxicity. We found that the XCR1Ab-IFNmut fusion specifically enhanced cDC1 activation in the tumor and spleen compared to an untargeted control IFN. However, multiple treatments with the XCR1Ab-IFNmut fusion resulted in robust anti-drug antibodies (ADA) and loss of drug exposure. Using other cDC1-targeting Ab-IFNmut fusions, we found that localizing IFN directly to cDC1s activates their ability to promote ADA responses, regardless of the cDC1 targeting antigen. The development of ADA remains a major hurdle in immunotherapy drug development and the cellular and molecular mechanisms governing the development of ADA responses in humans is not well understood. Our results reveal a role of cDC1s in ADA generation and highlight the potential ADA challenges with targeting immunostimulatory agents to this cellular compartment.
- Published
- 2023
- Full Text
- View/download PDF
4. NLRP3 Inflammasome Mediates Dormant Neutrophil Recruitment following Sterile Lung Injury and Protects against Subsequent Bacterial Pneumonia in Mice
- Author
-
Xiaoli Tian, He Sun, Amy-Jo Casbon, Edward Lim, Kevin P. Francis, Judith Hellman, and Arun Prakash
- Subjects
lung injury ,inflammasome ,NOD- ,LRR- ,and pyrin domain-containing 3 ,interleukin-1β ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Sterile lung injury is an important clinical problem that complicates the course of severely ill patients. Interruption of blood flow, namely ischemia–reperfusion (IR), initiates a sterile inflammatory response in the lung that is believed to be maladaptive. The rationale for this study was to elucidate the molecular basis for lung IR inflammation and whether it is maladaptive or beneficial. Using a mouse model of lung IR, we demonstrate that sequential blocking of inflammasomes [specifically, NOD-, LRR-, and pyrin domain-containing 3 (NLRP3)], inflammatory caspases, and interleukin (IL)-1β, all resulted in an attenuated inflammatory response. IL-1β production appeared to predominantly originate in conjunction with alveolar type 2 epithelial cells. Lung IR injury recruited unactivated or dormant neutrophils producing less reactive oxygen species thereby challenging the notion that recruited neutrophils are terminally activated. However, lung IR inflammation was able to limit or reduce the bacterial burden from subsequent experimentally induced pneumonia. Notably, inflammasome-deficient mice were unable to alter this bacterial burden following IR. Thus, we conclude that the NLRP3 inflammasome, through IL-1β production, regulates lung IR inflammation, which includes recruitment of dormant neutrophils. The sterile IR inflammatory response appears to serve an important function in inducing resistance to subsequent bacterial pneumonia and may constitute a critical part of early host responses to infection in trauma.
- Published
- 2017
- Full Text
- View/download PDF
5. Neutrophil-specific deletion of Syk results in recruitment-independent stabilization of the barrier and a long-term improvement in cognitive function after traumatic injury to the developing brain
- Author
-
Alpa Trivedi, Kayleen G. Tercovich, Amy Jo Casbon, Jacob Raber, Clifford Lowell, and Linda J. Noble-Haeusslein
- Subjects
Neutrophils ,Spleen tyrosine kinase ,Oxidative stress ,Barrier ,Learning ,Memory ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
While traumatic brain injury (TBI) is the leading cause of death and disability in children, we have yet to identify those pathogenic events that determine the extent of recovery. Neutrophils are best known as “first responders” to sites of infection and trauma where they become fully activated, killing pathogens via proteases that are released during degranulation. However, this activational state may generate substantial toxicity in the young brain after TBI that is partially due to developmentally regulated inadequate antioxidant reserves. Neutrophil degranulation is triggered via a downstream signaling pathway that is dependent on spleen tyrosine kinase (Syk). To test the hypothesis that the activational state of neutrophils is a determinant of early pathogenesis and long-term recovery, we compared young, brain-injured conditional knockouts of Syk (sykf/fMRP8-cre+) to congenic littermates (sykf/f). Based upon flow cytometry, there was an extended recruitment of distinct leukocyte subsets, including Ly6G+/Ly6C− and Ly6G+/Ly6Cint, over the first several weeks post-injury which was similar between genotypes. Subsequent assessment of the acutely injured brain revealed a reduction in blood-brain barrier disruption to both high and low molecular weight dextrans and reactive oxygen species in sykf/fMRP8-cre+ mice compared to congenic littermates, and this was associated with greater preservation of claudin 5 and neuronal integrity, as determined by Western blot analyses. At adulthood, motor learning was less affected in brain-injured sykf/fMRP8-cre+ mice as compared to sykf/f mice. Performance in the Morris Water Maze revealed a robust improvement in hippocampal-dependent acquisition and short and long-term spatial memory retention in sykf/fMRP8-cre+ mice. Subsequent analyses of swim path lengths during hidden platform training and probe trials showed greater thigmotaxis in brain-injured sykf/f mice than sham sykf/f mice and injured sykf/fMRP8-cre+ mice. Our results establish the first mechanistic link between the activation state of neutrophils and long-term functional recovery after traumatic injury to the developing brain. These results also highlight Syk kinase as a novel therapeutic target that could be further developed for the brain-injured child.
- Published
- 2021
- Full Text
- View/download PDF
6. Brief report: STING expressed in tumor and non-tumor compartments has distinct roles in regulating anti-tumor immunity
- Author
-
Jennie C. Kim, Xian Liu, Karen Fitzgerald, Jason S. Eng, Jessica Orf, Sarah A. O’Brien, Brian Belmontes, Amy-Jo Casbon, Sergey V. Novitskiy, Kristin V. Tarbell, Jason DeVoss, and Jackson G. Egen
- Subjects
Cancer Research ,Oncology ,Immunology ,Immunology and Allergy - Abstract
Type I interferon-mediated activation of immune cells can facilitate the generation of productive tumor antigen-specific T cell responses in solid tumors. The cGAS/STING DNA sensing pathway is a critical upstream mediator of type I interferon production and is an important regulator of anti-tumor immunity. Numerous STING pathway agonists are now being tested in clinical trials, but the effectiveness of this approach is not yet clear and a better understanding of the relative importance of this pathway in various tumor settings is needed. We have evaluated syngeneic tumor models with different baseline inflammatory states to determine the contributions of STING activity in both tumor and non-tumor cellular compartments to anti-tumor immune responses. We find that productive anti-tumor immune responses in the poorly immunogenic B16F10 model show a strong dependence on STING expression in non-tumor cells. In the immunogenic MC38 model, constitutive STING activation in tumor cells can partially bypass the requirement for STING-dependent activity from immune cells. Our findings reveal multiple, context-dependent roles for STING activity in the regulation of anti-tumor immunity and the response to immunotherapy. In preclinical models where STING is basally active, checkpoint inhibition is more likely to have a therapeutic effect and removal of STING signaling from either the tumor or the non-tumor compartment has a minimal effect. Removal of STING signaling in both, however, diminishes the efficacy derived from checkpoint therapy. Further work is needed to understand the heterogeneity of STING signaling in patients, both in tumor cells and the tumor microenvironment, and the best means of harnessing this pathway to generate anti-tumor immunity and improve therapeutic outcomes.
