289 results on '"Dragan Maric"'
Search Results
102. NMDAR-mediated transcriptional control of gene expression during the development of medial ganglionic eminence-derived interneurons
- Author
-
Ramesh Chittajallu, Chris J. McBain, Dragan Maric, Vivek Mahadevan, Kenneth A. Pelkey, Ryan K. Dale, Xiaoqing Yuan, Apratim Mitra, Yajun Zhang, Christopher T. Rhodes, Timothy J. Petros, Caroline Esnault, and Areg Peltekian
- Subjects
biology ,Ganglionic eminence ,Interneuron ,Glutamate receptor ,GRIN1 ,Hippocampal formation ,medicine.anatomical_structure ,nervous system ,Gene expression ,biology.protein ,Transcriptional regulation ,medicine ,Neuroscience ,Parvalbumin - Abstract
Medial ganglionic eminence (MGE)-derived parvalbumin (PV)+, somatostatin (SST)+ and Neurogliaform (NGFC)-type cortical and hippocampal interneurons, have distinct molecular, anatomical and physiological properties. However, the molecular mechanisms regulating their diversity remain poorly understood. Here, via single-cell transcriptomics, we show that the obligate NMDA-type glutamate receptor (NMDAR) subunit geneGrin1mediates subtype-specific transcriptional regulation of gene expression in MGE-derived interneurons, leading to altered subtype abundances. Notably, MGE-specific conditionalGrin1loss results in a systemic downregulation of diverse transcriptional, synaptogenic and membrane excitability regulatory programs. These widespread gene expression abnormalities mirror aberrations that are typically associated with neurodevelopmental disorders, particularly schizophrenia. Our study hence provides a road map for the systematic examination of NMDAR signaling in interneuron subtypes, revealing potential MGE-specific genetic targets that could instruct future therapies of psychiatric disorders.
- Published
- 2020
- Full Text
- View/download PDF
103. MerTK inhibition decreases immune suppressive glioblastoma-associated macrophages and neoangiogenesis in glioblastoma microenvironment
- Author
-
Stephen V. Frye, Lee Hwang, Christopher Ryan Miller, Henry Shelton Earp, Ryan E Bash, Masaki Terabe, Yu-Ting Su, Dragan Maric, Hua Song, Madison Butler, Wei Zhang, Mark R. Gilbert, Guangyang Yu, Allison N. Schorzman, Andy D Tran, Jing Wu, Ying Pang, William C. Zamboni, Xiaodong Wang, and Meili Zhang
- Subjects
0301 basic medicine ,Tube formation ,Tumor microenvironment ,Angiogenesis ,Chemistry ,medicine.medical_treatment ,C-Mer Tyrosine Kinase ,MERTK ,medicine.disease ,03 medical and health sciences ,MERTK Gene ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,030220 oncology & carcinogenesis ,Glioma ,Basic and Translational Investigations ,medicine ,Cancer research - Abstract
Background Glioblastoma-associated macrophages and microglia (GAMs) are the predominant immune cells in the tumor microenvironment. Activation of MerTK, a receptor tyrosine kinase, polarizes GAMs to an immunosuppressive phenotype, promoting tumor growth. Here, the role of MerTK inhibition in the glioblastoma microenvironment is investigated in vitro and in vivo. Methods Effects of MRX-2843 in glioblastoma microenvironment regulation were determined in vitro by cell viability, cytokine array, in vitro tube formation, Western blotting, and wound healing assays. A syngeneic GL261 orthotopic glioblastoma mouse model was used to evaluate the survival benefit of MRX-2843 treatment. Multiplex fluorescent immunohistochemistry was used to evaluate the expression of CD206, an anti-inflammatory marker on GAMs, and angiogenesis in murine brain tumor tissues. Results MRX-2843 inhibited cell growth and induced apoptosis in human glioblastoma cells and decreased protein expression of phosphorylated MerTK, AKT, and ERK, which are essential for cell survival signaling. Interleukin-8 and C-C motif chemokine ligand 2, the pro-glioma and pro-angiogenic cytokines, were decreased by MRX-2843. Decreased vascular formation and numbers of immunosuppressive (CD206+) GAMs were observed following MRX-2843 treatment in vivo, suggesting that in addition to alleviating immunosuppression, MRX-2843 also inhibits neoangiogenesis in the glioma microenvironment. These results were supported by a prolonged survival in the syngeneic mouse orthotopic GL261 glioblastoma model following MRX-2843 treatment. Conclusion Our findings suggest that MRX-2843 has a therapeutic benefit via promoting GAM polarization away from immunosuppressive condition, inhibiting neoangiogenesis in the glioblastoma microenvironment and inducing tumor cell death.
- Published
- 2020
104. VPS13D promotes peroxisome biogenesis
- Author
-
Dragan Maric, Gil Kanfer, William A. Prinz, Hetal V. Shah, Eric H. Baehrecke, Heather Baldwin, Richard J. Youle, Chunxin Wang, Norbert Brüggemann, Allyson L. Anding, Antonio Velayos-Baeza, and Marija Dulovic-Mahlow
- Subjects
Organelles ,Proteins ,Cell Biology ,Peroxisome ,Mitochondrion ,Biology ,medicine.disease ,Biochemistry ,Article ,Cell biology ,Mitochondria ,Systems and Computational Biology ,HEK293 Cells ,Mutation ,Spinocerebellar ataxia ,medicine ,Peroxisomes ,Gene family ,Humans ,Gene ,Biogenesis ,Gene knockout ,Membrane and lipid biology ,Abnormal mitochondrial morphology ,HeLa Cells - Abstract
The VPS13 proteins (VPS13A–D) are thought to mediate lipid transport between organelles and are linked to distinct neurological disorders in humans. Baldwin et al. found that, in addition to known involvement in mitochondrial morphology, VPS13D is essential for peroxisome biogenesis., The VPS13 gene family consists of VPS13A–D in mammals. Although all four genes have been linked to human diseases, their cellular functions are poorly understood, particularly those of VPS13D. We generated and characterized knockouts of each VPS13 gene in HeLa cells. Among the individual knockouts, only VPS13D-KO cells exhibit abnormal mitochondrial morphology. Additionally, VPS13D loss leads to either partial or complete peroxisome loss in several transformed cell lines and in fibroblasts derived from a VPS13D mutation–carrying patient with recessive spinocerebellar ataxia. Our data show that VPS13D regulates peroxisome biogenesis.
- Published
- 2020
105. Pain control through selective chemo-axotomy of centrally projecting TRPV1+ sensory neurons
- Author
-
Jason M. Keller, Stephen J. Raithel, Eric L. Rohrs, Andrew J. Mannes, Robert M. Caudle, Ethan M. Anderson, Dragan Maric, John A. Butman, Matthew R. Sapio, Dorothy Cimino Brown, Danielle M. LaPaglia, John K. Neubert, John D. Heiss, and Michael J. Iadarola
- Subjects
0301 basic medicine ,Agonist ,Sensory Receptor Cells ,medicine.drug_class ,medicine.medical_treatment ,Analgesic ,Resiniferatoxin ,TRPV1 ,TRPV Cation Channels ,03 medical and health sciences ,chemistry.chemical_compound ,Dogs ,0302 clinical medicine ,Dorsal root ganglion ,Ganglia, Spinal ,medicine ,Noxious stimulus ,Animals ,Humans ,Pain Management ,business.industry ,Axotomy ,Cancer Pain ,General Medicine ,Analgesics, Non-Narcotic ,Spinal cord ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,chemistry ,Diterpenes ,business ,Neuroscience ,030217 neurology & neurosurgery ,Research Article - Abstract
Agonists of the vanilloid receptor transient vanilloid potential 1 (TRPV1) are emerging as highly efficacious nonopioid analgesics in preclinical studies. These drugs selectively lesion TRPV1+ primary sensory afferents, which are responsible for the transmission of many noxious stimulus modalities. Resiniferatoxin (RTX) is a very potent and selective TRPV1 agonist and is a promising candidate for treating many types of pain. Recent work establishing intrathecal application of RTX for the treatment of pain resulting from advanced cancer has demonstrated profound analgesia in client-owned dogs with osteosarcoma. The present study uses transcriptomics and histochemistry to examine the molecular mechanism of RTX action in rats, in clinical canine subjects, and in 1 human subject with advanced cancer treated for pain using intrathecal RTX. In all 3 species, we observe a strong analgesic action, yet this was accompanied by limited transcriptional alterations at the level of the dorsal root ganglion. Functional and neuroanatomical studies demonstrated that intrathecal RTX largely spares susceptible neuronal perikarya, which remain active peripherally but unable to transmit signals to the spinal cord. The results demonstrate that central chemo-axotomy of the TRPV1+ afferents underlies RTX analgesia and refine the neurobiology underlying effective clinical use of TRPV1 agonists for pain control.
- Published
- 2018
- Full Text
- View/download PDF
106. Protein phosphatase 2A inhibition enhances radiation sensitivity and reduces tumor growth in chordoma
- Author
-
Wei Zhang, Dragan Maric, Amber J. Giles, Mark R. Gilbert, Zhengping Zhuang, Adrian Lita, Tamalee Kramp, Mones Abu-Asab, Martha Quezado, Shuyu Hao, Jinkyu Jung, Kevin Camphausen, Nicole Colwell, Marsha-Kay Hutchinson, Deric M. Park, Hua Song, Xiaoyu Cao, Ashlee Seldomridge, and Mioara Larion
- Subjects
0301 basic medicine ,Cancer Research ,DNA damage ,DNA repair ,Cellular differentiation ,Apoptosis ,Mice, SCID ,Radiation Tolerance ,Piperazines ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Radiation sensitivity ,Cell Movement ,Mice, Inbred NOD ,Biomarkers, Tumor ,Chordoma ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Protein Phosphatase 2 ,Cell Proliferation ,Cell growth ,Chemistry ,Cell cycle ,Bridged Bicyclo Compounds, Heterocyclic ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Basic and Translational Investigations ,Cancer cell ,Cancer research ,Female ,Neurology (clinical) ,Signal Transduction - Abstract
Background Standard therapy for chordoma consists of surgical resection followed by high-dose irradiation. Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase involved in signal transduction, cell cycle progression, cell differentiation, and DNA repair. LB100 is a small-molecule inhibitor of PP2A designed to sensitize cancer cells to DNA damage from irradiation and chemotherapy. A recently completed phase I trial of LB100 in solid tumors demonstrated its safety. Here, we show the therapeutic potential of LB100 in chordoma. Methods Three patient-derived chordoma cell lines were used: U-CH1, JHC7, and UM-Chor1. Cell proliferation was determined with LB100 alone and in combination with irradiation. Cell cycle progression was assessed by flow cytometry. Quantitative γ-H2AX immunofluorescence and immunoblot evaluated the effect of LB100 on radiation-induced DNA damage. Ultrastructural evidence for nuclear damage was investigated using Raman imaging and transmission electron microscopy. A xenograft model was established to determine potential clinical utility of adding LB100 to irradiation. Results PP2A inhibition in concert with irradiation demonstrated in vitro growth inhibition. The combination of LB100 and radiation also induced accumulation at the G2/M phase of the cell cycle, the stage most sensitive to radiation-induced damage. LB100 enhanced radiation-induced DNA double-strand breaks. Animals implanted with chordoma cells and treated with the combination of LB100 and radiation demonstrated tumor growth delay. Conclusions Combining LB100 and radiation enhanced DNA damage-induced cell death and delayed tumor growth in an animal model of chordoma. PP2A inhibition by LB100 treatment may improve the effectiveness of radiation therapy for chordoma.
- Published
- 2017
- Full Text
- View/download PDF
107. Comment on 'mt-Keima detects PINK1-PRKN mitophagy in vivo with greater sensitivity than mito-QC'
- Author
-
Mario K Shammas, Xiaoping Huang, Yi-Ting Liu, Hannah Calvelli, Dragan Maric, Chunxin Wang, Derek P. Narendra, and Danielle A. Sliter
- Subjects
In vivo ,Autophagy ,Mitophagy ,PINK1 ,Cell Biology ,Computational biology ,Sensitivity (control systems) ,Biology ,Molecular Biology - Abstract
We thank Ganley et al. for their thoughtful comment on our recent report [1]. We agree with Ganley et al. that both mito-Keima and mt-QC detect mitophagy. Our report demonstrates, however, that mit...
- Published
- 2021
- Full Text
- View/download PDF
108. NCMP-07. IDH MUTANT GLIOMAS PROMOTE EPILEPTOGENESIS VIA D-2-HYDROXYGLUTARATE DEPENDENT MTOR HYPER-ACTIVATION
- Author
-
Dragan Maric, Kareem A. Zaghloul, Chungzhang Yang, Alexander Ksendzovsky, Muzna Bachani, Jahandar Jahanipour, Islam Fayed, Armin Mortazavi, Anas Khan, Mioara Larion, and Tyrone Dowdy
- Subjects
Cancer Research ,Oncology ,Chemistry ,Mutant ,Cancer research ,Neurology (clinical) ,D-2-hydroxyglutarate ,Epileptogenesis ,PI3K/AKT/mTOR pathway - Abstract
Uncontrolled seizures in patients with low grade gliomas have a significant impact on quality of life and morbidity, yet the mechanisms through which these tumors cause seizures remain unknown. Albeit there are multiple features that contribute to tumor related epileptogenesis, IDH mutations are determined to be an independent factor, although the pathogenesis remains poorly understood. Here, we hypothesize that the active metabolite D-2-hydroxyglutarate (D-2-HG) produced by the IDH-mutant enzyme leads to metabolic disruptions in surrounding cortical neurons that consequently promote seizures. We use a complementary study of in vitro cortical cultures and electrographically sorted human cortical tissue from patients (n=5) with IDH-mutant gliomas to test this hypothesis. We demonstrate that D-2-HG leads to increased neuronal spiking activity (p< 0.0001) and promotes a distinct metabolic profile in surrounding neurons and upregulation of mTOR signaling (p< 0.0001), which is consistent in human epileptic cortex compared to peritumoral nonepileptic cortex. Furthermore, increases in neuronal activity are induced by mTOR activation and reversed with mTOR inhibition. Together, our data suggest that metabolic disruptions and mTOR signaling upregulation in the surrounding cortex due to D-2-HG may be a driving event for epileptogenesis in patients with IDH-mutant low grade gliomas.
- Published
- 2021
- Full Text
- View/download PDF
109. Development of a systems‐based in situ multiplex biomarker screening approach for the assessment of immunopathology and neural tissue plasticity in male rats after traumatic brain injury
- Author
-
Dragan Maric, John M. Hallenbeck, Brian M. Cox, Joshua D. Bernstock, Greg Bull, Shawn Gouty, and Tanya Bogoslovsky
- Subjects
Male ,0301 basic medicine ,Traumatic brain injury ,Systems biology ,Neuroimaging ,Neuropathology ,Biology ,Article ,Tongue Diseases ,Rats, Sprague-Dawley ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Brain Injuries, Traumatic ,Neuroplasticity ,medicine ,Animals ,Multiplex ,Neuroinflammation ,Cerebral Cortex ,Regeneration (biology) ,Brain ,medicine.disease ,Immunohistochemistry ,Oligodendroglia ,030104 developmental biology ,Astrocytes ,Biomarker (medicine) ,Endothelium, Vascular ,Neuroscience ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Traumatic brain injuries (TBIs) pose a massive burden of disease and continue to be a leading cause of morbidity and mortality throughout the world. A major obstacle in developing effective treatments is the lack of comprehensive understanding of the underlying mechanisms that mediate tissue damage and recovery after TBI. As such, our work aims to highlight the development of a novel experimental platform capable of fully characterizing the underlying pathobiology that unfolds after TBI. This platform encompasses an empirically optimized multiplex immunohistochemistry staining and imaging system customized to screen for a myriad of biomarkers required to comprehensively evaluate the extent of neuroinflammation, neural tissue damage, and repair in response to TBI. Herein, we demonstrate that our multiplex biomarker screening platform is capable of evaluating changes in both the topographical location and functional states of resident and infiltrating cell types that play a role in neuropathology after controlled cortical impact injury to the brain in male Sprague-Dawley rats. Our results demonstrate that our multiplex biomarker screening platform lays the groundwork for the comprehensive characterization of changes that occur within the brain after TBI. Such work may ultimately lead to the understanding of the governing pathobiology of TBI, thereby fostering the development of novel therapeutic interventions tailored to produce optimal tissue protection, repair, and/or regeneration with minimal side effects, and may ultimately find utility in a wide variety of other neurological injuries, diseases, and disorders that share components of TBI pathobiology.
- Published
- 2017
- Full Text
- View/download PDF
110. STEM-03. WAVE1 KNOCKDOWN ENHANCES THE ANTITUMOR EFFICACY IN PRIMARY GLIOBLASTOMA NEUROSPHERES
- Author
-
Shilpa Thammegowda, Abhik Ray-Chaudhury, Arunakumar Gangaplara, Dragan Maric, Sriya Namagiri, Hannah Sur, Sadhana Jackson, John D. Heiss, Yeshavanth Kumar Banasavadi-Siddegowda, Amelie Vezina, Ashis Chowdhury, and Nancy A. Edwards
- Subjects
Primary Glioblastoma ,Cancer Research ,Gene knockdown ,Oncology ,Cancer Stem Cells ,Neurosphere ,Cancer research ,Neurology (clinical) ,Biology - Abstract
INTRODUCTION Despite multi-model therapies that include maximal surgical resection, radiation, chemotherapy, and tumor treating fields, the median survival of Glioblastoma (GBM) patients is around 15 months. WASP-family verprolin homologous protein 1 (WAVE1) is a downstream effector that receives signals from small GTPases to regulate the actin cytoskeleton. WAVE1’s interaction with arp2/3 modulates critical roles, such as cell motility and morphologic changes. Expression of WAVE1 has been implicated in leukemia, ovarian, and prostate cancer. In this study, we tested the role of WAVE1 in GBM tumor biology. METHODS Expression of WAVE1 in normal brain and GBM tumor specimens was assessed by immunohistochemistry (IHC). The relevance of targeting WAVE1 for GBM therapy was evaluated in vitro by western blot, proliferation assay, cell cycle analysis, apoptosis assay, migration assay, and neurosphere formation assay using scrambled and target specific WAVE1-siRNA in patient-derived primary GBM neurospheres (GBMNS). RESULTS IHC data shows that the expression of WAVE1 is higher in GBM tumor samples than in low-grade gliomas and normal brain tissues. WAVE1 expression is upregulated in GBMNS compared to normal human astrocytes. WAVE1 knockdown significantly decreased the proliferation, migration, and self-renewal of GBMNS without affecting its cell cycle progression. Furthermore, WAVE1 depletion did not show any alterations in the apoptotic cell population, indicating that WAVE1 knockdown has a cytostatic effect on GBMNS. CONCLUSION Expression of WAVE1 positively correlates with GBM, and its knockdown imparts antitumor efficacy through cytostasis.
- Published
- 2020
- Full Text
- View/download PDF
111. Overexpression of fibroblast growth factor-21 (FGF-21) protects mesenchymal stem cells against caspase-dependent apoptosis induced by oxidative stress and inflammation
- Author
-
Gabriel R. Linares, Yan Leng, Dragan Maric, and De-Maw Chuang
- Subjects
0301 basic medicine ,Caspase 3 ,Apoptosis ,Biology ,Fibroblast growth factor ,Neuroprotection ,Caspase-Dependent Apoptosis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Staurosporine ,Animals ,Cells, Cultured ,Inflammation ,Tumor Necrosis Factor-alpha ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,General Medicine ,Hydrogen Peroxide ,Transplantation ,Fibroblast Growth Factors ,Mice, Inbred C57BL ,Oxidative Stress ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Stem cell ,medicine.drug - Abstract
The clinical application of stem cells offers great promise as a potential avenue for therapeutic use in neurodegenerative diseases. However, cell loss after transplantation remains a major challenge which currently plagues the field. Based on our previous findings that fibroblast growth factor 21 (FGF-21) protected neurons from glutamate excitotoxicity and that upregulation of FGF-21 in a rat model of ischemic stroke was associated with neuroprotection, we proposed that overexpression of FGF-21 protects bone marrow-derived mesenchymal stem cells (MSCs) from apoptosis. To test this hypothesis, we examined whether the detrimental effects of apoptosis can be mitigated by the transgenic overexpression of FGF-21 in MSCs. FGF-21 was transduced into MSCs by lentivirus and its overexpression was confirmed by quantitative PCR. Moreover, FGF-21 overexpression did not stimulate the expression of other FGF family members, suggesting it does not activate a positive feedback system. The effects of hydrogen peroxide (H2 O2 ), tumor necrosis factor-α (TNF-α), and staurosporine, known inducers of apoptosis, were evaluated in FGF-21 overexpressing MSCs and mCherry control MSCs. Caspase 3 and 7 activity was markedly and dose-dependently increased by all three stimuli in mCherry MSCs. FGF-21 overexpression robustly suppressed caspase activation induced by H2 O2 and TNF-α, but not staurosporine. Moreover, the assessment of apoptotic morphological changes confirmed the protective effects of FGF-21 overexpression. Taken together, these results provide compelling evidence that FGF-21 plays a crucial role in protecting MSCs from apoptosis induced by oxidative stress and inflammation, and merits further investigation as a strategy for enhancing the therapeutic efficacy of stem cell-based therapies. This article is protected by copyright. All rights reserved.
- Published
- 2019
112. TMOD-32. GL261 LUCIFERASE-EXPRESSING CELLS ELICIT AN ANTI-TUMOR IMMUNE RESPONSE: AN EVALUATION OF MURINE GLIOMA MODELS
- Author
-
Dragan Maric, Anthony Nwankwo, Nicholas Adamstein, Jeeva Munasinghe, Nancy A. Edwards, Xiang Wang, Victoria Sanchez, Pradeep K. Dagur, Arnold Obungu, Stuart Walbridge, John D. Heiss, Tianxia Wu, Gifty Dominah, John Lynes, and Edjah K. Nduom
- Subjects
Antitumor activity ,Cancer Research ,Immune system ,Oncology ,Chemistry ,Glioma ,Tumor Models ,medicine ,Cancer research ,Luciferase ,Neurology (clinical) ,medicine.disease - Abstract
Preclinical models that reliably recapitulate the immunosuppressive properties of human gliomas are essential to assess immune-based therapies. Intracranially injected GL261 cells are widely used as an immunocompetent animal model of glioma, but it is common practice to transfect these with luciferase to facilitate tumor monitoring during treatment. Our group has previously shown that the luciferase-expressing GL261 Red-FLuc cells create an inflammatory response when implanted intracranially. Now, we additionally explore the inflammatory response of GL261-Luc2 cells and demonstrate a similar host immune response occurs with this model as well. In our in vivo evaluation, C57BL/6 mice underwent stereotaxic, intracranial implantation with GL261, GL261 Red-FLuc or GL261-Luc2 cells at doses of 5x104cells/5mL or 3x105cells/5uL.MRIs were performed to monitor relative tumor growth. To assess intrinsic differences between cell lines, in vitro cytokine profiles were evaluated by proteome microarray. Kaplan-Meier survival analyses demonstrated median survival for mice implanted with GL261 cells at 5x104cells was 18 to 21 days. The GL261-Red FLuc implanted mice cells did not reach median survival at either tumor dose with greater than 60% of mice termed long-term survivors. Finally, mice injected with GL261-Luc2 cells at 3x105cells reached median survival at 23 days, but median survival was significantly prolonged for mice implanted with GL261-Luc2 at a dose of 5x104cells (37 days, with 40% becoming long-term survivors) compared to GL261 implanted mice. MRIs reveal differences in tumor growth that correspond with the differences in median survival between groups. In addition, proteomic analyses revealed significantly elevated inflammatory cytokines such as IFN-gamma, IL-7 and TNF-alpha in the supernatants of the GL261 Red-FLuc cells and GL261-Luc2 cells. Further immune characterization is ongoing. Our data suggests that GL261 Red-FLuc and GL261-Luc2 murine models elicit an anti-tumor immune response by increasing pro-inflammatory modulators which stimulate the tumoricidal function of immune cells in the tumor microenvironment.
- Published
- 2019
113. A novel in situ multiplex immunofluorescence panel for the assessment of tumor immunopathology and response to virotherapy in pediatric glioblastoma reveals a role for checkpoint protein inhibition
- Author
-
Li Rong, James M. Markert, G. Yancey Gillespie, Gregory K. Friedman, Nunzio Vicario, Richard J. Whitley, Joshua D. Bernstock, Asim K. Bag, T. Prescott Atkinson, Jason A. Chen, Diana S Osorio, Daniel DiToro, Florian Gessler, Kara Kachurak, James M. Johnston, Bryan D. Choi, Pablo A. Valdés, and Dragan Maric
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,checkpoint proteins ,medicine.medical_treatment ,Immunology ,multiplex immunofluorescence ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Downregulation and upregulation ,Glioma ,Immunopathology ,glioma ,medicine ,Immunology and Allergy ,ddc:610 ,Virotherapy ,oncolytic virotherapy ,business.industry ,Brief Report ,HSV ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,3. Good health ,Oncolytic virus ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,lcsh:RC581-607 ,business ,CD8 - Abstract
Immunotherapy with oncolytic herpes simplex virus-1 therapy offers an innovative, targeted, less-toxic approach for treating brain tumors. However, a major obstacle in maximizing oncolytic virotherapy is a lack of comprehensive understanding of the underlying mechanisms that unfold in CNS tumors/associated microenvironments after infusion of virus. We demonstrate that our multiplex biomarker screening platform comprehensively informs changes in both topographical location and functional states of resident/infiltrating immune cells that play a role in neuropathology after treatment with HSV G207 in a pediatric Phase 1 patient. Using this approach, we identified robust infiltration of CD8+ T cells suggesting activation of the immune response following virotherapy; however there was a corresponding upregulation of checkpoint proteins PD-1, PD-L1, CTLA-4, and IDO revealing a potential role for checkpoint inhibitors. Such work may ultimately lead to an understanding of the governing pathobiology of tumors, thereby fostering development of novel therapeutics tailored to produce optimal responses.
- Published
- 2019
114. CD8+ T cells target cerebrovasculature in children with cerebral malaria
- Author
-
Susan K. Pierce, Karl B. Seydel, Terrie E. Taylor, Dorian B. McGavern, Monica Manglani, Louis H. Miller, Kory R. Johnson, Osorio Abath Neto, Myoung-Hwa Lee, Avindra Nath, Brittany A. Riggle, and Dragan Maric
- Subjects
0301 basic medicine ,Male ,CD3 ,T cell ,Malaria, Cerebral ,CD8-Positive T-Lymphocytes ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,parasitic diseases ,Medicine ,Cytotoxic T cell ,Humans ,Child ,biology ,business.industry ,Brain ,Infant ,General Medicine ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Cerebral Malaria ,030220 oncology & carcinogenesis ,Child, Preschool ,Immunology ,biology.protein ,Commentary ,Female ,business ,CD8 ,Malaria - Abstract
BACKGROUNDCerebral malaria (CM) accounts for nearly 400,000 deaths annually in African children. Current dogma suggests that CM results from infected RBC (iRBC) sequestration in the brain microvasculature and resulting sequelae. Therapies targeting these events have been unsuccessful; findings in experimental models suggest that CD8+ T cells drive disease pathogenesis. However, these data have largely been ignored because corroborating evidence in humans is lacking. This work fills a critical gap in our understanding of CM pathogenesis that is impeding development of therapeutics.METHODSUsing multiplex immunohistochemistry, we characterized cerebrovascular immune cells in brain sections from 34 children who died from CM or other causes. Children were grouped by clinical diagnosis (CM+ or CM-), iRBC sequestration (Seqhi, Seqlo, Seq0) and HIV status (HIV+ or HIV-).RESULTSWe identified effector CD3+CD8+ T cells engaged on the cerebrovasculature in 69% of CM+ HIV- children. The number of intravascular CD3+CD8+ T cells was influenced by CM status (CM+ > CM-, P = 0.004) and sequestration level (Seqhi > Seqlo, P = 0.010). HIV coinfection significantly increased T cell numbers (P = 0.017) and shifted cells from an intravascular (P = 0.004) to perivascular (P < 0.0001) distribution.CONCLUSIONWithin the studied cohort, CM is associated with cerebrovascular engagement of CD3+CD8+ T cells, which is exacerbated by HIV coinfection. Thus, CD3+CD8+ T cells are highly promising targets for CM adjunctive therapy, opening new avenues for the treatment of this deadly disease.FUNDINGThis research was supported by the Intramural Research Program of the National Institutes of Health.