- Published
- 2022
- Full Text
- View/download PDF
7. 1342 Therapeutic targeting of MARCO with PY265 antibody promotes myeloid cell reprogramming and unleashes anti-tumor immunity
- Author
-
Linnea Haeggblom, Subhadra Dash, Yelena Vayn, Josh Rudolph, Rachael Palmer, Xi Yang, Sergio Lacayo, Cesar Juarez, Erin Mayes, Rachel Greathouse, Kara Mojica, Carlos Santamaria, Johanna Ramoth, Ranna Mehta, Maryam Yousefi, Nadine Jahchan, Svetlana Gratch, Amy-Jo Casbon, Joshua Pollack, Marc Chamberlain, Leonard Reyno, Matthew Myers, Vladislava Juric, Linda Liang, Kristen Pierce, and Kevin Baker
- Published
- 2022
- Full Text
- View/download PDF
8. Abstract 1768: Multiplexed shRNA cassettes targeting orthogonal pathways (FAS/PTPN2/TGFBR) enhance the potency of integrated circuit T cells (ICTs) in multiple solid tumor models
- Author
-
Thomas J. Gardner, Adam Litterman, Brenal K. Singh, Luisa Silva, Mukund Hari, Stanley Zhou, Colin Tang, Sahil Joshi, John Gagnon, Oliver Takacsi-Nagy, Jason Hall, Hans Pope, James Zhang, Alma Gomez, Jeremy Chen, Suchismita Mohanty, Vince Thomas, Nicholas Quant, Beatriz Millare, Amy-Jo Casbon, Natalie Bezman, Levi Gray-Rupp, Angela C. Boroughs, and W. Nicholas Haining
- Subjects
Cancer Research ,Oncology - Abstract
T cell exhaustion resulting from chronic antigen stimulation and immunosuppression in the tumor microenvironment (TME) limits CAR T efficacy in the solid tumor setting. We have previously shown that engineering therapeutic T cell products using CRISPR-based gene insertion of a dual shRNA cassette targeting Fas and PTPN2 significantly increased antitumor efficacy of Integrated Circuit T cells (ICTs) in ovarian cancer models. We sought to build on this finding to induce additional gene perturbations that improve the efficacy of ICTs. Transforming growth factor (TGF)-b is an immunosuppressive cytokine that potently inhibits T cell responses and is present at high levels in numerous solid tumors, including renal cell carcinoma (RCC). In order to render ICT cells less susceptible to TGF-b-mediated suppression, we developed a quadruple shRNA cassette that simultaneously targets Fas, PTPN2, and TGFBR. Candidate TGFBR-targeting shRNAs were selected for their ability to reduce surface TGFBR receptor expression and impair proximal (pSMAD) or distal (CD103, PD-1) signaling through TGFBR. While single shRNAs against TGFBR did not rescue ICT cell activity in the presence of TGF-b, likely due to partial knockdown of TGFBR signaling, a cassette encoding two shRNAs against TGFBR restored ICT function to similar levels observed in the absence of TGF-b. The quadruple shRNA cassettes targeting Fas/PTPN2/TGFBR significantly enhanced antitumor activity of ICT cells in multiple xenograft tumor models relative to Fas/PTPN2 cassettes. These results demonstrate the utility of multiplexed shRNA strategies to render therapeutic T cells resistant to orthogonal suppressive pathways in solid tumors. Citation Format: Thomas J. Gardner, Adam Litterman, Brenal K. Singh, Luisa Silva, Mukund Hari, Stanley Zhou, Colin Tang, Sahil Joshi, John Gagnon, Oliver Takacsi-Nagy, Jason Hall, Hans Pope, James Zhang, Alma Gomez, Jeremy Chen, Suchismita Mohanty, Vince Thomas, Nicholas Quant, Beatriz Millare, Amy-Jo Casbon, Natalie Bezman, Levi Gray-Rupp, Angela C. Boroughs, W. Nicholas Haining. Multiplexed shRNA cassettes targeting orthogonal pathways (FAS/PTPN2/TGFBR) enhance the potency of integrated circuit T cells (ICTs) in multiple solid tumor models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1768.
- Published
- 2023
- Full Text
- View/download PDF
9. Abstract 4073: Tunable STAT activation by synthetic pathway activators (SPAs) increases engineered T-cell potency and persistence
- Author
-
Thomas J. Gardner, Beatriz Millare, Anzhi Yao, Ashley Cass, Suchismita Mohanty, Jeremy Chen, Alma Gomez, David DeTomaso, Manching Ku, Lionel Berthoin, Meng Lim, Azalea Ong, Vince Thomas, Nicholas Quant, Brian Hsu, Amy-Jo Casbon, Natalie Bezman, Aaron Cooper, Levi Gray-Rupp, Angela C. Boroughs, and W. Nicholas Haining
- Subjects
Cancer Research ,Oncology - Abstract
Clinically effective adoptive T cell therapy for the treatment of solid tumors will require robust T cell expansion, persistence, and potency. The Janus-kinase signal transducer and activator of transcription (JAK-STAT) pathway governs T cell activation and differentiation, and can thereby serve as a critical regulator of these properties. To take advantage of the benefits of STAT signaling in programming an antitumor T-cell response, we used synthetic biology to create a library of proteins, termed Synthetic Pathway Activators (SPAs) which constitutively drive STAT signaling without the need for external cytokine input. SPAs can be designed to engage activity of multiple STAT family transcription factors at variable levels through rational design. We have developed several classes of SPAs, including but not limited to Class I SPAs, which primarily increase pSTAT3 activity, and Class II SPAs, which increase pSTAT5 activity. When constitutively expressed in ArsenalBio Integrated Circuit T (ICT) cells, SPAs result in significant enhancements in T-cell potency and expansion. Repetitive stimulation assays, wherein T cells are challenged with tumor cells every 2 days, reveal that Class I SPAs result in 6-log or higher improved tumor cell clearance over a 2-week assay period. Across various mouse xenograft models, SPA-expressing ICTs reach at least 6-fold improved tumor growth inhibition. RNAseq and ATACseq analysis indicate dramatic changes to gene expression profiles in T cells expressing Class I SPAs, with maintenance of T cell stem-like phenotypes, and restricted accessibility of various exhaustion marker genes. Importantly, despite significantly increased levels of expansion, ICTs equipped with SPAs are not immortalized, showing no signs of cytokine-independent outgrowth. In addition, SPA-expressing ICT cells rapidly contract following tumor clearance in-vivo. The SPA platform represents a novel, tunable, and T cell intrinsic approach for engineering cell fates that result in potent anti-tumor properties. Citation Format: Thomas J. Gardner, Beatriz Millare, Anzhi Yao, Ashley Cass, Suchismita Mohanty, Jeremy Chen, Alma Gomez, David DeTomaso, Manching Ku, Lionel Berthoin, Meng Lim, Azalea Ong, Vince Thomas, Nicholas Quant, Brian Hsu, Amy-Jo Casbon, Natalie Bezman, Aaron Cooper, Levi Gray-Rupp, Angela C. Boroughs, W. Nicholas Haining. Tunable STAT activation by synthetic pathway activators (SPAs) increases engineered T-cell potency and persistence. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4073.