- Published
- 2019
115. Nitrosative Stress Is Associated with Dopaminergic Dysfunction in the HIV-1 Transgenic Rat
- Author
-
Dragan Maric, Ashutosh Kumar, Swati Shah, Frank Denaro, Dima A. Hammoud, Wael G. Ibrahim, William Reid, and Ronald P. Mason
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Transgene ,Dopamine ,Neurocognitive Disorders ,HIV Infections ,medicine.disease_cause ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Humans ,Dopamine transporter ,Dopamine Plasma Membrane Transport Proteins ,biology ,business.industry ,Dopaminergic ,Colocalization ,Glutathione ,Rats ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,chemistry ,biology.protein ,HIV-1 ,Tyrosine ,Thioredoxin ,Rats, Transgenic ,business ,030217 neurology & neurosurgery ,Oxidative stress ,Immunostaining - Abstract
Advances in antiretroviral therapy have resulted in significantly decreased HIV-related mortality. HIV-associated neurocognitive disorders, however, continue to be a major problem in infected patients. The neuropathology underlying HIV-associated neurocognitive disorders has not been well characterized, and evidence suggests different contributing mechanisms. One potential mechanism is the induction of oxidative stress. Using the HIV-1 transgenic (Tg) rat model of HIV, we found increased striatal NADPH oxidase-4 and neuronal nitric oxide synthase expression in the adult (7- to 9-month-old) Tg rat compared with control rats but not in the young (1-month-old) Tg rats. This was accompanied by increased 3-nitrotyrosine (3-NT) immunostaining in the adult Tg rats, which worsened significantly in the old Tg rats (18 to 20 months old). There was, however, no concurrent induction of the antioxidant systems because there was no change in the expression of the nuclear factor-erythroid 2-related factor 2 and its downstream targets (thioredoxin and glutathione antioxidant systems). Colocalization of 3-NT staining with neurofilament proteins and evidence of decreased tyrosine hydroxylase and dopamine transporter expression in the old rats support dopaminergic involvement. We conclude that the HIV-1 Tg rat brain shows evidence of nitrosative stress without appropriate oxidation-reduction adaptation, whereas 3-NT modification of striatal neurofilament proteins likely points to the ensuing dopaminergic neuronal loss and dysfunction in the aging HIV-1 Tg rat.
- Published
- 2019
116. Neuroinflammatory Changes in Relation to Cerebrospinal Fluid Viral Load in Simian Immunodeficiency Virus Encephalitis
- Author
-
Paul Wakim, Avindra Nath, Sanhita Sinharay, William Reid, Vanessa M. Hirsch, William Schreiber-Stainthorp, Swati Shah, Dragan Maric, Siva Muthusamy, Falguni Basuli, Dianne E. Lee, Cheri A. Lee, Dima A. Hammoud, Michele Di Mascio, and Kenta Matsuda
- Subjects
Male ,Pathology ,medicine.medical_specialty ,positron emission tomography ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,medicine.disease_cause ,Microbiology ,Host-Microbe Biology ,Proinflammatory cytokine ,neuroinflammation ,03 medical and health sciences ,rhesus macaques ,0302 clinical medicine ,Cerebrospinal fluid ,Virology ,Translocator protein ,Animals ,Medicine ,Neuroinflammation ,030304 developmental biology ,Inflammation ,0303 health sciences ,biology ,Microglia ,business.industry ,translocator protein ,SIV encephalitis ,Brain ,DPA714 ,Viral Load ,Simian immunodeficiency virus ,Macaca mulatta ,QR1-502 ,Disease Models, Animal ,medicine.anatomical_structure ,Positron-Emission Tomography ,biology.protein ,Female ,Simian Immunodeficiency Virus ,business ,Neuroglia ,Viral load ,030217 neurology & neurosurgery ,Immunostaining ,Research Article - Abstract
Neurological and cognitive problems are a common complication of HIV infection and are prevalent even in treated individuals. Although the molecular processes underlying brain involvement with HIV are not completely understood, inflammation is suspected to play a significant role. Our work presents an in vivo assessment of neuroinflammation in an animal model of HIV, the simian immunodeficiency virus (SIV)-infected rhesus macaque. Using positron emission tomography (PET) imaging, we identified changes in brain inflammation after inoculation with SIV over time. Interestingly, we found decreased binding of the PET ligand in the presence of very high cerebrospinal fluid (CSF) viral loads. These findings were supported by immunostaining which showed marked glial loss instead of inflammation. This study provides insight into glial and neuronal changes associated with very high CSF viral load and could reflect similar changes occurring in HIV-infected patients., The exact cause of neurocognitive dysfunction in HIV-positive patients despite successful control of the infection in the periphery is not completely understood. One suggested mechanism is a vicious cycle of microglial activation and release of proinflammatory chemokines/cytokines that eventually leads to neuronal loss and dysfunction. However, the exact role of microglial activation in the earliest stages of the infection with high cerebrospinal fluid (CSF) viral loads (VL) is unclear. In this study, we imaged the translocator protein (TSPO), a mitochondrial membrane receptor known to be upregulated in activated microglia and macrophages, in rhesus macaques before and multiple times after inoculation with a neurotropic simian immunodeficiency virus (SIV) strain (SIVsm804E), using 18F-DPA714 positron emission tomography (PET). The whole-brain standardized uptake values of TSPO at equilibrium reflecting total binding (SUVT) and binding potentials (BPND) were calculated and correlated with CSF and serum markers of disease, and a corresponding postmortem immunostaining analysis was also performed. SUVT was found to be inversely correlated with both CSF VL and monocyte chemoattractant protein 1 (MCP-1) levels. In SIV-infected macaques with very high CSF VL at necropsy (>106 copies/ml), we found decreased TSPO binding by PET, and this was supported by immunostaining which showed glial and neuronal apoptosis rather than microglial activation. On the other hand, with only moderately elevated CSF VL (∼104 copies/ml), we found increased TSPO binding as well as focal and diffuse microglial activation on immunostaining. Our results in the SIV-infected macaque model provide insights into the relationship between HIV neuropathology and CSF VL at various stages of the disease.
- Published
- 2019
117. CRH stimulation improves
- Author
-
Jacqueline, Boyle, Nicholas J, Patronas, James, Smirniotopoulos, Peter, Herscovitch, William, Dieckman, Corina, Millo, Dragan, Maric, Grégoire P, Chatain, Christina Piper, Hayes, Sarah, Benzo, Gretchen, Scott, Nancy, Edwards, Abhik, Ray Chaudhury, Maya B, Lodish, Susmeeta, Sharma, Lynnette K, Nieman, Constantine A, Stratakis, Russell R, Lonser, and Prashant, Chittiboina
- Subjects
Adenoma ,Adult ,Male ,Adolescent ,Corticotropin-Releasing Hormone ,Middle Aged ,Sensitivity and Specificity ,Diagnosis, Differential ,Young Adult ,ACTH-Secreting Pituitary Adenoma ,Fluorodeoxyglucose F18 ,Positron-Emission Tomography ,Humans ,Female ,Child ,Pituitary ACTH Hypersecretion - Abstract
In MRI-negative cases Cushing's disease (CD), surgeons perform a more extensive exploration of the pituitary gland, with fewer instances of hormonal remission.Subjects with a likely diagnosis of CD (n = 27, 20 females) each underwent twoThe mean-SUV of adenomas was significantly increased from baseline (3.6 ± 1.5) with oCRH administration (3.9 ± 1.7; one-tailed p = 0.003). Neuroradiologists agreed that adenomas were visible on 21 scans, not visible on 26 scans (disagreed about 7, kappa = 0.7). oCRH-stimulation led to the detection of additional adenomas (n = 6) not visible on baseline-PET study. Of the MRI-negative adenomas (n = 5), two were detected on PET imaging (one only after oCRH-stimulation). USP8 mutations or glycolytic pathway proteins were not associated with SUV in corticotropinomas.The results of the current study suggest that oCRH-stimulation may lead to increased
- Published
- 2019
118. Abstract WP550: Pathophysiology of Acute, Subacute and Chronic Lesions in Stroke: A Radiological-Pathological Analysis
- Author
-
Abhik Ray-Chaudhury, John Lynch, Nancy J. Edwards, Jill B. De Vis, Allison Griffin, Lawrence L. Latour, Richard Leigh, and Dragan Maric
- Subjects
Advanced and Specialized Nursing ,Pathology ,medicine.medical_specialty ,business.industry ,medicine.disease ,Pathophysiology ,Lesion ,Pathogenesis ,Radiological weapon ,medicine ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Stroke ,Pathological ,Diffusion MRI - Abstract
Introduction: The imaging characteristics of ischemic lesions seen on MRI aid in the interpretation of the lesion pathogenesis. Early ischemic lesions are seen on diffusion weighted imaging (DWI) with low apparent diffusion coefficient (ADC), later evolve on T2-FLAIR, develop elevated ADC, then disappear on DWI. Chronic white matter lesions (WML) seen on T2-FLAIR but not DWI may or may not be of ischemic origin. Here we present an MRI targeted pathology approach to better describe in-vivo MRI based on cellular changes. Methods: The brain of a patient was obtained for pathological evaluation early after stroke recurrence. As part of clinical care and study procedures, 3T MRI was obtained at baseline (within 2-hours), multiple follow-up visits, and during second admission. Radiological-pathological methods previously validated in trauma was employed. Whole brain 7T MRI was performed after formalin fixation, and MRI targeted pathology was performed, co-localizing three lesions of different ages based on history and presumed imaging characteristics; i) acute, ii) subacute, and iii) chronic vs WML. Homologous contralateral tissue was used as control. Results: Lesions seen in-vivo were observed in post mortem MRI, and reconfirmed in histology. An example of in-vivo and post-mortem MRI, along with corresponding tissue sections and H&E are shown in figure. Qualitative delineation of the boundaries of the chronic lesion on H&E was challenging. A large number of small vessels with enlarged perivascular spaces dominated. The subacute lesion, was characterized by loss of white mater, infiltration of macrophages, and astrocytosis. The acute lesion had a text-book appearance. Conclusion: Dramatic differences are seen in the volume fraction of free water, which likely a major contributor to the appearance on DWI/ADC and T2 FLAIR. Work continues with immunohistochemistry to better delineate the boundaries of the lesions and cellular morphology relevant to in-vivo MRI.
- Published
- 2019
- Full Text
- View/download PDF
119. Dynorphin and Enkephalin Opioid Peptides and Transcripts in Spinal Cord and Dorsal Root Ganglion During Peripheral Inflammatory Hyperalgesia and Allodynia
- Author
-
Michael J. Iadarola, Andrew J. Mannes, Dragan Maric, Jenny Kim, Jean Thierry-Mieg, Amelia J. Loydpierson, Danielle Thierry-Mieg, and Matthew R. Sapio
- Subjects
Male ,medicine.medical_specialty ,Enkephalin ,Dynorphin ,Dynorphins ,Article ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Dorsal root ganglion ,Internal medicine ,Ganglia, Spinal ,Medicine ,Animals ,RNA, Messenger ,Opioid peptide ,Endogenous opioid ,business.industry ,Dynorphin A ,Enkephalins ,Peptide Fragments ,Proenkephalin ,Rats ,Anesthesiology and Pain Medicine ,Endocrinology ,Allodynia ,medicine.anatomical_structure ,Neurology ,chemistry ,nervous system ,Opioid Peptides ,Spinal Cord ,Hyperalgesia ,Neurology (clinical) ,medicine.symptom ,Inflammation Mediators ,business - Abstract
Understanding molecular alterations associated with peripheral inflammation is a critical factor in selectively controlling acute and persistent pain. The present report employs in situ hybridization of the 2 opioid precursor mRNAs coupled with quantitative measurements of 2 peptides derived from the prodynorphin and proenkephalin precursor proteins: dynorphin A 1-8 and [Met5]-enkephalin-Arg6-Gly7-Leu8. In dorsal spinal cord ipsilateral to the inflammation, dynorphin A 1-8 was elevated after inflammation, and persisted as long as the inflammation was sustained. Qualitative identification by high performance liquid chromatography and gel permeation chromatography revealed the major immunoreactive species in control and inflamed extracts to be dynorphin A 1-8. In situ hybridization in spinal cord after administration of the inflammatory agent, carrageenan, showed increased expression of prodynorphin (Pdyn) mRNA somatotopically in medial superficial dorsal horn neurons. The fold increase in preproenkephalin mRNA (Penk) was comparatively lower, although the basal expression is substantially higher than Pdyn. While Pdyn is not expressed in the dorsal root ganglion (DRG) in basal conditions, it can be induced by nerve injury, but not by inflammation alone. A bioinformatic meta-analysis of multiple nerve injury datasets confirmed Pdyn upregulation in DRG across different nerve injury models. These data support the idea that activation of endogenous opioids, notably dynorphin, is a dynamic indicator of persistent pain states in spinal cord and of nerve injury in DRG. PERSPECTIVE: This is a systematic, quantitative assessment of dynorphin and enkephalin peptides and mRNA in dorsal spinal cord and DRG neurons in response to peripheral inflammation and axotomy. These studies form the foundational framework for understanding how endogenous spinal opioid peptides are involved in nociceptive circuit modulation.
- Published
- 2018
120. SUMOylation promotes survival and integration of neural stem cell grafts in ischemic stroke
- Author
-
Luca Peruzzotti-Jametti, Kory R. Johnson, Mark Winterbone, Yongshan Mou, Yang-ja Lee, Aletta Van Den Bosch, Joshua D. Bernstock, Gregory K. Friedman, Dragan Maric, Nunzio Vicario, Robin J.M. Franklin, John M. Hallenbeck, Tommaso Leonardi, Stefano Pluchino, Daniel Ye, Bernstock, Joshua [0000-0002-7814-3867], Peruzzotti Jametti, Luca [0000-0002-9396-5607], Leonardi, Tommaso [0000-0002-4449-1863], Franklin, Robin [0000-0001-6522-2104], Pluchino, Stefano [0000-0002-6267-9472], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Male ,Research paper ,SUMO protein ,Gene Expression ,Regenerative medicine ,Transgenic ,Cell therapy ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Neural stem cells (NSCs) ,Ubc9 ,Stroke ,Neurons ,Cell Cycle ,Cellular engineering ,Ischemia/reperfusion ,SUMOylation ,Animals ,Biomarkers ,Computational Biology ,Energy Metabolism ,Gene Expression Profiling ,Glucose ,Mice, Transgenic ,Neurogenesis ,Oxygen ,Signal Transduction ,Stem Cell Transplantation ,Sumoylation ,Ubiquitin-Conjugating Enzymes ,Cell Survival ,General Medicine ,Neural stem cell ,030220 oncology & carcinogenesis ,Stem cell ,Ischemia ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Downregulation and upregulation ,medicine ,business.industry ,medicine.disease ,030104 developmental biology ,nervous system ,business ,Neuroscience - Abstract
Background Neural stem cell (NSC)-based therapies hold great promise for treating diseases of the central nervous system (CNS). However, several fundamental problems still need to be overcome to fully exploit the clinical potential of NSC therapeutics. Chief among them is the limited survival of NSC grafts within hostile microenvironments. Methods Herein, we sought to engineer NSCs in an effort to increase graft survival within ischemic brain lesions via upregulation of global SUMOylation, a post-translational modification critically involved in mediating tolerance to ischemia/reperfusion. Findings NSCs overexpressing the SUMO E2-conjugase Ubc9 displayed resistance to oxygen-glucose-deprivation/restoration of oxygen/glucose (OGD/ROG) and enhanced neuronal differentiation in vitro, as well as increased survival and neuronal differentiation when transplanted in mice with transient middle cerebral artery occlusion in vivo. Interpretation Our work highlights a critical role for SUMOylation in NSC biology and identifies a biological pathway that can be targeted to increase the effectiveness of exogenous stem cell medicines in ischemic stroke. Fund Intramural Research Program of the NINDS/NIH, the Italian Multiple Sclerosis Foundation (FISM), the Bascule Charitable Trust, NIH-IRTA-OxCam and Wellcome Trust Research Training Fellowships.
- Published
- 2018
121. MRI demonstrates glutamine antagonist-mediated reversal of cerebral malaria pathology in mice
- Author
-
Susan K. Pierce, Louis H. Miller, Jeeva Munasinghe, Sanhita Sinharay, William Schreiber-Stainthorp, Eva Prchalova, Jonathan D. Powell, Dragan Maric, Dima A. Hammoud, Barbara S. Slusher, and Brittany A. Riggle
- Subjects
Adult ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Glutamine ,Diazooxonorleucine ,Plasmodium falciparum ,Malaria, Cerebral ,Brain Edema ,Antimalarials ,Mice ,03 medical and health sciences ,In vivo ,Edema ,medicine ,Animals ,Humans ,Malaria, Falciparum ,Child ,Multidisciplinary ,biology ,business.industry ,Antagonist ,Brain ,biology.organism_classification ,Magnetic Resonance Imaging ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,PNAS Plus ,Blood-Brain Barrier ,Cerebral Malaria ,Adjunctive treatment ,Disease Progression ,Female ,medicine.symptom ,Complication ,business ,Biomarkers - Abstract
The deadliest complication of Plasmodium falciparum infection is cerebral malaria (CM), with a case fatality rate of 15 to 25% in African children despite effective antimalarial chemotherapy. No adjunctive treatments are yet available for this devastating disease. We previously reported that the glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON) rescued mice from experimental CM (ECM) when administered late in the infection, a time by which mice had already suffered blood–brain barrier (BBB) dysfunction, brain swelling, and hemorrhaging. Herein, we used longitudinal MR imaging to visualize brain pathology in ECM and the impact of a new DON prodrug, JHU-083, on disease progression in mice. We demonstrate in vivo the reversal of disease markers in symptomatic, infected mice following treatment, including the resolution of edema and BBB disruption, findings usually associated with a fatal outcome in children and adults with CM. Our results support the premise that JHU-083 is a potential adjunctive treatment that could rescue children and adults from fatal CM.
- Published
- 2018
- Full Text
- View/download PDF
122. DDRE-27. IDH MUTATED GLIOMAS PROMOTE EPILEPTOGENESIS VIA D-2-HYDROXYGLUTARATE DEPENDENT MTOR HYPERACTIVATION
- Author
-
Muzna Bachani, Joseph P. Steiner, Dragan Maric, Chunzhang Yang, Islam Fayed, Mioara Larion, Armin Mortazavi, Alexander Ksendzovsky, Tyrone Dowdy, and Kareem A. Zaghloul
- Subjects
Mutation ,Hyperactivation ,Chemistry ,Metabolic Drug Targets, Resistance ,medicine.disease ,medicine.disease_cause ,Epileptogenesis ,Supplement Abstracts ,Isocitrate dehydrogenase ,Glioma ,medicine ,Cancer research ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,D-2-hydroxyglutarate ,Signal transduction ,PI3K/AKT/mTOR pathway - Abstract
INTRODUCTION Epileptic seizures in patients with low-grade, isocitrate dehydrogenase (IDH) mutated gliomas reach 90%, a major source of morbidity for these patients. Albeit there are multiple features that contribute to tumor related epileptogenesis, IDH mutations are determined to be an independent factor, although the pathogenesis remains poorly understood. We demonstrate IDH-mutated tumors promote epileptogenesis through D-2-hydroxyglutarate (D-2-HG) dependent mTOR hyperactivation and metabolic reprogramming. METHODS Human epileptic and nonepileptic cortex were identified via subdural electrodes in patients with IDH-mutated gliomas (n=5). An in vitro rat cortical neuronal model on microelectrode arrays were utilized to investigate the role of D-2-HG on neuronal excitability. mTOR and lysine demethylase (KDM) modulators were applied to elucidate the epileptogenic mechanism. Tetrodotoxin was utilized to evaluate the contribution of neuronal activity to mTOR signaling and metabolism. mTOR signaling was evaluated through western blot analysis and multiplex immunofluorescence. Metabolic function were analyzed via Seahorse assays and metabolomic analysis. RESULTS D-2-HG increased normalized bursting rate in the neuronal cultures (p CONCLUSION We demonstrate IDH-mutated gliomas promote epileptogenesis through a D-2-HG dependent mTOR hyperactivation via KDM inhibition, a putative mechanism and potential therapeutic targets. Furthermore, we argue mTOR hyperactivation results in metabolic reprogramming, independent of neuronal firing, which may contribute to epileptogenesis, a heretofore unrecognized aspect of pathologic mTOR signaling in neurological diseases.
- Published
- 2021
- Full Text
- View/download PDF
123. Preconditioning mesenchymal stem cells with the mood stabilizers lithium and valproic acid enhances therapeutic efficacy in a mouse model of Huntington's disease
- Author
-
Gabriel R. Linares, Yan Leng, Lisa Scheuing, Hsiao-Mei Liao, Chi-Tso Chiu, De-Maw Chuang, and Dragan Maric
- Subjects
Male ,0301 basic medicine ,Dopamine and cAMP-Regulated Phosphoprotein 32 ,Huntingtin ,medicine.medical_treatment ,Gene Expression ,Mice, Transgenic ,Pharmacology ,Mesenchymal Stem Cell Transplantation ,Neuroprotection ,Drug Administration Schedule ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,Huntington's disease ,Antimanic Agents ,Huntingtin Protein ,Animals ,Medicine ,Receptors, Cytokine ,business.industry ,Valproic Acid ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Stem-cell therapy ,medicine.disease ,Up-Regulation ,Mice, Inbred C57BL ,Transplantation ,Disease Models, Animal ,Proto-Oncogene Proteins c-kit ,Huntington Disease ,030104 developmental biology ,Neurology ,Phosphopyruvate Hydratase ,Mutation ,Cytokines ,Female ,Stem cell ,Lithium Chloride ,business ,030217 neurology & neurosurgery - Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder caused by CAG repeat expansions in the huntingtin gene. Although, stem cell-based therapy has emerged as a potential treatment for neurodegenerative diseases, limitations remain, including optimizing delivery to the brain and donor cell loss after transplantation. One strategy to boost cell survival and efficacy is to precondition cells before transplantation. Because the neuroprotective actions of the mood stabilizers lithium and valproic acid (VPA) induce multiple pro-survival signaling pathways, we hypothesized that preconditioning bone marrow-derived mesenchymal stem cells (MSCs) with lithium and VPA prior to intranasal delivery to the brain would enhance their therapeutic efficacy, and thereby facilitate functional recovery in N171-82Q HD transgenic mice. MSCs were treated in the presence or absence of combined lithium and VPA, and were then delivered by brain-targeted single intranasal administration to eight-week old HD mice. Histological analysis confirmed the presence of MSCs in the brain. Open-field test revealed that ambulatory distance and mean velocity were significantly improved in HD mice that received preconditioned MSCs, compared to HD vehicle-control and HD mice transplanted with non-preconditioned MSCs. Greater benefits on motor function were observed in HD mice given preconditioned MSCs, while HD mice treated with non-preconditioned MSCs showed no functional benefits. Moreover, preconditioned MSCs reduced striatal neuronal loss and huntingtin aggregates in HD mice. Gene expression profiling of preconditioned MSCs revealed a robust increase in expression of genes involved in trophic effects, antioxidant, anti-apoptosis, cytokine/chemokine receptor, migration, mitochondrial energy metabolism, and stress response signaling pathways. Consistent with this finding, preconditioned MSCs demonstrated increased survival after transplantation into the brain compared to non-preconditioned cells. Our results suggest that preconditioning stem cells with the mood stabilizers lithium and VPA before transplantation may serve as an effective strategy for enhancing the therapeutic efficacy of stem cell-based therapies.
- Published
- 2016
- Full Text
- View/download PDF
124. Characterization of neuropathology in the HIV-1 transgenic rat at different ages
- Author
-
Dianne E. Lee, Rafael Casas, Dima A. Hammoud, Wael G. Ibrahim, William Reid, Dragan Maric, Frank Denaro, and Saejeong J. Kim
- Subjects
Gene Expression Regulation, Viral ,Male ,0301 basic medicine ,Aging ,Pathology ,medicine.medical_specialty ,Tyrosine 3-Monooxygenase ,Immunology ,Subventricular zone ,Hippocampus ,Enzyme-Linked Immunosorbent Assay ,Neuropathology ,Striatum ,HIV Envelope Protein gp120 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Glial Fibrillary Acidic Protein ,Animals ,Humans ,Immunology and Allergy ,Medicine ,AIDS-Associated Nephropathy ,CD11b Antigen ,Glial fibrillary acidic protein ,biology ,business.industry ,Calcium-Binding Proteins ,Microfilament Proteins ,Neurogenesis ,Dopaminergic ,Neurotoxicity ,Brain ,medicine.disease ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Case-Control Studies ,HIV-1 ,biology.protein ,Neurology (clinical) ,Rats, Transgenic ,business ,030217 neurology & neurosurgery - Abstract
The transgenic HIV-1 rat (Tg) is a commonly used neuroHIV model with documented neurologic/behavioral deficits. Using immunofluorescent staining of the Tg brain, we found astrocytic dysfunction/damage, as well as dopaminergic neuronal loss/dysfunction, both of which worsening significantly in the striatum with age. We saw mild microglial activation in young Tg brains, but this decreased with age. There were no differences in neurogenesis potential suggesting a neurodegenerative rather than a neurodevelopmental process. Gp120 CSF levels exceeded serum gp120 levels in some animals, suggesting local viral protein production in the brain. Further probing of the pathophysiology underlying astrocytic injury in this model is warranted.
- Published
- 2016
- Full Text
- View/download PDF
125. Insufficient disease inhibition by intrathecal rituximab in progressive multiple sclerosis
- Author
-
Yen Chih Lin, Joan Ohayon, Peter Kosa, Andrew Blake, Dragan Maric, Irene Cortese, Tianxia Wu, Jamie Cherup, Mika Komori, and Bibiana Bielekova
- Subjects
0301 basic medicine ,Central nervous system ,Inflammation ,Pharmacology ,Placebo ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,medicine ,Research Articles ,CD20 ,biology ,business.industry ,General Neuroscience ,Interim analysis ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,biology.protein ,Biomarker (medicine) ,Rituximab ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Research Article ,medicine.drug - Abstract
Objective Inaccessibility of the inflammation compartmentalized to the central nervous system (CNS) may underlie the lack of efficacy of immunomodulatory treatments in progressive multiple sclerosis (MS). The double blind combination of Rituximab by IntraVenous and IntraThecAl injection versus placebo in patients with Low-Inflammatory SEcondary progressive MS (RIVITALISE; NCT01212094) trial was designed to answer: (1) Whether an induction dose of intravenous and intrathecal rituximab efficiently depletes CNS B cells? and (2) If so, whether this leads to global inhibition of CNS inflammation and slowing of CNS tissue destruction? Methods Patients aged 18–65 years were randomly assigned to rituximab or placebo. Protocol-stipulated interim analysis quantified the efficacy of B-cell depletion. Results The efficacy on cerebrospinal fluid (CSF) biomarkers failed to reach criteria for continuation of the trial. B-cell-related CSF biomarkers (sCD21 and B-cell activating factor) changed only in the active-treatment arm. While CSF B cells were killed robustly (median −79.71%, P = 0.0176), B cells in CNS tissue were depleted inadequately (~−10–20%, P
- Published
- 2016
- Full Text
- View/download PDF
126. PP2A inhibition with LB100 enhances cisplatin cytotoxicity and overcomes cisplatin resistance in medulloblastoma cells
- Author
-
Abhik Ray-Chaudhury, Juan-Carlos Vera, Dragan Maric, Martin J. Lizak, R. Aaron Robison, Michael J. Feldman, Matthew D. Hall, Winson S. Ho, Zhengping Zhuang, Lauren Amable, John D. Heiss, and Gerald M. Feldman
- Subjects
0301 basic medicine ,cisplatin ,Mice, SCID ,medulloblastoma ,Piperazines ,STAT3 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Animals ,Humans ,Medicine ,Protein Phosphatase 2 ,Viability assay ,Cerebellar Neoplasms ,Cytotoxicity ,Cisplatin ,Medulloblastoma ,business.industry ,LB100 ,Drug Synergism ,Bridged Bicyclo Compounds, Heterocyclic ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,PP2A ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Apoptosis ,Cell culture ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,business ,Research Paper ,medicine.drug - Abstract
The protein phosphatase 2A (PP2A) inhibitor, LB100, has been shown in pre-clinical studies to be an effective chemo- and radio-sensitizer for treatment of various cancers. We investigated effects associated with LB100 treatment alone and in combination with cisplatin for medulloblastoma (MB) in vitro and in vivo in an intracranial xenograft model. We demonstrated that LB100 had a potent effect on MB cells. By itself, LB100 inhibited proliferation and induced significant apoptosis in a range of pediatric MB cell lines. It also attenuated MB cell migration, a pre-requirement for invasion. When used in combination, LB100 enhanced cisplatin-mediated cytotoxic effects. Cell viability in the presence of 1 uM cisplatin alone was 61% (DAOY), 100% (D341), and 58% (D283), but decreased with the addition of 2 μM of LB100 to 26% (DAOY), 67% (D341), and 27% (D283), (p < 0.005). LB100 suppressed phosphorylation of the STAT3 protein and several STAT3 downstream targets. Also, LB100 directly increased cisplatin uptake and overcame cisplatin-resistance in vitro. Finally, LB100 exhibited potent in vivo anti-neoplastic activity in combination with cisplatin in an intracranial xenograft model.