- Published
- 2023
- Full Text
- View/download PDF
10. Zena Werb (1945–2020)
- Author
-
Amy-Jo Casbon, Mikala Egeblad, Matilda F. Chan, Sylvain Provot, Laurie E. Littlepage, Kai Kessenbrock, Jonathan Chou, Hosein Kouros-Mehr, Andrew J. Ewald, Jennifer N. Lilla, Devon A. Lawson, George A. Lemieux, Joanna J. Phillips, Vicki Plaks, Bryan E. Welm, Mark D. Sternlicht, Caroline M. Alexander, and Pengfei Lu
- Subjects
Genetics ,Molecular Medicine ,Art history ,Cell Biology ,Obituary ,Biology ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology ,Developmental Biology - Published
- 2020
- Full Text
- View/download PDF
11. Neutrophil-specific deletion of Syk results in recruitment-independent stabilization of the barrier and a long-term improvement in cognitive function after traumatic injury to the developing brain
- Author
-
Amy Jo Casbon, Jacob Raber, Linda J. Noble-Haeusslein, Alpa Trivedi, Clifford A. Lowell, and Kayleen G. Tercovich
- Subjects
0301 basic medicine ,Barrier ,Traumatic brain injury ,Neutrophils ,Congenic ,Morris water navigation task ,Syk ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Pathogenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cognition ,Memory ,Morris Water Maze Test ,Brain Injuries, Traumatic ,medicine ,Animals ,Learning ,Syk Kinase ,Spatial Memory ,Mice, Knockout ,Neurons ,Thigmotaxis ,business.industry ,Degranulation ,Brain ,Spleen tyrosine kinase ,Recovery of Function ,medicine.disease ,030104 developmental biology ,Neurology ,Neutrophil Infiltration ,Oxidative stress ,Blood-Brain Barrier ,Immunology ,Neutrophil degranulation ,business ,Reactive Oxygen Species ,030217 neurology & neurosurgery ,RC321-571 - Abstract
While traumatic brain injury (TBI) is the leading cause of death and disability in children, we have yet to identify those pathogenic events that determine the extent of recovery. Neutrophils are best known as “first responders” to sites of infection and trauma where they become fully activated, killing pathogens via proteases that are released during degranulation. However, this activational state may generate substantial toxicity in the young brain after TBI that is partially due to developmentally regulated inadequate antioxidant reserves. Neutrophil degranulation is triggered via a downstream signaling pathway that is dependent on spleen tyrosine kinase (Syk). To test the hypothesis that the activational state of neutrophils is a determinant of early pathogenesis and long-term recovery, we compared young, brain-injured conditional knockouts of Syk (sykf/fMRP8-cre+) to congenic littermates (sykf/f). Based upon flow cytometry, there was an extended recruitment of distinct leukocyte subsets, including Ly6G+/Ly6C− and Ly6G+/Ly6Cint, over the first several weeks post-injury which was similar between genotypes. Subsequent assessment of the acutely injured brain revealed a reduction in blood-brain barrier disruption to both high and low molecular weight dextrans and reactive oxygen species in sykf/fMRP8-cre+ mice compared to congenic littermates, and this was associated with greater preservation of claudin 5 and neuronal integrity, as determined by Western blot analyses. At adulthood, motor learning was less affected in brain-injured sykf/fMRP8-cre+ mice as compared to sykf/f mice. Performance in the Morris Water Maze revealed a robust improvement in hippocampal-dependent acquisition and short and long-term spatial memory retention in sykf/fMRP8-cre+ mice. Subsequent analyses of swim path lengths during hidden platform training and probe trials showed greater thigmotaxis in brain-injured sykf/f mice than sham sykf/f mice and injured sykf/fMRP8-cre+ mice. Our results establish the first mechanistic link between the activation state of neutrophils and long-term functional recovery after traumatic injury to the developing brain. These results also highlight Syk kinase as a novel therapeutic target that could be further developed for the brain-injured child.
- Published
- 2021
12. HIF signaling in osteoblast-lineage cells promotes systemic breast cancer growth and metastasis in mice
- Author
-
Sylvain Provot, Koen Schepers, Claire-Sophie Devignes, Amy-Jo Casbon, Audrey Brenot, Stéphanie Fabre, Audrey Devillers, Yetki Aslan, Jonathan Chou, and Zena Werb
- Subjects
0301 basic medicine ,Chemokine ,Amino Acid Motifs ,Osteoclasts ,Ligands ,medicine.disease_cause ,bone ,CXCR4 ,Transgenic ,Metastasis ,Mice ,2.1 Biological and endogenous factors ,Aetiology ,Neoplasm Metastasis ,skin and connective tissue diseases ,Hypoxia ,Cancer ,Multidisciplinary ,Bone metastasis ,medicine.anatomical_structure ,PNAS Plus ,Disease Progression ,Stem Cell Research - Nonembryonic - Non-Human ,Female ,Hypoxia-Inducible Factor 1 ,Signal transduction ,Signal Transduction ,Stromal cell ,Green Fluorescent Proteins ,Bone Neoplasms ,Mice, Transgenic ,Biology ,alpha Subunit ,Bone and Bones ,Experimental ,03 medical and health sciences ,breast cancer ,medicine ,HIF ,metastasis ,Animals ,Cell Lineage ,Alleles ,Osteoblasts ,Mammary Neoplasms ,Mammary Neoplasms, Experimental ,Stem Cell Research ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Chemokine CXCL12 ,030104 developmental biology ,Cancer research ,biology.protein ,Bone marrow ,Carcinogenesis - Abstract
Bone metastasis involves dynamic interplay between tumor cells and the local stromal environment. In bones, local hypoxia and activation of the hypoxia-inducible factor (HIF)-1α in osteoblasts are essential to maintain skeletal homeostasis. However, the role of osteoblast-specific HIF signaling in cancer metastasis is unknown. Here, we show that osteoprogenitor cells (OPCs) are located in hypoxic niches in the bone marrow and that activation of HIF signaling in these cells increases bone mass and favors breast cancer metastasis to bone locally. Remarkably, HIF signaling in osteoblast-lineage cells also promotes breast cancer growth and dissemination remotely, in the lungs and in other tissues distant from bones. Mechanistically, we found that activation of HIF signaling in OPCs increases blood levels of the chemokine C-X-C motif ligand 12 (CXCL12), which leads to a systemic increase of breast cancer cell proliferation and dissemination through direct activation of the CXCR4 receptor. Hence, our data reveal a previously unrecognized role of the hypoxic osteogenic niche in promoting tumorigenesis beyond the local bone microenvironment. They also support the concept that the skeleton is an important regulator of the systemic tumor environment.
- Published
- 2018
- Full Text
- View/download PDF
13. Clonal Deletion of Tumor-Specific T Cells by Interferon-γ Confers Therapeutic Resistance to Combination Immune Checkpoint Blockade
- Author
-
E Liu, Gillian A. Kinsbury, Katsunobu Hagihara, Adil Daud, Amy Jo Casbon, Marcella Fasso, Michel DuPage, Katy K. Tsai, Ravi Sachidanandam, Xiaoqing Lu, Kole T. Roybal, Li Zhang, Ingrid Lin, Jane Seagal, David Y. Oh, Mingyi Chen, Chanhyuk Park, Xiao X. Wei, Anitha D. Jayaprakash, Chien-Chun Steven Pai, Lawrence Fong, Anthony Chang, Whitney Tamaki, John Ting Wei Huang, Donald M. Simons, Serena S. Kwek, and Clint Wu
- Subjects
Male ,0301 basic medicine ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,Drug Resistance ,Inbred C57BL ,activation-induced cell death ,Mice ,0302 clinical medicine ,Neoplasms ,Monoclonal ,Immunology and Allergy ,CTLA-4 Antigen ,Receptor ,IFN-γ ,Mice, Knockout ,Tumor ,Melanoma ,Antibodies, Monoclonal ,Tumor Burden ,Infectious Diseases ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,immunotherapy ,Immunotherapy ,Development of treatments and therapeutic interventions ,Combination therapy ,Cell Survival ,Knockout ,Immunology ,Clonal Deletion ,Biology ,Antibodies ,Article ,Clonal deletion ,Cell Line ,Vaccine Related ,Experimental ,Interferon-gamma ,03 medical and health sciences ,Immunity ,Cell Line, Tumor ,medicine ,Animals ,cancer ,Humans ,Inflammatory and immune system ,Neoplasms, Experimental ,medicine.disease ,Immune checkpoint ,Blockade ,Mice, Inbred C57BL ,030104 developmental biology ,Drug Resistance, Neoplasm ,Apoptosis ,anti-CTLA-4 ,Cancer research ,Neoplasm ,anti-PD-1 ,Immunization - Abstract
Resistance to checkpoint-blockade treatments is a challenge in the clinic. We found that although treatment with combined anti-CTLA-4 and anti-PD-1 improved control of established tumors, this combination compromised anti-tumor immunity in the low tumor burden (LTB) state in pre-clinical models as well as in melanoma patients. Activated tumor-specific Tcells expressed higher amounts of interferon-γ (IFN-γ) receptor and were more susceptible to apoptosis than naive Tcells. Combination treatment induced deletion of tumor-specific Tcells and altered the Tcell repertoire landscape, skewing the distribution of Tcells toward lower-frequency clonotypes. Additionally, combination therapy induced higher IFN-γ production in the LTB state than in the high tumor burden (HTB) state on a per-cell basis, reflecting a less exhausted immune status in the LTB state. Thus, elevated IFN-γ secretion in the LTB state contributes to the development of an immune-intrinsic mechanism of resistance to combination checkpoint blockade, highlighting the importance of achieving the optimal magnitude of immune stimulation for successful combination immunotherapy strategies.
- Published
- 2019
- Full Text
- View/download PDF
14. Abstract 2981: Clonal deletion of tumor-specific T cells by combination checkpoint blockade compromises antitumor efficacy in low tumor burden states
- Author
-
Chien-Chun Steven Pai, Gillian A. Kinsbury, Lawrence Fong, Amy-Jo Casbon, Donald M. Simons, Mingyi Chen, Serena S. Kwek, Michel DuPage, Kole T. Roybal, and Xiaoqing Lu
- Subjects
Cancer Research ,Combination therapy ,business.industry ,medicine.medical_treatment ,Melanoma ,Cancer ,Immunotherapy ,medicine.disease ,Immune checkpoint ,Clonal deletion ,Blockade ,Immune system ,Oncology ,medicine ,Cancer research ,business - Abstract
A myriad of combination immunotherapies is being developed as cancer treatments with the goal of enhancing antitumor immunity. We found that depending on the disease setting, combined immune checkpoint blockade may not always lead to synergistic antitumor effects. While combined anti-CTLA-4 and anti-PD-1 improves control of established tumors, this combination can paradoxically compromise antitumor immunity in the low tumor burden state in preclinical models as well as in melanoma patients. This paradoxical outcome results from treatment-induced apoptosis of tumor-specific T cells. These changes further alter the overall T-cell receptor repertoire. Activated tumor-specific T cells express higher levels of IFN-γ receptor and are more susceptible to apoptosis. Deficiency of IFN-γ receptor on immune cells rescues this phenotype and restores antitumor activity. Additionally, tumor-specific T cells lacking the IFN-γ receptor demonstrate a significant survival advantage compared to their wild-type counterparts in tumor-bearing mice receiving combination therapy. Finally, combination therapy induces significantly higher levels of IFN-γ in the low versus high tumor burden state on a per cell basis, reflecting their less exhausted immune status. Thus, the optimal immunotherapy strategy may depend on disease context, and may not always favor more potent combination immunotherapies. Citation Format: Chien-Chun Steven Pai, Xiaoqing Lu, Donald Simons, Michel DuPage, Kole T. Roybal, Mingyi Chen, Serena Kwek, Amy-Jo Casbon, Gillian A. Kinsbury, Lawrence Fong. Clonal deletion of tumor-specific T cells by combination checkpoint blockade compromises antitumor efficacy in low tumor burden states [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2981.
- Published
- 2018
- Full Text
- View/download PDF
15. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils
- Author
-
Emmanuelle Passegué, Amy-Jo Casbon, Emily Khuc, Koen Schepers, Chanhyuk Park, Damien Reynaud, Dennis D. Gan, and Zena Werb
- Subjects
Myeloid ,Neutrophils ,Knockout ,Breast Neoplasms ,Biology ,Inbred C57BL ,stem cell biology ,Cell Line ,Immune tolerance ,immunology ,Mice ,Stem Cell Research - Nonembryonic - Human ,Cell Line, Tumor ,Granulocyte Colony-Stimulating Factor ,Receptors ,Breast Cancer ,Immune Tolerance ,medicine ,Animals ,cancer ,2.1 Biological and endogenous factors ,Myeloid Cells ,Neoplasm Invasiveness ,Mice, Knockout ,Tumor ,Multidisciplinary ,Hematology ,Flow Cytometry ,myeloid-derived suppressor cells ,Stem Cell Research ,hematopoiesis ,Hematopoiesis ,Mice, Inbred C57BL ,Haematopoiesis ,medicine.anatomical_structure ,PNAS Plus ,Bromodeoxyuridine ,Tumor progression ,Receptors, Granulocyte Colony-Stimulating Factor ,Immunology ,Myeloid-derived Suppressor Cell ,Erythropoiesis ,Female ,Stem Cell Research - Nonembryonic - Non-Human ,Bone marrow ,Stem cell - Abstract
Expansion of myeloid cells associated with solid tumor development is a key contributor to neoplastic progression. Despite their clinical relevance, the mechanisms controlling myeloid cell production and activity in cancer remains poorly understood. Using a multistage mouse model of breast cancer, we show that production of atypical T cell-suppressive neutrophils occurs during early tumor progression, at the onset of malignant conversion, and that these cells preferentially accumulate in peripheral tissues but not in the primary tumor. Production of these cells results from activation of a myeloid differentiation program in bone marrow (BM) by a novel mechanism in which tumor-derived granulocyte-colony stimulating factor (G-CSF) directs expansion and differentiation of hematopoietic stem cells to skew hematopoiesis toward the myeloid lineage. Chronic skewing of myeloid production occurred in parallel to a decrease in erythropoiesis in BM in mice with progressive disease. Significantly, we reveal that prolonged G-CSF stimulation is both necessary and sufficient for the distinguishing characteristics of tumor-induced immunosuppressive neutrophils. These results demonstrate that prolonged G-CSF may be responsible for both the development and activity of immunosuppressive neutrophils in cancer.