- Published
- 2016
- Full Text
- View/download PDF
127. CSIG-17. PRMT5 INHIBITION SENSITIZES GLIOBLASTOMA MODELS TO TRAMETINIB TREATMENT
- Author
-
Hannah Sur, Ji Young Yoo, Shilpa Thammegowda, Toshihiko Shimizu, Balveen Kaur, John D. Heiss, Yeshavanth Kumar Banasavadi-Siddegowda, Dragan Maric, Yoshihiro Otani, Sriya Namagiri, Ashis Chowdhury, Tae Jin Lee, and Cole T. Lewis
- Subjects
Trametinib ,Cancer Research ,Proto-Oncogene Proteins c-akt ,Chemistry ,Protein arginine methyltransferase 5 ,Cell Signaling and Signaling Pathways ,Cell cycle ,Oncology ,Apoptosis ,Tumor progression ,Cancer research ,Neurology (clinical) ,Signal transduction ,Survival rate - Abstract
INTRODUCTION Glioblastoma (GBM) is the most common malignant primary brain tumor. With limited effective therapeutic strategies, prognosis for GBM is very poor. Our previous study shows that the expression of Protein Arginine Methyltransferase 5 (PRMT5) is upregulated in GBM; its inhibition promotes anti-GBM effect through apoptosis and senescence of mature and immature tumor cells, respectively. In GBM, RAS-RAF- MEK-ERK signaling is aberrantly activated and promotes tumor growth. Therefore, MEK inhibitors, including trametinib are currently under investigation for GBM therapy. In this study, we tested whether inhibition of PRMT5 can enhance the anti-tumor efficacy of trametinib in GBM. METHODS Patient-derived primary GBM neurospheres (GBMNS) with transient PRMT5 knockdown were treated with trametinib and cell viability, proliferation, cell cycle progression, and western blot analysis were conducted. In vivo, PRMT5-intact and -depleted GBMNS were intracranially implanted in NSG mice and treated with trametinib by daily oral gavage, and tumor progression and mice survival rate were analyzed by MRI and Kaplan-Meier survival curve, respectively. RESULTS Trametinib treatment upregulated the expression of PRMT5 in GBMNS. Depletion of PRMT5 increased the cytotoxic effect of trametinib in GBMNS. In concurrence with the trametinib-induced PRMT5 upregulation, trametinib treatment increased the activity of AKT that was blocked with PRMT5 knockdown. In vivo, PRMT5-depletion extended the survival of the tumor bearing mice that further increased in combination with trametinib treatment. Interestingly, trametinib treatment alone had no survival benefit. CONCLUSION Trametinib treatment induces PRMT5 expression. Depletion of trametinib-induced PRMT5 expression sensitizes GBMNS for trametinib by inhibiting AKT activity.
- Published
- 2020
- Full Text
- View/download PDF
128. Cytokine Microdialysis for Real-Time Immune Monitoring in Glioblastoma Patients Undergoing Checkpoint Blockade
- Author
-
Prashant Chittiboina, Averie Kuek, Mark R. Gilbert, John Lynes, Deric M. Park, Sarah Benzo, Sadhana Jackson, Kareem A. Zaghloul, Xiang Wang, Christina Hayes, John D. Heiss, Tianxia Wu, Christopher S. Hourigan, Dragan Maric, Jing Wu, Gifty Dominah, Victoria Sanchez, Jinguo Chen, Gretchen Scott, Martha Quezado, Amber J. Giles, and Edjah K. Nduom
- Subjects
Oncology ,Adult ,medicine.medical_specialty ,Microdialysis ,Cell cycle checkpoint ,Side effect ,medicine.medical_treatment ,Brain tumor ,Phases of clinical research ,03 medical and health sciences ,Interferon-gamma ,0302 clinical medicine ,Monitoring, Immunologic ,Internal medicine ,medicine ,Humans ,Brain Chemistry ,business.industry ,Brain Neoplasms ,Immunotherapy ,medicine.disease ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Cytokines ,Surgery ,Neurology (clinical) ,Nivolumab ,Neoplasm Recurrence, Local ,business ,Glioblastoma ,Research—Human—Study Protocols ,030217 neurology & neurosurgery - Abstract
Background Glioblastoma is the most common primary malignancy of the brain, with a dismal prognosis. Immunomodulation via checkpoint inhibition has provided encouraging results in non-CNS malignancies, but prediction of responders has proven to be challenging in glioblastoma patients. Objective To determine the proportion of patients who have a measurable increase of interferon gamma levels in brain tumor tissue after their first dose of nivolumab, and to evaluate the safety of using brain tumor microdialysis to monitor for immune response while evaluating the safety of the combination of anti-programmed death 1 (PD-1) and anti-lymphocyte activation gene 3 (LAG-3) checkpoint inhibition. Methods The study design is a single-center, nonrandomized phase 1 clinical trial. Up to 15 adult patients with recurrent glioblastoma will be enrolled with the goal of 10 patients completing the trial over an anticipated 18 mo. Patients will undergo biopsy; placement of microdialysis catheters and lumbar drains; treatment with anti-PD-1 checkpoint inhibition; comprehensive immune biomarker collection; tumor resection; and then treatment with anti-PD-1 and anti-LAG-3 checkpoint inhibition until progression. Expected outcomes We expect interferon gamma levels to increase in the brain as measured via microdialysis in treated patients. Based on published reports, microdialysis in this patient population is expected to be safe, and anti-LAG-3 and anti-PD-1 combined will likely have a similar side effect profile to other checkpoint inhibitor combinations. Discussion The failure of recent trials of immune therapies in glioblastoma underscores the need to appropriately measure response in the treated tissue. This trial may provide insight on indicators of which patients will respond to immune therapy.
- Published
- 2018
129. Neural precursor cells form integrated brain-like tissue when implanted into rat cerebrospinal fluid
- Author
-
Stephen J. Dodd, Jung-Hwa Tao-Cheng, Dragan Maric, Alan P. Koretsky, Alec Calac, Kathryn Sharer, James Pickel, Nikorn Pothayee, and Nadia Bouraoud
- Subjects
0301 basic medicine ,Medicine (miscellaneous) ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Cortex (anatomy) ,Precursor cell ,otorhinolaryngologic diseases ,medicine ,lcsh:QH301-705.5 ,Microglia ,Chemistry ,Regeneration (biology) ,fungi ,Neural stem cell ,Cell biology ,Transplantation ,stomatognathic diseases ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,lcsh:Biology (General) ,GABAergic ,General Agricultural and Biological Sciences ,030217 neurology & neurosurgery - Abstract
There is tremendous interest in transplanting neural precursor cells for brain tissue regeneration. However, it remains unclear whether a vascularized and integrated complex neural tissue can be generated within the brain through transplantation of cells. Here, we report that early stage neural precursor cells recapitulate their seminal properties and develop into large brain-like tissue when implanted into the rat brain ventricle. Whereas the implanted cells predominantly differentiated into glutamatergic neurons and astrocytes, the host brain supplied the intact vasculature, oligodendrocytes, GABAergic interneurons, and microglia that seamlessly integrated into the new tissue. Furthermore, local and long-range axonal connections formed mature synapses between the host brain and the graft. Implantation of precursor cells into the CSF-filled cavity also led to a formation of brain-like tissue that integrated into the host cortex. These results may constitute the basis of future brain tissue replacement strategies. Nikorn Pothayee et al. show that early neural precursor cells (NPCs) derived from the embryonic telencephalon or midbrain can develop into brain-like tissue when implanted into the rat brain ventricle. Telencephalon-derived NPCs also form brain tissue in the host cortex when implanted into a CSF-filled cavity formed by cortical ablation.
- Published
- 2018
- Full Text
- View/download PDF
130. Neocortical Projection Neurons Instruct Inhibitory Interneuron Circuit Development in a Lineage-Dependent Manner
- Author
-
Jason C. Wester, Yajun Zhang, Timothy J. Petros, Sanan Venkatesh, Xiaoqing Yuan, Daniela Calvigioni, Christopher T. Rhodes, Chris J. McBain, Dragan Maric, Vivek Mahadevan, and Steven Hunt
- Subjects
0301 basic medicine ,Telencephalon ,Cell signaling ,animal structures ,Ganglionic eminence ,Interneuron ,Pyramidal Tracts ,Gene Expression ,Neocortex ,Biology ,Inhibitory postsynaptic potential ,Article ,03 medical and health sciences ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Cell Movement ,Interneurons ,Neural Pathways ,medicine ,Animals ,Cell Lineage ,Transcription factor ,Neurons ,Pyramidal tracts ,Sequence Analysis, RNA ,Pyramidal Cells ,General Neuroscience ,RNA ,Neural Inhibition ,Matrix Attachment Region Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Synapses ,Excitatory postsynaptic potential ,sense organs ,Single-Cell Analysis ,Neuroscience ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Neocortical circuits consist of stereotypical motifs that must self-assemble during development. Recent evidence suggests that the subtype identity of both excitatory projection neurons (PNs) and inhibitory interneurons (INs) is important for this process. We knocked out the transcription factor Satb2 in PNs to induce those of the intratelencephalic (IT) type to adopt a pyramidal tract (PT)-type identity. Loss of IT-type PNs selectively disrupted the lamination and circuit integration of INs derived from the caudal ganglionic eminence (CGE). Strikingly, reprogrammed PNs demonstrated reduced synaptic targeting of CGE-derived INs relative to controls. In control mice, IT-type PNs targeted neighboring CGE INs, while PT-type PNs did not in deep layers, confirming this lineage-dependent motif. Finally, single-cell RNA sequencing revealed that major CGE IN subtypes were conserved after loss of IT PNs, but with differential transcription of synaptic proteins and signaling molecules. Thus, IT-type PNs influence CGE-derived INs in a non-cell-autonomous manner during cortical development.
- Published
- 2018
131. Novel Targeting of Transcription and Metabolism in Glioblastoma
- Author
-
Chunzhang Yang, Dragan Maric, Jing Wu, Li-Yuan Chen, Herui Wang, Kristan Meetze, Wei Zhang, Zhengping Zhuang, Mioara Larion, Hallie Lappin, Gabriel Vasconcelos, Adrian Lita, Mones Abu-Asab, Mark R. Gilbert, Yu-Ting Su, Hua Song, Tomas Estok, Qi Zhang, Orieta Celiku, Aiguo Li, and Robert T. Chen
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Transcription, Genetic ,Drug Evaluation, Preclinical ,Apoptosis ,Biology ,Heterocyclic Compounds, 4 or More Rings ,Article ,03 medical and health sciences ,Mice ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Temozolomide ,Animals ,Humans ,Cytotoxicity ,Cell Proliferation ,Cell growth ,Brain Neoplasms ,Brain ,Drug Synergism ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Cell killing ,Oncology ,Cell culture ,Cancer cell ,Cancer research ,Energy Metabolism ,Glioblastoma ,medicine.drug - Abstract
Purpose: Glioblastoma (GBM) is highly resistant to treatment, largely due to disease heterogeneity and resistance mechanisms. We sought to investigate a promising drug that can inhibit multiple aspects of cancer cell survival mechanisms and become an effective therapeutic for GBM patients. Experimental Design: To investigate TG02, an agent with known penetration of the blood–brain barrier, we examined the effects as single agent and in combination with temozolomide, a commonly used chemotherapy in GBM. We used human GBM cells and a syngeneic mouse orthotopic GBM model, evaluating survival and the pharmacodynamics of TG02. Mechanistic studies included TG02-induced transcriptional regulation, apoptosis, and RNA sequencing in treated GBM cells as well as the investigation of mitochondrial and glycolytic function assays. Results: We demonstrated that TG02 inhibited cell proliferation, induced cell death, and synergized with temozolomide in GBM cells with different genetic background but not in astrocytes. TG02-induced cytotoxicity was blocked by the overexpression of phosphorylated CDK9, suggesting a CDK9-dependent cell killing. TG02 suppressed transcriptional progression of antiapoptotic proteins and induced apoptosis in GBM cells. We further demonstrated that TG02 caused mitochondrial dysfunction and glycolytic suppression and ultimately ATP depletion in GBM. A prolonged survival was observed in GBM mice receiving combined treatment of TG02 and temozolomide. The TG02-induced decrease of CDK9 phosphorylation was confirmed in the brain tumor tissue. Conclusions: TG02 inhibits multiple survival mechanisms and synergistically decreases energy production with temozolomide, representing a promising therapeutic strategy in GBM, currently under investigation in an ongoing clinical trial. Clin Cancer Res; 24(5); 1124–37. ©2017 AACR.
- Published
- 2017
132. Molecular Signatures of Mouse TRPV1-Lineage Neurons Revealed by RNA-Seq Transcriptome Analysis
- Author
-
Gian Luigi Gonnella, Jason M. Keller, Michael J. Iadarola, Dragan Maric, Samuel Clokie, Mark A. Hoon, Samridhi C. Goswami, Santosh K. Mishra, Andrew J. Mannes, Hal D. Kominsky, Jacklyn R. Gross, and Krisztian Kaszas
- Subjects
Lineage (genetic) ,Population ,TRPV1 ,Gene Expression ,Pain ,TRPV Cation Channels ,Mice, Transgenic ,RNA-Seq ,Biology ,Article ,Transcriptome ,Transient receptor potential channel ,Species Specificity ,Dorsal root ganglion ,Ganglia, Spinal ,medicine ,Animals ,Cell Lineage ,Neurons, Afferent ,Trigeminal Nerve ,education ,In Situ Hybridization ,education.field_of_study ,Gene Expression Profiling ,Immunohistochemistry ,Rats ,Anesthesiology and Pain Medicine ,medicine.anatomical_structure ,nervous system ,Neurology ,Neurology (clinical) ,Neuroglia ,Neuroscience ,Nucleus - Abstract
Disorders of pain neural systems are frequently chronic and, when recalcitrant to treatment, can severely degrade the quality of life. The pain pathway begins with sensory neurons in dorsal root or trigeminal ganglia, and the neuronal subpopulations that express the transient receptor potential cation channel, subfamily V, member 1 (TRPV1) ion channel transduce sensations of painful heat and inflammation and play a fundamental role in clinical pain arising from cancer and arthritis. In the present study, we elucidate the complete transcriptomes of neurons from the TRPV1 lineage and a non-TRPV1 neuroglial population in sensory ganglia through the combined application of next-gen deep RNA-Seq, genetic neuronal labeling with fluorescence-activated cell sorting, or neuron-selective chemoablation. RNA-Seq accurately quantitates gene expression, a difficult parameter to determine with most other methods, especially for very low and very high expressed genes. Differentially expressed genes are present at every level of cellular function from the nucleus to the plasma membrane. We identified many ligand receptor pairs in the TRPV1 population, suggesting that autonomous presynaptic regulation may be a major regulatory mechanism in nociceptive neurons. The data define, in a quantitative, cell population–specific fashion, the molecular signature of a distinct and clinically important group of pain-sensing neurons and provide an overall framework for understanding the transcriptome of TRPV1 nociceptive neurons. Perspective Next-gen RNA-Seq, combined with molecular genetics, provides a comprehensive and quantitative measurement of transcripts in TRPV1 lineage neurons and a contrasting transcriptome from non-TRPV1 neurons and cells. The transcriptome highlights previously unrecognized protein families, identifies multiple molecular circuits for excitatory or inhibitory autocrine and paracrine signaling, and suggests new combinatorial approaches to pain control.
- Published
- 2014
- Full Text
- View/download PDF
133. EXTH-62. TG02, A NOVEL TARGETING OF TRANSCRIPTION AND METABOLISM IN GLIOBLASTOMA
- Author
-
Jing Wu, Hua Song, Orieta Celiku, Dragan Maric, Tom Estok, Chunzhang Yang, Yu-Ting Su, Kristan Meetze, Wei Zhang, Zhengping Zhuang, Robert T. Chen, Qi Zhang, Aiguo Li, and Mark R. Gilbert
- Subjects
Cancer Research ,business.industry ,Computational biology ,Metabolism ,Biology ,medicine.disease ,Abstracts ,Text mining ,Oncology ,Transcription (biology) ,medicine ,Neurology (clinical) ,business ,Glioblastoma - Published
- 2017
134. IMMU-02. VALIDATION OF PD-L1 EXPRESSION IN HIGH GRADE GLIOMAS AS AN INDEPENDENT NEGATIVE PROGNOSTIC MARKER
- Author
-
Edjah K. Nduom, Xiang Wang, Graham Lobel, Gifty Dominah, Martha Quezado, Nancy A. Edwards, Arnold Obungu, Dragan Maric, Mark R. Gilbert, and Drew Pratt
- Subjects
Cancer Research ,business.industry ,Cancer ,T lymphocyte ,medicine.disease ,Log-rank test ,Abstracts ,Immunophenotyping ,Text mining ,Plasmid ,Oncology ,Cancer research ,Medicine ,Immunohistochemistry ,Pd l1 expression ,Neurology (clinical) ,business - Published
- 2017
135. LB-100, a novel Protein Phosphatase 2A (PP2A) inhibitor, sensitizes malignant meningioma cells to the therapeutic effects of radiation
- Author
-
Deric M. Park, Hua Song, John D. Heiss, Dragan Maric, Tamalee Kramp, Anita Tandle, Shuyu Hao, Rongze Lu, Zhengping Zhuang, Mark R. Gilbert, Ashlee Seldomridge, Saman Sizdahkhani, Winson S. Ho, and Kevin Camphausen
- Subjects
Cancer Research ,Malignant meningioma ,DNA repair ,Cell Survival ,Mice, Nude ,Disease-Free Survival ,Piperazines ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Meningeal Neoplasms ,Animals ,Humans ,Radiosensitivity ,Protein Phosphatase 2 ,Clonogenic assay ,Mitotic catastrophe ,Mice, Inbred BALB C ,Chemistry ,Protein phosphatase 2 ,Chemoradiotherapy ,Bridged Bicyclo Compounds, Heterocyclic ,Xenograft Model Antitumor Assays ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Radiosensitizing Agent ,Female ,Meningioma ,030217 neurology & neurosurgery - Abstract
Atypical and anaplastic meningiomas (AAM) represent 20% of all meningiomas. They are associated with poor outcomes due to their tendency to recur. While surgery and radiation (RT) are first line therapy, no effective systemic medical treatment has been identified. Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase involved in cell cycle regulation and DNA repair. Here, we examined radiosensitizing effects of LB-100, a novel inhibitor of PP2A against AAM as a novel treatment strategy. Three human-derived immortalized meningioma cell lines, IOMM-LEE, GAR, and CH-157, were used to investigate the radio-sensitizing potential of LB-100 in AAM. Survival fraction by clonogenic assay, immunofluorescence, cell cycle analysis and protein expression were evaluated in vitro. The antitumor effects of combining LB-100 with RT were verified in vivo by using intracranial orthotopic xenograft mouse model. Pharmacologic PP2A inhibition with LB-100 prior to RT enhanced the radiosensitivity of meningioma cells and reduced survival fraction in clonogenic assays. LB-100 increased DNA double-strand breakage (measured by γ-H2AX), mitotic catastrophe cell death, and G2/M cell cycle arrest in irradiated meningioma cells. Also, LB-100 decreased activation of STAT3 and expression of its downstream proteins. In vivo, LB-100 and RT combined treatment prolonged the survival of mice with xenografts compared to RT alone. Taken together, these results provide convincing preclinical data to support the use of LB-100 as a radiosensitizing agent for treatment of malignant meningioma. Its potential for clinical application deserves further investigation.
- Published
- 2017
136. Cross-sectional and longitudinal small animal PET shows pre and post-synaptic striatal dopaminergic deficits in an animal model of HIV
- Author
-
Sanhita Sinharay, Georgios Z. Papadakis, Dianne E. Lee, William Reid, Dima A. Hammoud, Siva Muthusamy, Dragan Maric, Swati Shah, and Xiang Zhang
- Subjects
0301 basic medicine ,Male ,Cancer Research ,medicine.medical_specialty ,Pathology ,Dopamine ,HIV Infections ,Mice, Transgenic ,HIV Envelope Protein gp120 ,Neuroprotection ,Article ,03 medical and health sciences ,0302 clinical medicine ,Dopamine receptor D3 ,Postsynaptic potential ,Internal medicine ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Dopamine transporter ,Tyrosine hydroxylase ,biology ,business.industry ,Dopaminergic ,Neurotoxicity ,medicine.disease ,Rats ,Neostriatum ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Cross-Sectional Studies ,Positron-Emission Tomography ,Synapses ,biology.protein ,HIV-1 ,Molecular Medicine ,NeuN ,Rats, Transgenic ,business ,030217 neurology & neurosurgery - Abstract
Introduction In vivo imaging biomarkers of various HIV neuropathologies, including dopaminergic dysfunction, are still lacking. Towards developing dopaminergic biomarkers of brain involvement in HIV, we assessed the pre and postsynaptic components of the dopaminergic system in the HIV-1 transgenic rat (Tg), a well-characterized model of treated HIV + patients, using small-animal PET imaging. Methods Fifteen to 18 month-old Tg and wild type (WT) rats were imaged with both [18F]-FP-CMT, a dopamine transporter (DAT) ligand (n = 16), and [18F]-Fallypride, a D2/D3 dopamine receptor (D2/D3DR) ligand (n = 16). Five to 8 month-old Tg and WT rats (n = 18) were also imaged with [18F]-FP-CMT. A subset of animals was imaged longitudinally at 7 and 17 months of age. Multiplex immunohistochemistry staining for DAT, tyrosine hydroxylase, D2DR, D3DR, GFAP, Iba1 and NeuN was performed on a subgroup of the scanned animals. Results [18F]-FP-CMT and [18F]-Fallypride binding potential (BPND) values were significantly lower in 15–18 month-old Tg compared to age-matched WT rats (p Conclusions We found presynaptic and postsynaptic dopaminergic dysfunction/loss in older Tg compared to WT rats. We believe this to be related to neurotoxicity of viral proteins present in the Tg rats’ serum and brain. Advances in knowledge Our findings confirm prior reports of neurobehavioral abnormalities suggestive of dopaminergic dysfunction in this model. They also suggest similarities between the Tg rat and HIV + patients as far as dopaminergic dysfunction. Implications for patient care The Tg rat, along with the above-described quantitative PET imaging biomarkers, can have a role in the evaluation of HIV neuroprotective therapies prior to human translation.
- Published
- 2017
137. Histone Deacetylase Inhibitor SAHA Is a Promising Treatment of Cushing Disease
- Author
-
Prashant Chittiboina, Xiang Wang, Alejandro Bugarini, Stuart Walbridge, Jie Lu, Dragan Maric, Grégoire P Chatain, and Zhengping Zhuang
- Subjects
0301 basic medicine ,Pro-Opiomelanocortin ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Apoptosis ,Hydroxamic Acids ,Biochemistry ,Mice ,0302 clinical medicine ,Endocrinology ,Medicine ,Corticotrophs ,Vorinostat ,Histone deacetylase inhibitor ,Liver X receptor alpha ,Flow Cytometry ,ACTH-Secreting Pituitary Adenoma ,030220 oncology & carcinogenesis ,Female ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug ,Adenoma ,medicine.medical_specialty ,endocrine system ,medicine.drug_class ,Cell Survival ,Blotting, Western ,Mice, Nude ,Enzyme-Linked Immunosorbent Assay ,In Vitro Techniques ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,Adrenocorticotropic Hormone ,In vivo ,Internal medicine ,Cell Line, Tumor ,Animals ,Humans ,Viability assay ,Pituitary ACTH Hypersecretion ,Clinical Research Articles ,business.industry ,Gene Expression Profiling ,Biochemistry (medical) ,Xenograft Model Antitumor Assays ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Cell culture ,Corticotropic cell ,business - Abstract
Context Remission failure following transsphenoidal surgery in Cushing disease (CD) from pituitary corticotroph tumors (CtTs) remains clinically challenging. Histone deacetylase inhibitors (HDACis) are antitumor drugs approved for clinical use, with the potential to affect adrenocorticotropin hormone (ACTH) hypersecretion by inhibiting pro-opiomelanocortin (POMC) transcription. Objective Testing the efficacy of suberoylanilide hydroxamic acid (SAHA) on human and murine ACTH-secreting tumor (AtT-20) cells. Design Cell viability, ACTH secretion (enzyme-linked immunosorbent assay), apoptosis, and gene expression profile were investigated on AtT-20 cells. In vivo efficacy was examined in an athymic nude mouse AtT-20 xenograft model. SAHA efficacy against human-derived corticotroph tumor (hCtT) (n = 8) was tested in vitro. Setting National Institutes of Health. Intervention SAHA (0.5 to 8 µM). Main Outcome Measures AtT-20 and hCtT cell survival, in vitro/invivo ACTH measurements. Results SAHA (1 µM) reduced AtT-20 viability to 75% at 24 hours, 43% at 48 hours (analysis of variance; P = 0.002). Apoptosis was confirmed with elevated BAX/Bcl2 ratio and FACS. Intriguingly, early (3-hour) significant decline (70%; P < 0.0001) of secreted ACTH and diminished POMC transcription was observed with SAHA (1 µM). Microarray analysis revealed a direct association between liver X receptor alpha (LXRα) and POMC expression. Accordingly, SAHA reduced LXRα in AtT-20 cells but not in normal murine corticotrophs. Xenografted nude-mice tumor involution (126 ± 33/160 ± 35 vs 337 ± 49 mm3; P = 0.0005) was observed with 5-day intraperitoneal SAHA, with reversal of elevated ACTH (P < 0.0001). SAHA did not affect serum ACTH in nontumor mice. Lastly, we confirmed that SAHA (1 µM/24 h) decreased hCtT survival (78.92%; P = 0.0007) and ACTH secretion (83.64%; P = 0.03). Conclusion Our findings demonstrate SAHA’s efficacy in reducing survival and ACTH secretion in AtT-20 and hCtT cells, providing a potential intervention for recurrent/unremitting CD.
- Published
- 2017
138. Neural stem cell transplantation in ischemic stroke: A role for preconditioning and cellular engineering
- Author
-
John M. Hallenbeck, Luca Peruzzotti-Jametti, Stefano Pluchino, Yang-ja Lee, Nunzio Vicario, Daniel Ye, Joshua D. Bernstock, Dragan Maric, and Florian Gessler
- Subjects
0301 basic medicine ,Transplantation Conditioning ,Cell Survival ,cellular engineering ,Brain Ischemia ,Brain ischemia ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,preconditioning ,post-translational modifications ,medicine ,Humans ,Stroke ,Cell Engineering ,Ischemic stroke ,biology ,business.industry ,Mini-Review ,medicine.disease ,Neural stem cell ,Cellular engineering ,Transplantation ,030104 developmental biology ,Neurology ,neural stem cells ,Stem Cell Transplantation ,biology.protein ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business ,Neuroscience ,030217 neurology & neurosurgery ,Neurotrophin - Abstract
Ischemic stroke continues to be a leading cause of morbidity and mortality throughout the world. To protect and/or repair the ischemic brain, a multitiered approach may be centered on neural stem cell (NSC) transplantation. Transplanted NSCs exert beneficial effects not only via structural replacement, but also via immunomodulatory and/or neurotrophic actions. Unfortunately, the clinical translation of such promising therapies remains elusive, in part due to their limited persistence/survivability within the hostile ischemic microenvironment. Herein, we discuss current approaches for the development of NSCs more amenable to survival within the ischemic brain as a tool for future cellular therapies in stroke.
- Published
- 2017
139. Somatostatin receptor expression on von Hippel-Lindau-associated hemangioblastomas offers novel therapeutic target
- Author
-
Prashant Chittiboina, Zhengping Zhuang, Saman Sizdahkhani, Marsha J. Merrill, Abhik Ray-Chaudhury, Michael J. Feldman, Nancy A. Edwards, Alexander Ksendzovsky, Martin G. Piazza, Karel Pacak, and Dragan Maric
- Subjects
Stromal cell ,von Hippel-Lindau Disease ,endocrine system diseases ,Octreotide ,Apoptosis ,urologic and male genital diseases ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Hemangioblastoma ,Organometallic Compounds ,Medicine ,Humans ,Somatostatin receptor 1 ,Receptors, Somatostatin ,neoplasms ,Cells, Cultured ,Cytopenia ,Multidisciplinary ,Somatostatin receptor ,business.industry ,Brain Neoplasms ,Middle Aged ,medicine.disease ,female genital diseases and pregnancy complications ,3. Good health ,Vascular endothelial growth factor ,Somatostatin ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Radiopharmaceuticals ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Von Hippel-Lindau (VHL)-associated hemangioblastomas (VHL-HB) arise in the central nervous system (CNS), and are a leading cause of morbidity and mortality in VHL disease. Currently, surgical resection is the most effective way to manage symptomatic VHL-HBs. Surgically unresectable VHL-HBs or those in frail patients are challenging problems. Therapies targeting oncologic and vascular endothelial growth factor (VEGF) pathways have failed to demonstrate tumor control. Our experience and previous reports on VHL-HB avidity to somatostatin analogues suggested somatostatin receptor (SSTR) expression in VHL-HBs, offering an alternative therapeutic strategy. We explored this possibility by demonstrating consistent histologic expression of SSTR1, 2a, 4, and 5 in VHL-HBs. We found that somatostatin analogue octreotide induces apoptosis in VHL-HB stromal cells in a dose-dependent fashion by BAX – caspase-3 pathway unrelated to canonical VHL pathway. When administered to a patient with unresectable symptomatic suprasellar hemangioblastoma, octreotide resulted in tumor volume reduction, symptom stabilization, and tumor cytopenia on repeat 68Ga-DOTA-TATE positron emission tomography (PET) within 6 months, suggesting tumor infarction. We conclude that VHL-HBs harbor multiple SSTR subtypes that offer actionable chemo-therapeutic strategy for management of symptomatic, unresectable tumors by somatostatin analogue therapy.