- Published
- 2015
- Full Text
- View/download PDF
16. Delineating CSF-1-dependent regulation of myeloid cell diversity in tumors
- Author
-
Amy-Jo Casbon, Marja Lohela, and Zena Werb
- Subjects
Myeloid ,matrix metalloproteinase ,Cell ,Population ,Immunology ,Oncology and Carcinogenesis ,Matrix metalloproteinase ,Malignancy ,Breast cancer ,breast cancer ,Immunology and Allergy ,Medicine ,tumor microenvironment ,education ,Author's View ,Cancer ,Tumor microenvironment ,education.field_of_study ,MMP ,business.industry ,tumor-associated macrophages ,Intravital Imaging ,medicine.disease ,M-CSF ,medicine.anatomical_structure ,Oncology ,TAM ,business - Abstract
© 2015, Taylor & Francis Group, LLC. Myeloid cells contribute to increased malignancy and poor prognosis in breast cancer. We demonstrate that anti-CSF-1R therapy depletes a cell population sharing characteristics of tumor-associated macrophages (TAMs) and dendritic cells (DCs). Intravital imaging combined with cellular characterization has refined our understanding of anti-CSF-1R therapy on the tumor microenvironment.
- Published
- 2015
- Full Text
- View/download PDF
17. La signalisation HIF dans les ostéoprogéniteurs stimule la croissance et la dissémination des tumeurs mammaires
- Author
-
Amy-Jo Casbon, Koen Schepers, Stéphanie Fabre, Audrey Brenot, Sylvain Provot, Jonathan Chou, Y. Aslan, Claire-Sophie Devignes, Zena Werb, and A. Devillers
- Subjects
Rheumatology - Published
- 2016
- Full Text
- View/download PDF
18. Intravital imaging reveals distinct responses of depleting dynamic tumor-associated macrophage and dendritic cell subpopulations
- Author
-
Ning Weng, Marja Lohela, Zena Werb, Lynn Bonham, Aleksandra Olow, Jeffrey Smith, Daniel Branstetter, and Amy-Jo Casbon
- Subjects
Stromal cell ,Myeloid ,Lung Neoplasms ,matrix metalloproteinase ,Neutrophils ,medicine.medical_treatment ,Receptor, Macrophage Colony-Stimulating Factor ,Tumor-associated macrophage ,Biology ,Targeted therapy ,Metastasis ,Mice ,Experimental ,Breast Cancer ,medicine ,Tumor Microenvironment ,Animals ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,CSF-1 receptor ,Tumor microenvironment ,Multidisciplinary ,Macrophages ,Macrophage Colony-Stimulating Factor ,lung metastasis ,Mammary Neoplasms ,Mammary Neoplasms, Experimental ,Dendritic cell ,medicine.disease ,Endocytosis ,Cell biology ,medicine.anatomical_structure ,PNAS Plus ,inflammation ,myeloid cells ,Female ,Intravital microscopy ,Cell Division ,Receptor - Abstract
Tumor-infiltrating inflammatory cells comprise a major part of the stromal microenvironment and support cancer progression by multiple mechanisms. High numbers of tumor myeloid cells correlate with poor prognosis in breast cancer and are coupled with the angiogenic switch and malignant progression. However, the specific roles and regulation of heterogeneous tumor myeloid populations are incompletely understood. CSF-1 is a major myeloid cell mitogen, and signaling through its receptor CSF-1R is also linked to poor outcomes. To characterize myeloid cell function in tumors, we combined confocal intravital microscopy with depletion of CSF-1R-dependent cells using a neutralizing CSF-1R antibody in the mouse mammary tumor virus long-terminal region-driven polyoma middle T antigen breast cancer model. The depleted cells shared markers of tumor-associated macrophages and dendritic cells (M-DCs), matching the phenotype of tumor dendritic cells that take up antigens and interact with T cells. We defined functional subgroups within the M-DC population by imaging endocytic and matrix metalloproteinase activity. Anti-CSF-1R treatment altered stromal dynamics and impaired both survival of M-DCs and accumulation of new M-DCs, but did not deplete Gr-1(+) neutrophils or block doxorubicin-induced myeloid cell recruitment, and had a minimal effect on lung myeloid cells. Nevertheless, prolonged treatment led to delayed tumor growth, reduced vascularity, and decreased lung metastasis. Because the myeloid infiltrate in metastatic lungs differed significantly from that in mammary tumors, the reduction in metastasis may result from the impact on primary tumors. The combination of functional analysis by intravital imaging with cellular characterization has refined our understanding of the effects of experimental targeted therapies on the tumor microenvironment.
- Published
- 2014
19. Balancing the innate immune system in tumor development
- Author
-
Amy-Jo Casbon, Catharina Hagerling, and Zena Werb
- Subjects
Tumor initiation ,Biology ,Cell Transformation ,Medical and Health Sciences ,Article ,Classical complement pathway ,Immune system ,cancer development ,Neoplasms ,Tumor Microenvironment ,Innate ,2.1 Biological and endogenous factors ,Humans ,Aetiology ,innate immune cells ,Cancer ,Inflammation ,Tumor microenvironment ,Neoplastic ,Innate immune system ,Inflammatory and immune system ,Innate lymphoid cell ,CCL18 ,Immunity ,Cell Biology ,Biological Sciences ,Immunity, Innate ,Cell Transformation, Neoplastic ,Tumor progression ,Immunology ,Disease Progression ,Developmental Biology - Abstract
Cells of the innate immune system have a dual role in cancer development in both tumor initiation and progression. Innate immune cells can, on the one hand, aid malignant transformation and tumor outgrowth and, on the other hand, prevent tumor progression. The innate immune system has the ability to tune the inflammatory response and is a key player in cancer-related inflammation, which can precede the development of malignancy or be induced by oncogenic changes promoting a protumor inflammatory milieu. In this review, we discuss the emerging cellular and molecular mechanisms of the innate immune system and inflammation in tumor initiation and progression, and point to the outstanding questions that remain.
- Published
- 2014
20. Protective effects of matrix metalloproteinase-12 following corneal injury
- Author
-
Matilda F. Chan, Inna Maltseva, Anthony Bertrand, Amy-Jo Casbon, Zena Werb, Jing Li, and Jeffrey H. Lin
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Chemokine CXCL1 ,Neovascularization, Physiologic ,Inflammation ,Biology ,Matrix metalloproteinase ,Corneal inflammation ,Collagen Type I ,Extracellular matrix ,Cornea ,Mice ,Fibrosis ,Matrix Metalloproteinase 12 ,medicine ,Animals ,Chemokine CCL2 ,Bone Marrow Transplantation ,Mice, Knockout ,Wound Healing ,Macrophages ,Cell Biology ,medicine.disease ,Actins ,CXCL1 ,Neutrophil Infiltration ,Immunology ,Female ,sense organs ,medicine.symptom ,Wound healing ,Corneal Injuries ,Research Article - Abstract
Corneal scarring due to injury is a leading cause of blindness worldwide and results from dysregulated inflammation and angiogenesis during wound healing. Here we demonstrate that the extracellular matrix metalloproteinase MMP12 (macrophage metalloelastase) is an important regulator of these repair processes. Chemical injury resulted in higher expression of the fibrotic markers α-smooth muscle actin and type I collagen, and increased levels of angiogenesis in corneas of Mmp12(-/-) mice compared with corneas of wild-type mice. In vivo, we observed altered immune cell dynamics in Mmp12(-/-) corneas by confocal imaging. We determined that the altered dynamics were the result of an altered inflammatory response, with delayed neutrophil infiltration during the first day and excessive macrophage infiltration 6 days later, mediated by altered expression levels of chemokines CXCL1 and CCL2, respectively. Corneal repair returned to normal upon inhibition of these chemokines. Taken together, these data show that MMP12 has a protective effect on corneal fibrosis during wound repair through regulation of immune cell infiltration and angiogenesis.
- Published
- 2013
21. Effects of IFN-γ on intracellular trafficking and activity of macrophage NADPH oxidase flavocytochrome b558
- Author
-
Amy Jo Casbon, Kenneth W. Dunn, Mary C. Dinauer, Lee-Ann H. Allen, and Matthew E. Long
- Subjects
Time Factors ,Phagocyte ,Endosome ,Phagocytosis ,Immunology ,Cell Line ,chemistry.chemical_compound ,Interferon-gamma ,Mice ,Cell Movement ,medicine ,Immunology and Allergy ,Macrophage ,Animals ,NADPH oxidase ,biology ,Dose-Response Relationship, Drug ,Superoxide ,Macrophages ,NADPH Oxidases ,Cell Biology ,Host Defense & Pathophysiology ,Cytochrome b Group ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Biochemistry ,chemistry ,biology.protein ,Tetradecanoylphorbol Acetate ,P22phox ,Intracellular - Abstract
IFNγ regulates trafficking and synthesis of flavocytochrome b558, suggesting a role to control superoxide production in macrophages. Flavocytochrome b558, the catalytic core of the phagocyte NADPH oxidase (NOX2), mediates electron transfer from NADPH to molecular oxygen to generate superoxide, the precursor of highly ROS for host defense. Flavocytochrome b558 is an integral membrane heterodimer consisting of a large glycosylated subunit, gp91phox, and a smaller subunit, p22phox. We recently showed in murine macrophages that flavocytochrome b558 localizes to the PM and Rab11-positive recycling endosomes, whereas in primary hMDMs, gp91phox and p22phox reside in the PM and the ER. The antimicrobial activity of macrophages, including ROS production, is greatly enhanced by IFN-γ, but how this is achieved is incompletely understood. To further define the mechanisms by which IFN-γ enhances macrophage NADPH oxidase activity, we evaluated changes in flavocytochrome b558 expression and localization, along with NADPH oxidase activity, in IFN-γ stimulated RAW 264.7 cells and primary murine BMDMs and hMDMs. We found that enhanced capacity for ROS production is, in part, a result of increased protein expression of gp91phox and p22phox but also demonstrate that IFN-γ induced a shift in the predominant localization of gp91phox and p22phox from intracellular membrane compartments to the PM. Our results are the first to show that a cytokine can change the distribution of macrophage flavocytochrome b558 and provide a potential, new mechanism by which IFN-γ modulates macrophage antimicrobial activity. Altogether, our data suggest that the mechanisms by which IFN-γ regulates antimicrobial activity of macrophages are more complex than previously appreciated.
- Published
- 2012
22. Macrophage NADPH oxidase flavocytochrome B localizes to the plasma membrane and Rab11-positive recycling endosomes
- Author
-
Kenneth W. Dunn, Mary C. Dinauer, Amy Jo Casbon, and Lee-Ann H. Allen
- Subjects
Endosome ,Immunology ,Blotting, Western ,Fluorescent Antibody Technique ,CHO Cells ,Endosomes ,Article ,Cell membrane ,Mice ,Cricetulus ,Cricetinae ,medicine ,Image Processing, Computer-Assisted ,Immunology and Allergy ,Animals ,Humans ,Transgenes ,Phagosome ,NADPH oxidase ,Microscopy, Confocal ,biology ,Chinese hamster ovary cell ,Endoplasmic reticulum ,Macrophages ,Cell Membrane ,NADPH Oxidases ,Cytochrome b Group ,Flow Cytometry ,Cell biology ,Transport protein ,Protein Transport ,medicine.anatomical_structure ,rab GTP-Binding Proteins ,biology.protein ,Intracellular - Abstract
Flavocytochrome b558, the catalytic core of the phagocytic NADPH oxidase, mediates the transfer of electrons from NADPH to molecular oxygen to generate superoxide for host defense. Flavocytochrome b is a membrane heterodimer consisting of a large subunit gp91phox (NOX2) and a smaller subunit, p22phox. Although in neutrophils flavocytochrome b has been shown to localize to the plasma membrane and specific granules, little is known about its distribution in macrophages. Using immunofluorescent staining and live cell imaging of fluorescently tagged gp91phox and p22phox, we demonstrate in a Chinese hamster ovary cell model system and in RAW 264.7 and primary murine bone marrow-derived macrophages that flavocytochrome b is found in the Rab11-positive recycling endocytic compartment, as well as in Rab5-positive early endosomes and plasma membrane. Additionally, we show that unassembled p22phox and gp91phox subunits localize to the endoplasmic reticulum, which redistribute to the cell surface and endosomal compartments following heterodimer formation. These studies show for the first time that flavocytochrome b localizes to intracellular compartments in macrophages that recycle to the plasma membrane, which may act as a reservoir to deliver flavocytochrome b to the cell surface and phagosome membranes.
- Published
- 2009
23. Abstract IA09: Building the metastatic microenvironment
- Author
-
Amy-Jo Casbon, Zena Werb, and Vicki Plaks
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Tumor microenvironment ,Stromal cell ,Wnt signaling pathway ,Biology ,medicine.disease ,Primary tumor ,Metastasis ,Extracellular matrix ,Oncology ,Cancer cell ,Cancer research ,medicine ,Stem cell - Abstract
The major cause of cancer-associated mortality is metastasis, but our understanding of this process is far from complete. Tumor cells invade the surrounding tissue of the primary tumor, intravasate into blood and lymphatic vessels, survive and translocate to distant tissues, extravasate, adapt to the new microenvironment and eventually seed, proliferate and colonize to form metastases. Nearly 125 years ago, Paget enunciated the seed and soil hypothesis of cancer. More recently, emerging data suggested that the cellular and extracellular matrix (ECM) microenvironments—both in the primary tumor and in metastatic sites—are crucial at multiple stages of metastasis. Significantly, targeting the tumor microenvironment in metastasis might hold promise for therapy as stromal cells are not mutated and the effects may be widespread as the ECM interacts with multiple tumor cells. The ECM, which is a rich reservoir of pro- and anti-angiogenic cues that regulate neovascularization of the tumor, a crucial process in tumor cell dissemination. The developing metastatic lesions result from a complex crosstalk between disseminating tumor cells and the different players in the microenvironment of the metastatic lesion. The specific recruitment of distinct populations of leukocytes and stromal cells with overlapping functions in metastasis, may open new avenues to the development of metastasis-targeted therapies. Finally, the function of the tumor microenvironment in modulating sensitivity to chemotherapy is of clinical importance. But the aspects of the microenvironment contribute to loss of drug efficacy are still poorly understood in the metastatic setting. The microenvironment is an important source of anticancer drug resistance and also a therapeutic target, as drugs might not need to be completely penetrant to be effective because altering some immune cells, ECM components or the vasculature may have profound effects as seen with regulation of microRNAs. The microenvironment faced by cells at metastatic sites also determines whether these cells die, proliferate or become dormant. How does the microenvironment evolve from the pre-metastatic stage to established metastases? Determining whether the metastatic niche arises from changes in the ECM, decreased immune surveillance or changes in specific pro-inflammatory molecules poses a challenge for the future. We need a more complete understanding of the role of the metastatic microenvironment to uncover how these processes promote metastasis. Chou, J., J.H. Lin, A. Brenot, J.