- Published
- 2017
140. Tumor-Infiltrating Myeloid Cells Activate Dll4/Notch/TGF-β Signaling to Drive Malignant Progression
- Author
-
Dunrui Wang, Ombretta Salvucci, Giovanna Tosato, Kan Jiang, Dragan Maric, Hyeongil Kwak, David Sánchez-Martín, and Hidetaka Ohnuki
- Subjects
Male ,Cancer Research ,Myeloid ,Notch signaling pathway ,Article ,Mice ,Transforming Growth Factor beta ,Tumor Microenvironment ,medicine ,Animals ,Myeloid Cells ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Tumor microenvironment ,Receptors, Notch ,biology ,Calcium-Binding Proteins ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Neoplasms, Experimental ,Transforming growth factor beta ,Squamous carcinoma ,medicine.anatomical_structure ,Oncology ,Tumor progression ,Hes3 signaling axis ,Disease Progression ,biology.protein ,Cancer research ,Cyclin-dependent kinase 8 ,Female ,Signal Transduction - Abstract
Myeloid cells that orchestrate malignant progression in the tumor microenvironment offer targets for a generalized strategy to attack solid tumors. Through an analysis of tumor microenvironments, we explored an experimental model of lung cancer that uncovered a network of Dll4/Notch/TGF-β1 signals that links myeloid cells to cancer progression. Myeloid cells attracted to the tumor microenvironment by the tumor-derived cytokines CCL2 and M-CSF expressed increased levels of the Notch ligand Dll4, thereby activating Notch signaling in the tumor cells and amplifying tumor-intrinsic Notch activation. Heightened Dll4/Notch signaling in tumor cells magnified TGF-β–induced pSMAD2/3 signaling and was required to sustain TGF-β–induced tumor cell growth. Conversely, Notch blockade reduced TGF-β signaling and limited lung carcinoma tumor progression. Corroborating these findings, by interrogating RNAseq results from tumor and adjacent normal tissue in clinical specimens of human head and neck squamous carcinoma, we found evidence that TGF-β/Notch crosstalk contributed to progression. In summary, the myeloid cell-carcinoma signaling network we describe uncovers novel mechanistic links between the tumor microenvironment and tumor growth, highlighting new opportunities to target tumors where this network is active. Cancer Res; 74(7); 2038–49. ©2014 AACR.
- Published
- 2014
- Full Text
- View/download PDF
141. Dephosphorylation of Barrier-to-autointegration Factor by Protein Phosphatase 4 and Its Role in Cell Mitosis
- Author
-
Robert Craigie, Elena Semenova, Dragan Maric, and Xiaolei Zhuang
- Subjects
Blotting, Western ,Barrier-to-autointegration factor ,Mitosis ,DNA and Chromosomes ,Protein Serine-Threonine Kinases ,Biology ,Biochemistry ,Phosphoprotein Phosphatases ,Serine ,medicine ,Humans ,Phosphorylation ,Nuclear membrane ,Telophase ,Nuclear protein ,Molecular Biology ,Microscopy, Confocal ,Cell Cycle ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,Cell Biology ,Cell cycle ,Molecular biology ,Cell biology ,Chromatin ,DNA-Binding Proteins ,HEK293 Cells ,medicine.anatomical_structure ,Mutation ,RNA Interference ,HeLa Cells - Abstract
Barrier-to-autointegration factor (BAF or BANF1) is highly conserved in multicellular eukaryotes and was first identified for its role in retroviral DNA integration. Homozygous BAF mutants are lethal and depletion of BAF results in defects in chromatin segregation during mitosis and subsequent nuclear envelope assembly. BAF exists both in phosphorylated and unphosphorylated forms with phosphorylation sites Thr-2, Thr-3, and Ser-4, near the N terminus. Vaccinia-related kinase 1 is the major kinase responsible for phosphorylation of BAF. We have identified the major phosphatase responsible for dephosphorylation of Ser-4 to be protein phosphatase 4 catalytic subunit. By examining the cellular distribution of phosphorylated BAF (pBAF) and total BAF (tBAF) through the cell cycle, we found that pBAF is associated with the core region of telophase chromosomes. Depletion of BAF or perturbing its phosphorylation state results not only in nuclear envelope defects, including mislocalization of LEM domain proteins and extensive invaginations into the nuclear interior, but also impaired cell cycle progression. This phenotype is strikingly similar to that seen in cells from patients with progeroid syndrome resulting from a point mutation in BAF.
- Published
- 2014
- Full Text
- View/download PDF
142. EXTH-44. INHIBITION OF MerTK ACTIVATES GLIOBLASTOMA-ASSOCIATED MACROPHAGES AND INDUCES TUMOR CELL DEATH IN GLIOMA MICROENVIRONMENT
- Author
-
Shelton Earp, Masaki Terabe, Jing Wu, Madison Butler, Yu-Ting Su, Dragan Maric, Lee Hwang, and Mark R. Gilbert
- Subjects
Cancer Research ,Tumor microenvironment ,Microglia ,Cell growth ,Chemistry ,C-Mer Tyrosine Kinase ,MERTK ,medicine.disease ,Apoptotic cell clearance ,MERTK Gene ,medicine.anatomical_structure ,Oncology ,Glioma ,medicine ,Cancer research ,Experimental Therapeutics ,Neurology (clinical) - Abstract
BACKGROUND Glioblastoma-associated macrophages and microglia (GAMs) are the predominant immune cells in the tumor microenvironment. Activation of MerTK, a receptor tyrosine kinase, triggers efferocytosis and polarizes GAMs to an immunosuppressive phenotype, promoting glioma growth. Our previous findings showed that UNC2371, a small-molecule inhibitor of MerTK, induced a less immunosuppressive phenotype of GAMs. Here, we investigate the role of MerTK inhibition on glioblastoma cells in the tumor microenvironment in vitro and in vivo. METHODS Cytotoxicity of UNC2371 in glioblastoma cells was determined by cell viability and colony formation assays. The protein expression of MerTK, AKT, and Erk were quantified by Western blotting in UNC2371-treated glioblastoma cells. A syngeneic GL261 mouse orthotopic glioblastoma model was used to evaluate the survival benefit of UNC2371 treatment. Fluorescent multiplex immunohistochemistry (IHC) was used to evaluate the expression of CD206, an anti-inflammatory marker on GAMs in murine brain tumor tissues. RESULTS UNC2371 inhibited GBM cell growth with an EC50 < 100 nM in both human U251 and mouse GL261 glioma cells, but not in GAMs. UNC2371-induced cell death and decreased cell proliferation were demonstrated by colony formation assays. UNC2371 decreased protein expression of phosphorylated MerTK, AKT, and Erk, which are essential for cell survival signaling, in U251 and GL261 cells. Furthermore, UNC2371 treatment prolonged survival in the mouse orthotopic GL261 glioblastoma model, suggesting that UNC2371 induces glioma cell death. A decreased of CD206+ GAMs was found in mice glioma tissues by fluorescent multiplex IHC, consistent with our previous findings in the in vitro cell-based assays. These data suggest that in addition to alleviate immunosuppression in the glioma microenvironment, UNC2371 directly inhibits GBM cell growth in vitro and in vivo. CONCLUSION Our findings suggest that UNC2371 has a therapeutic benefit via promoting GAM polarization towards proinflammatory status in the glioblastoma microenvironment and unexpectedly, inducing tumor cell death.
- Published
- 2019
- Full Text
- View/download PDF
143. ATIM-32. PREDICTORS OF IMPROVED SURVIVAL FOLLOWING ONCOLYTIC VIRUS TREATMENT IN PATIENTS WITH RECURRENT GLIOBLASTOMA: GENE EXPRESSION ANALYSIS FROM THE PHASE IB G207 CLINICAL TRIAL
- Author
-
Nripesh Prasad, G. Yancey Gillespie, Gary Cutter, Naomi J. Barker, Justin C. Roth, Jennifer M. Coleman, Bernard Roizman, Kathleen M. Schieffer, Anthony R. Miller, Kevin A. Cassady, Josh Bernstock, James M. Markert, Katherine E. Miller, Kristen M. Leraas, Dragan Maric, Richard J. Whitley, Elaine R. Mardis, and Jianmei W. Leavenworth
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Adult Clinical Trials–Immunologic ,Recurrent glioblastoma ,Improved survival ,Oncolytic virus ,Clinical trial ,Internal medicine ,Gene expression ,medicine ,In patient ,Neurology (clinical) ,business - Abstract
Our Phase I trials of experimental virotherapy for recurrent glioblastoma (GBM) have shown that inoculation with a conditionally replication-competent early generation oncolytic herpes simplex virus (oHSV), G207, is safe. However, while 17 of 37 subjects experienced objective clinical responses, the highly attenuated oHSV did not uniformly improve survival. We sought to identify predictors that would identify mechanisms contributing to survival and improve future trial design, by studying accrued samples. We analyzed pre-treatment biopsy and post-G207-treatment tumor samples (collected D2-5 post injection) banked from the patients enrolled in the phase IB G207 trial. The key findings from these patients suggest that productive G207 infection and G207-induced changes in gene expression were predictive of oHSV therapeutic success. RNAseq-based transcriptome analysis of these samples revealed that both the intrinsic IFN mediated antiviral response and adaptive immune functional response in patients correlated significantly with improved survival following G207 inoculation. Further, GBM tissue stained using multiplex fluorescent immunohistochemistry supported differences in the tumor microenvironments that were identified from RNAseq data analysis. Our data indicate that both viral gene expression and the resulting intrinsic anti-viral and recruited adaptive response were critical for survival after G207 inoculation and predict survival with this early generation oHSV in patients with recurrent malignant glioma.
- Published
- 2019
- Full Text
- View/download PDF
144. Spatiotemporal Control of ULK1 Activation by NDP52 and TBK1 during Selective Autophagy
- Author
-
Dragan Maric, Chunxin Wang, Richard J. Youle, Ling Hao, Giampietro Schiavo, Eric Bunker, Jose Norberto S. Vargas, and Felix Randow
- Subjects
Autophagosome ,TAX1BP1 ,ATG13 ,P62 ,Autophagy-Related Proteins ,PINK1 ,Protein Serine-Threonine Kinases ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,AMP-Activated Protein Kinase Kinases ,Lysosome ,Mitophagy ,Autophagy ,Peroxisomes ,medicine ,Autophagy-Related Protein-1 Homolog ,Humans ,Clustered Regularly Interspaced Short Palindromic Repeats ,FIP200 ,Phosphorylation ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Parkin ,030304 developmental biology ,0303 health sciences ,TOR Serine-Threonine Kinases ,Nuclear Proteins ,Cell Biology ,Protein-Tyrosine Kinases ,Autophagy-related protein 13 ,ULK1 ,optineurin ,Mitochondria ,Cell biology ,mitophagy ,medicine.anatomical_structure ,Multiprotein Complexes ,lysosome ,Protein Multimerization ,Protein Kinases ,030217 neurology & neurosurgery ,HeLa Cells ,Signal Transduction - Abstract
Summary Selective autophagy recycles damaged organelles and clears intracellular pathogens to prevent their aberrant accumulation. How ULK1 kinase is targeted and activated during selective autophagic events remains to be elucidated. In this study, we used chemically inducible dimerization (CID) assays in tandem with CRISPR KO lines to systematically analyze the molecular basis of selective autophagosome biogenesis. We demonstrate that ectopic placement of NDP52 on mitochondria or peroxisomes is sufficient to initiate selective autophagy by focally localizing and activating the ULK1 complex. The capability of NDP52 to induce mitophagy is dependent on its interaction with the FIP200/ULK1 complex, which is facilitated by TBK1. Ectopically tethering ULK1 to cargo bypasses the requirement for autophagy receptors and TBK1. Focal activation of ULK1 occurs independently of AMPK and mTOR. Our findings provide a parsimonious model of selective autophagy, which highlights the coordination of ULK1 complex localization by autophagy receptors and TBK1 as principal drivers of targeted autophagosome biogenesis., Graphical Abstract, Highlights • NDP52 associates with the ULK1 complex through FIP200, facilitated by TBK1 • NDP52/TBK1 targets ULK1 to cargo to initiate autophagy in the absence of LC3 • ULK1 is activated on cargo independently of AMPK and mTOR activity • Ectopic recruitment of FIP200-binding peptide is sufficient to degrade cargo, The targeting and activation mechanism of ULK1 complex during selective autophagy has been unclear. Here, Vargas et al. show that NDP52/TBK1 target the ULK1 complex to cargo in an LC3-independent manner and that ULK1 kinase activation is coupled to cargo localization, circumventing energy-sensing pathways.
- Published
- 2019
- Full Text
- View/download PDF
145. Meningeal inflammation drives long-term engraftment by monocytes that impair CNS immunity
- Author
-
Rua, Rejane, primary, Lee, Jane, additional, Silva, Alex, additional, Dragan, Maric, additional, Johnson, Kory, additional, and McGavern, Dorian B., additional
- Published
- 2018
- Full Text
- View/download PDF
146. Induction of IL-17 and nonclassical T-cell activation by HIV-Tat protein
- Author
-
Peter A. Calabresi, Rodrigo Hasbun, Avindra Nath, Kory R. Johnson, Karan Patel, Dragan Maric, and Tory P. Johnson
- Subjects
Gene Expression Regulation, Viral ,Virulence Factors ,T-Lymphocytes ,Lymphocyte ,T cell ,Immunoblotting ,Population ,Enzyme-Linked Immunosorbent Assay ,HIV Infections ,Biology ,Lymphocyte Activation ,Proinflammatory cytokine ,Immune Reconstitution Inflammatory Syndrome ,medicine ,Humans ,Secretion ,education ,Receptor ,education.field_of_study ,Multidisciplinary ,Interleukin-17 ,Brain ,Biological Sciences ,Flow Cytometry ,Microarray Analysis ,Cell biology ,medicine.anatomical_structure ,Anti-Retroviral Agents ,Immunology ,Leukocytes, Mononuclear ,tat Gene Products, Human Immunodeficiency Virus ,Interleukin 17 ,Signal transduction ,Signal Transduction - Abstract
Chronic immune activation is a major complication of antiretroviral therapy (ART) for HIV infection and can cause a devastating immune reconstitution inflammatory syndrome (IRIS) in the brain. The mechanism of T-cell activation in this population is not well understood. We found HIV-Tat protein and IL-17–expressing mononuclear cells in the brain of an individual with IRIS. Tat was also present in the CSF of individuals virologically controlled on ART. Hence we examined if Tat protein could directly activate T cells. Tat transcriptionally dysregulated 94 genes and induced secretion of 11 cytokines particularly activation of IL-17 signaling pathways supporting the development of a proinflammatory state. Tat increased IL-17 transcription and secretion in T cells. Tat entered the T cells rapidly by clathrin-mediated endocytosis and localized to both the cytoplasm and the nucleus. Tat activated T cells through a nonclassical pathway dependent upon vascular endothelial growth factor receptor-2 and downstream secondary signaling pathways but independent of the T-cell receptor. However, Tat stimulation of T cells did not induce T-cell proliferation but increased viral infectivity. This study demonstrates Tat’s role as a virulence factor, by driving T-cell activation and contributing to IRIS pathophysiology. This supports the necessity of an anti-Tat therapy in conjunction with ART and identifies multiple targetable pathways to prevent Tat-mediated T-cell activation.