-w. Kim, S. Provot & Z. Werb (2013). GATA3 suppresses metastasis and modulates the tumor microenvironment by regulating miR-29 expression. Nat. Cell Biol. 15: 201-213. PMCID: PMC3660859. Egeblad, M., A. J. Ewald, H. A. Askautrud, B. E. Welm, M. Truitt, E. Bainbridge, G. Peeters, M. Krummell & Z. Werb (2008). Visualizing stromal cell dynamics in different tumor microenvironments by spinning disk confocal microscopy. Dis. Model. Mech. 1:155-167. PMCID: PMC2562195. Kessenbrock, K., G.J.P. Dijkgraaf, D. A. Lawson, L. E. Littlepage, P. Shahi, U. Pieper & Z. Werb (2013). A role for matrix metalloproteinases in regulating mammary stem cell function via the Wnt signaling pathway. Cell Stem Cell. 13:300-313. PMCID: PMC3769456. Lu, P., V. M. Weaver & Z. Werb (2012). Extracellular matrix: a dynamic niche component during cancer progression. J. Cell Biol. 196:396-406. PMCID: PMC3283993. Nakasone, E., H. A.Askautrud, T. Kees, V. Plaks, A. J. Ewald, M. G. Rasch, Y. X. Tan, J. Qin, M. Fein, J. Park, P. Sinha, M. J. Bissell, E. Frengen, Z. Werb & M. Egeblad (2012). Imaging tumor-stroma interactions during chemotherapy reveals microenvironmental contributions to chemoresistance. Cancer Cell. 21:488-503. PMCID: PMC3332002. Citation Format: Amy-Jo Casbon, Vicki Plaks, Zena Werb. Building the metastatic microenvironment. [abstract]. In: Abstracts: AACR Special Conference on Cellular Heterogeneity in the Tumor Microenvironment; 2014 Feb 26-Mar 1; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(1 Suppl):Abstract nr IA09. doi:10.1158/1538-7445.CHTME14-IA09
- Published
- 2015
- Full Text
- View/download PDF
24. Deletion mutagenesis of p22phox subunit of flavocytochrome b558: identification of regions critical for gp91phox maturation and NADPH oxidase activity
- Author
-
Yanmin, Zhu, Christophe C, Marchal, Amy-Jo, Casbon, Natalie, Stull, Katharina, von Löhneysen, Ulla G, Knaus, Algirdas J, Jesaitis, Sally, McCormick, William M, Nauseef, and Mary C, Dinauer
- Subjects
Membrane Glycoproteins ,Sequence Homology, Amino Acid ,Mutagenesis ,Cell Membrane ,Molecular Sequence Data ,NADPH Oxidase 2 ,Animals ,Humans ,NADPH Oxidases ,Cattle ,Amino Acid Sequence ,Cytochrome b Group ,Gene Deletion - Abstract
The heterodimeric flavocytochrome b558, comprised of the two integral membrane proteins p22phox and gp91phox, mediates the transfer of electrons from NADPH to molecular oxygen in the phagocyte NADPH oxidase to generate the superoxide precursor of microbicidal oxidants. This study uses deletion mutagenesis to identify regions of p22phox required for maturation of gp91phox and for NADPH oxidase activity. N-terminal, C-terminal, or internal deletions of human p22phox were generated and expressed in Chinese hamster ovary cells with transgenes for gp91phox and two other NADPH oxidase subunits, p47phox, and p67phox. The results demonstrate that p22phox-dependent maturation of gp91phox carbohydrate, cell surface expression of gp91phox, and the enzymatic function of flavocytochrome b558 are closely correlated. Whereas the 5 N-terminal and 25 C-terminal amino acids are dispensable for these functions, the N-terminal 11 amino acids of p22phox are required, as is a hydrophilic region between amino acids 65 and 90. Upon deletion of 54 residues at the C terminus of p22phox (amino acids 142-195), maturation and cell surface expression of gp91phox was still preserved, although NADPH oxidase activity was absent, as expected, due to removal of a proline-rich domain between amino acids 151-160 that is required for recruitment of p47phox. Antibody binding studies indicate that the extreme N terminus of p22phox is inaccessible in the absence of cell permeabilization, supporting a model in which both the N- and C-terminal domains of p22phox extend into the cytoplasm, anchored by two membrane-embedded regions.
- Published
- 2006
25. Abstract IA24: How tumors regulate their microenvironment in primary sites and metastasis
- Author
-
Amy Jo Casbon, Vicki Plaks, Marja Lohela, and Zena Werb
- Subjects
Cancer Research ,Tumor microenvironment ,Stromal cell ,Angiogenesis ,Cancer ,Biology ,Cell cycle ,medicine.disease ,Primary tumor ,Metastasis ,Oncology ,Cancer cell ,medicine ,Cancer research ,Molecular Biology - Abstract
In 1863, Virchow hypothesized that the cancer originated at sites of chronic tissue injury and that the ensuing inflammation they cause enhances cell proliferation. While it is now clear that proliferation of tumor cells alone does not cause cancer, sustained cell proliferation in an environment rich in inflammatory cells, growth factors, activated stroma certainly potentiates and/or promotes neoplastic risk and leads to metastasis. In fact, sometimes the trigger for neoplastic progression may come from signals within the stromal microenvironment. The stromal microenvironment consists of fibroblasts, macrophages, neutrophils, vascular components, nerves and inflammatory cells of the innate and acquired immune response, as well as the extracellular matrix that they elaborate along with the molecules that are concentrated and immobilized on it. All of these components communicate with each other and with the neoplastic cells to contribute to the aberrant tumor organ. Factors elaborated by tumors cause dramatic increases in inflammatory myeloid cells in the microenvironment of the primary tumor as well as in distant sites upon the malignant conversion of the tumor cells. These myeloid cells modulate the tumor ecology and regulate metastatic take, altering vascular permeability and responses to chemotherapy. The dynamic interplay of tumor cells and host cells responding to the tumor in the microenvironment contribute to tumor evolution, metastasis and evasion of therapeutic responses. Supported by grants from the National Cancer Institute. Chou, J., J.H. Lin, A. Brenot, J.-w. Kim, S. Provot & Z. Werb (2013). GATA3 suppresses metastasis and modulates the tumor microenvironment by regulating miR-29 expression. Nat. Cell Biol. 15: 201-213. [2013 Jan 27, PMCID: PMC3660859. Chou, J., P. Shahi & Z. Werb (2013). MicroRNA-mediated regulation of the tumor microenvironment. Cell Cycle. In press. Egeblad, M., A. J. Ewald, H. A. Askautrud, B. E. Welm, M. Truitt, E. Bainbridge, G. Peeters, M. Krummell & Z. Werb (2008). Visualizing stromal cell dynamics in different tumor microenvironments by spinning disk confocal microscopy. Dis. Model. Mech. 1:155-167. PMCID: PMC2562195. Egeblad, M., N. Nakasone & Z. Werb (2010). Tumors as organs: complex tissues that interface with the entire organism. Dev. Cell. 18:884-901. PMCID: PMC2905377. Kessenbrock, K., V. Plaks & Z. Werb (2010). Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell. 141: 52-67. PMCID: PMC286205. Kessenbrock, K., G.J.P. Dijkgraaf, D. A. Lawson, L. E. Littlepage, P. Shahi, U. Pieper & Z. Werb (2013). A role for matrix metalloproteinases in regulating mammary stem cell function via the Wnt signaling pathway. Cell Stem Cell. [Epub ahead of print July 18]. PMID: 23871604. Kouros-Mehr, H., S. K. Bechis, E.M. Slorach, L. E. Littlepage, M. Egeblad, A. J. Ewald, S.-Y. Pai, I-C. Ho & Z. Werb (2008). GATA-3 links tumor differentiation and dissemination in a luminal breast cancer model. Cancer Cell. 13:141-152. PMCID: PMC2262951. Littlepage, L. E., M. D. Sternlicht, N. Rougier, J. Phillips, E. Gallo, Y. Yu, K. Williams, A. Brenot, J. I. Gordon & Z. Werb (2010). Matrix metalloproteinases contribute distinct roles in neuroendocrine prostate carcinogenesis, metastasis, and angiogenesis progression. Cancer Res. 70:2224-2234. PMCID: PMC2840052. Littlepage, L.E., A. S. Adler, H. Kouros-Mehr, G. Huang, J. Chou, S. R. Krig, O. L. Griffith, J. E. Korkola, K. Qu, D. A. Lawson, Q. Xue, M. D. Sternlicht, G. J. P. Dijkgraaf, P. Yaswen, H. S. Rugo, C. A. Sweeney, C. C. Collins, J. W. Gray, H. Y. Chang & Z. Werb (2012). The transcription factor ZNF217 is a prognostic biomarker and therapeutic target during breast cancer progression. Cancer Discov. 2 : 638-651. PMCID : PMC3546490. Lu, P., V. M. Weaver & Z. Werb (2012). Extracellular matrix: a dynamic niche component during cancer progression. J. Cell Biol. 196:396-406. PMCID: PMC3283993. Nakasone, E., H. A.Askautrud, T. Kees, V. Plaks, A. J. Ewald, M. G. Rasch, Y. X. Tan, J. Qin, M. Fein, J. Park, P. Sinha, M. J. Bissell, E. Frengen, Z. Werb & M. Egeblad (2012). Imaging tumor-stroma interactions during chemotherapy reveals microenvironmental contributions to chemoresistance. Cancer Cell. 21:488-503. PMCID: PMC3332002. Nguyen-Ngoc, K.-V., K. J. Cheung, A. Brenot, E. R. Shamir, R. S. Gray, W. C. Hines, P. Yaswen, Z. Werb* & A. J. Ewald* (2012). The ECM microenvironment regulates collective migration and local dissemination in normal and malignant mammary epithelium. Proc. Natl. Acad. Sci. USA. 109:E2595-E2604. PMCID: PMC3465416. Sounni, N. E., K. Dehne, L. van Kempen, M. Egeblad, N.I. Affara, I. Cuevas, J. Wiesen, J. Junankar, L. Korets, L. Lee, J. Shen, C. J. Morrison, C. M. Overall, S. M. Krane, Z. Werb, N. Boudreau & L. M. Coussens (2010). Stromal regulation of vessel stability by MMP9 and TGFβ. Dis. Model. Mech. 3: 317-332. PMCID: PMC20223936. Citation Format: Amy Jo Casbon, Marja Lohela, Vicki Plaks, Zena Werb. How tumors regulate their microenvironment in primary sites and metastasis. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research: Genetics, Biology, and Clinical Applications; Oct 3-6, 2013; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2013;11(10 Suppl):Abstract nr IA24.
- Published
- 2013
- Full Text
- View/download PDF
26. Abstract IA2: Microenvironmental control of bone metastasis
- Author
-
Audrey Brenot, Sylvain Provot, Zena Werb, Ying Yu, and Amy-Jo Casbon
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Bone metastasis ,Cancer ,Osteoblast ,medicine.disease ,Metastasis ,Haematopoiesis ,medicine.anatomical_structure ,Endocrinology ,Breast cancer ,Oncology ,Internal medicine ,Conditional gene knockout ,medicine ,Cancer research ,Bone marrow ,business - Abstract
Breast cancer is often associated with a poor prognosis largely due to its tendency to metastasize. Breast cancer preferentially metastasizes to the skeleton. Since Paget's seed and soil hypothesis from more than a century ago, it is known that bones constitute a very hospitable soil that attracts and allows breast cancer cells to thrive. Surprisingly, however, only a few studies have addressed the role of the bone microenvironment in bone metastasis, and most of them emphasized the role of osteoclasts (bone-destructing cells). Osteoclasts are known to stimulate bone metastasis because of their bone resorbing activity, which releases into the bone marrow space growth factors normally embedded in the bone matrix. On the other hand, the role of osteoblasts (bone-making cells) in this process has not been addressed directly. There is growing evidence that osteoblasts' function is not limited to making bone. In particular, these cells regulate various aspects of hematopoiesis, insulin secretion in the pancreas, or even male fertility in mice. We hypothesized that osteoblasts play an important role in breast cancer metastasis to bone. We tested this possibility by inoculating a breast cancer cell line (PyMT-FVB-Luc+ cells) into either the mammary fat pad, or directly into the blood stream of syngeneic recipient mice, which presented either an increased or a decreased osteoblast number. Hypoxia and the hypoxia inducible factor 1alpha (Hif-1alpha) play a fundamental role in bone by stimulating the differentiation of the osteoblasts in hypoxic niches where metastatic cells coming from distant tumors are often found. However, the role of the hypoxic microenvironment in bone metastasis is unknown. We used a conditional knockout approach to suppress either Hif-1alpha or von Hippel Lindau (VHL, an E3 ubiquitin-ligase that targets Hif-1alpha for degradation) specifically in osteoblasts (ΔHif-1alphaOB and ΔVHLOB mice). VHL suppression leads to ectopic expression of Hif-1alpha, and thus functions as a gain-of-function model for Hif-1alpha. Tissue specific ablation of Hif-1alpha and VHL in osteoblasts was performed by mating floxed Hif-1alpha or VHL mice, with mice expressing the Cre recombinase under the control of the osterix promoter, which is solely activated in immature osteoblasts. ΔHif-1alphaOB mice presented a decreased number of osteoblasts and reduced bone mass. Conversely, ΔVHLOB mice exhibited a dramatic increase in osteoblast number and bone mass. Intracardiac injection of PyMT-FVB-Luc+ cells into these loss- or gain-of-function models revealed that osteoblasts directly or indirectly stimulate breast cancer metastasis to bone. Strikingly, our results also show that osteoblasts produce systemic changes, controlling breast cancer metastasis to the lungs, as well as the growth of primary mammary tumors. These results were obtained by either transplanting PyMT-FVB-Luc+ cells into the mammary gland, or by tail vein (intravenous) injections. Thus, osteoblasts can affect distant tissues, well beyond the bone microenvironment. By providing the first evidence that the skeleton exerts a systemic control of breast cancer growth and dissemination, our study expands the biological importance of this organ. Citation Format: Sylvain Provot, Audrey Brenot, Amy-Jo Casbon, Ying Yu, Zena Werb. Microenvironmental control of bone metastasis. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Invasion and Metastasis; Jan 20-23, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;73(3 Suppl):Abstract nr IA2.
- Published
- 2013
- Full Text
- View/download PDF
27. NADPH oxidase flavocytochrome b localizes to Rab11‐positive recycling endosomes in macrophages
- Author
-
Lee-Ann H. Allen, Amy Jo Casbon, Kenneth W. Dunn, and Mary C. Dinauer
- Subjects
NADPH oxidase ,biology ,Biochemistry ,Endosome ,Chemistry ,Genetics ,biology.protein ,Molecular Biology ,Flavocytochrome b ,Biotechnology - Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.