- Published
- 2013
- Full Text
- View/download PDF
147. IRF-1 responsiveness to IFN-γ predicts different cancer immune phenotypes
- Author
-
Jennifer Reinboth, Patricia Fetsch, Lotfi Chouchane, Fm Marincola, Dragan Maric, Daniela Murtas, Armando C. Filie, Ml Ascierto, Ena Wang, Qiuzhen Liu, Andrea Worschech, De . Giorgi V, Sara Tomei, Davide Bedognetti, and Lorenzo Uccellini
- Subjects
Cancer Research ,Transcription, Genetic ,medicine.medical_treatment ,nuclear translocation ,Biology ,NF-κB ,Interferon-gamma ,Immune system ,Cell Line, Tumor ,medicine ,Humans ,Interferon gamma ,Melanoma ,IFN-γ ,beta Catenin ,IRF-1 ,Tumor Necrosis Factor-alpha ,TOR Serine-Threonine Kinases ,NF-kappa B ,Cancer ,Immunotherapy ,medicine.disease ,Phenotype ,Enzyme Activation ,Wnt Proteins ,IRF1 ,Oncology ,TNF-α ,Immunology ,immune phenotype ,Tumor necrosis factor alpha ,Translational Therapeutics ,Interferon Regulatory Factor-1 ,medicine.drug ,Interferon regulatory factors - Abstract
Background: Several lines of evidence suggest a dichotomy between immune active and quiescent cancers, with the former associated with a good prognostic phenotype and better responsiveness to immunotherapy. Central to such dichotomy is the master regulator of the acute inflammatory process interferon regulatory factor (IRF)-1. However, it remains unknown whether the responsiveness of IRF-1 to cytokines is able to differentiate cancer immune phenotypes. Methods: IRF-1 activation was measured in 15 melanoma cell lines at basal level and after treatment with IFN-γ, TNF-α and a combination of both. Microarray analysis was used to compare transcriptional patterns between cell lines characterised by high or low IRF-1 activation. Results: We observed a strong positive correlation between IRF-1 activation at basal level and after IFN-γ and TNF-α treatment. Microarray demonstrated that three cell lines with low and three with high IRF-1 inducible translocation scores differed in the expression of 597 transcripts. Functional interpretation analysis showed mTOR and Wnt/β-cathenin as the top downregulated pathways in the cell lines with low inducible IRF-1 activation, suggesting that a low IRF-1 inducibility recapitulates a cancer phenotype already described in literature characterised by poor prognosis. Conclusion: Our findings support the central role of IRF-1 in influencing different tumour phenotypes.
- Published
- 2013
- Full Text
- View/download PDF
148. 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016): part two
- Author
-
Casey Ager, Matthew Reilley, Courtney Nicholas, Todd Bartkowiak, Ashvin Jaiswal, Michael Curran, Tina C. Albershardt, Anshika Bajaj, Jacob F. Archer, Rebecca S. Reeves, Lisa Y. Ngo, Peter Berglund, Jan ter Meulen, Caroline Denis, Hormas Ghadially, Thomas Arnoux, Fabien Chanuc, Nicolas Fuseri, Robert W. Wilkinson, Nicolai Wagtmann, Yannis Morel, Pascale Andre, Michael B. Atkins, Matteo S. Carlino, Antoni Ribas, John A. Thompson, Toni K. Choueiri, F. Stephen Hodi, Wen-Jen Hwu, David F. McDermott, Victoria Atkinson, Jonathan S. Cebon, Bernie Fitzharris, Michael B. Jameson, Catriona McNeil, Andrew G. Hill, Eric Mangin, Malidi Ahamadi, Marianne van Vugt, Mariëlle van Zutphen, Nageatte Ibrahim, Georgina V. Long, Robyn Gartrell, Zoe Blake, Ines Simoes, Yichun Fu, Takuro Saito, Yingzhi Qian, Yan Lu, Yvonne M. Saenger, Sadna Budhu, Olivier De Henau, Roberta Zappasodi, Kyle Schlunegger, Bruce Freimark, Jeff Hutchins, Christopher A. Barker, Jedd D. Wolchok, Taha Merghoub, Elena Burova, Omaira Allbritton, Peter Hong, Jie Dai, Jerry Pei, Matt Liu, Joel Kantrowitz, Venus Lai, William Poueymirou, Douglas MacDonald, Ella Ioffe, Markus Mohrs, William Olson, Gavin Thurston, Cristian Capasso, Federica Frascaro, Sara Carpi, Siri Tähtinen, Sara Feola, Manlio Fusciello, Karita Peltonen, Beatriz Martins, Madeleine Sjöberg, Sari Pesonen, Tuuli Ranki, Lukasz Kyruk, Erkko Ylösmäki, Vincenzo Cerullo, Fabio Cerignoli, Biao Xi, Garret Guenther, Naichen Yu, Lincoln Muir, Leyna Zhao, Yama Abassi, Víctor Cervera-Carrascón, Mikko Siurala, João Santos, Riikka Havunen, Suvi Parviainen, Akseli Hemminki, Angus Dalgleish, Satvinder Mudan, Mark DeBenedette, Ana Plachco, Alicia Gamble, Elizabeth W. Grogan, John Krisko, Irina Tcherepanova, Charles Nicolette, Pooja Dhupkar, Ling Yu, Eugenie S. Kleinerman, Nancy Gordon, Italia Grenga, Lauren Lepone, Sofia Gameiro, Karin M. Knudson, Massimo Fantini, Kwong Tsang, James Hodge, Renee Donahue, Jeffrey Schlom, Elizabeth Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Cathie Foster, Alan Howell, Leslie Balch, Alyssa Knapp, John E. Leonard, Mark Paris, Terry Fisher, Siwen Hu-Lieskovan, Ernest Smith, Maurice Zauderer, William Fogler, Marilyn Franklin, Matt Thayer, Dan Saims, John L. Magnani, Jian Gong, Michael Gray, George Fromm, Suresh de Silva, Louise Giffin, Xin Xu, Jason Rose, Taylor H. Schreiber, Sofia R. Gameiro, Paul E. Clavijo, Clint T. Allen, James W. Hodge, Kwong Y. Tsang, Jane Grogan, Nicholas Manieri, Eugene Chiang, Patrick Caplazi, Mahesh Yadav, Patrick Hagner, Hsiling Chiu, Michelle Waldman, Anke Klippel, Anjan Thakurta, Michael Pourdehnad, Anita Gandhi, Ian Henrich, Laura Quick, Rob Young, Margaret Chou, Andrew Hotson, Stephen Willingham, Po Ho, Carmen Choy, Ginna Laport, Ian McCaffery, Richard Miller, Kimberly A. Tipton, Kenneth R. Wong, Victoria Singson, Chihunt Wong, Chanty Chan, Yuanhiu Huang, Shouchun Liu, Jennifer H. Richardson, W. Michael Kavanaugh, James West, Bryan A. Irving, Ritika Jaini, Matthew Loya, Charis Eng, Melissa L. Johnson, Alex A. Adjei, Mateusz Opyrchal, Suresh Ramalingam, Pasi A. Janne, George Dominguez, Dmitry Gabrilovich, Laura de Leon, Jeannette Hasapidis, Scott J. Diede, Peter Ordentlich, Scott Cruickshank, Michael L. Meyers, Matthew D. Hellmann, Pawel Kalinski, Amer Zureikat, Robert Edwards, Ravi Muthuswamy, Nataša Obermajer, Julie Urban, Lisa H. Butterfield, William Gooding, Herbert Zeh, David Bartlett, Olga Zubkova, Larissa Agapova, Marina Kapralova, Liudmila Krasovskaia, Armen Ovsepyan, Maxim Lykov, Artem Eremeev, Vladimir Bokovanov, Olga Grigoryeva, Andrey Karpov, Sergey Ruchko, Alexandr Shuster, Danny N. Khalil, Luis Felipe Campesato, Yanyun Li, Adam S. Lazorchak, Troy D. Patterson, Yueyun Ding, Pottayil Sasikumar, Naremaddepalli Sudarshan, Nagaraj Gowda, Raghuveer Ramachandra, Dodheri Samiulla, Sanjeev Giri, Rajesh Eswarappa, Murali Ramachandra, David Tuck, Timothy Wyant, Jasmin Leshem, Xiu-fen Liu, Tapan Bera, Masaki Terabe, Birgit Bossenmaier, Gerhard Niederfellner, Yoram Reiter, Ira Pastan, Leiming Xia, Yang Xia, Yangyang Hu, Yi Wang, Yangyi Bao, Fu Dai, Shiang Huang, Elaine Hurt, Robert E. Hollingsworth, Lawrence G. Lum, Alfred E. Chang, Max S. Wicha, Qiao Li, Thomas Mace, Neil Makhijani, Erin Talbert, Gregory Young, Denis Guttridge, Darwin Conwell, Gregory B. Lesinski, Rodney JM Macedo Gonzales, Austin P. Huffman, Ximi K. Wang, Ran Reshef, Andy MacKinnon, Jason Chen, Matt Gross, Gisele Marguier, Peter Shwonek, Natalija Sotirovska, Susanne Steggerda, Francesco Parlati, Amani Makkouk, Mark K. Bennett, Ethan Emberley, Tony Huang, Weiqun Li, Silinda Neou, Alison Pan, Jing Zhang, Winter Zhang, Netonia Marshall, Thomas U. Marron, Judith Agudo, Brian Brown, Joshua Brody, Christopher McQuinn, Matthew Farren, Hannah Komar, Reena Shakya, Thomas Ludwug, Y. Maurice Morillon, Scott A. Hammond, John W. Greiner, Pulak R. Nath, Anthony L. Schwartz, Dragan Maric, David D. Roberts, Aung Naing, Kyriakos P. Papadopoulos, Karen A. Autio, Deborah J. Wong, Manish Patel, Gerald Falchook, Shubham Pant, Patrick A. Ott, Melinda Whiteside, Amita Patnaik, John Mumm, Filip Janku, Ivan Chan, Todd Bauer, Rivka Colen, Peter VanVlasselaer, Gail L. Brown, Nizar M. Tannir, Martin Oft, Jeffrey Infante, Evan Lipson, Ajay Gopal, Sattva S. Neelapu, Philippe Armand, Stephen Spurgeon, John P. Leonard, Rachel E. Sanborn, Ignacio Melero, Thomas F. Gajewski, Matthew Maurer, Serena Perna, Andres A. Gutierrez, Raphael Clynes, Priyam Mitra, Satyendra Suryawanshi, Douglas Gladstone, Margaret K. Callahan, James Crooks, Sheila Brown, Audrey Gauthier, Marc Hillairet de Boisferon, Andrew MacDonald, Laura Rosa Brunet, William T. Rothwell, Peter Bell, James M. Wilson, Fumi Sato-Kaneko, Shiyin Yao, Shannon S. Zhang, Dennis A. Carson, Cristina Guiducci, Robert L. Coffman, Kazutaka Kitaura, Takaji Matsutani, Ryuji Suzuki, Tomoko Hayashi, Ezra E. W. Cohen, David Schaer, Yanxia Li, Julie Dobkin, Michael Amatulli, Gerald Hall, Thompson Doman, Jason Manro, Frank Charles Dorsey, Lillian Sams, Rikke Holmgaard, Krishnadatt Persaud, Dale Ludwig, David Surguladze, John S. Kauh, Ruslan Novosiadly, Michael Kalos, Kyla Driscoll, Hardev Pandha, Christy Ralph, Kevin Harrington, Brendan Curti, Wallace Akerley, Sumati Gupta, Alan Melcher, David Mansfield, David R. Kaufman, Emmett Schmidt, Mark Grose, Bronwyn Davies, Roberta Karpathy, Darren Shafren, Katerina Shamalov, Cyrille Cohen, Naveen Sharma, James Allison, Tala Shekarian, Sandrine Valsesia-Wittmann, Christophe Caux, Aurelien Marabelle, Brian M. Slomovitz, Kathleen M. Moore, Hagop Youssoufian, Marshall Posner, Poonam Tewary, Alan D. Brooks, Ya-Ming Xu, Kithsiri Wijeratne, Leslie A. A. Gunatilaka, Thomas J. Sayers, John P. Vasilakos, Tesha Alston, Simon Dovedi, James Elvecrog, Iwen Grigsby, Ronald Herbst, Karen Johnson, Craig Moeckly, Stefanie Mullins, Kristen Siebenaler, Julius SternJohn, Ashenafi Tilahun, Mark A. Tomai, Katharina Vogel, Eveline E. Vietsch, Anton Wellstein, Martin Wythes, Stefano Crosignani, Joseph Tumang, Shilpa Alekar, Patrick Bingham, Sandra Cauwenberghs, Jenny Chaplin, Deepak Dalvie, Sofie Denies, Coraline De Maeseneire, JunLi Feng, Kim Frederix, Samantha Greasley, Jie Guo, James Hardwick, Stephen Kaiser, Katti Jessen, Erick Kindt, Marie-Claire Letellier, Wenlin Li, Karen Maegley, Reece Marillier, Nichol Miller, Brion Murray, Romain Pirson, Julie Preillon, Virginie Rabolli, Chad Ray, Kevin Ryan, Stephanie Scales, Jay Srirangam, Jim Solowiej, Al Stewart, Nicole Streiner, Vince Torti, Konstantinos Tsaparikos, Xianxian Zheng, Gregory Driessens, Bruno Gomes, Manfred Kraus, Chunxiao Xu, Yanping Zhang, Giorgio Kradjian, Guozhong Qin, Jin Qi, Xiaomei Xu, Bo Marelli, Huakui Yu, Wilson Guzman, Rober Tighe, Rachel Salazar, Kin-Ming Lo, Jessie English, Laszlo Radvanyi, Yan Lan, Michael Postow, Yasin Senbabaoglu, Billel Gasmi, Hong Zhong, Cailian Liu, Daniel Hirschhorhn-Cymerman, Yuanyuan Zha, Gregory Malnassy, Noreen Fulton, Jae-Hyun Park, Wendy Stock, Yusuke Nakamura, Hongtao Liu, Xiaoming Ju, Rachelle Kosoff, Kimberly Ramos, Brandon Coder, Robert Petit, Michael Princiotta, Kyle Perry, Jun Zou, Ainhoa Arina, Christian Fernandez, Wenxin Zheng, Michael A. Beckett, Helena J. Mauceri, Yang-Xin Fu, Ralph R. Weichselbaum, Whitney Lewis, Yanyan Han, Yeting Wu, Chou Yang, Jing Huang, Dongyun Wu, Jin Li, Xiaoling Liang, Xiangjun Zhou, Jinlin Hou, Raffit Hassan, Thierry Jahan, Scott J. Antonia, Hedy L. Kindler, Evan W. Alley, Somayeh Honarmand, Weiqun Liu, Meredith L. Leong, Chan C. Whiting, Nitya Nair, Amanda Enstrom, Edward E. Lemmens, Takahiro Tsujikawa, Sushil Kumar, Lisa M. Coussens, Aimee L. Murphy, Dirk G. Brockstedt, Sven D. Koch, Martin Sebastian, Christian Weiss, Martin Früh, Miklos Pless, Richard Cathomas, Wolfgang Hilbe, Georg Pall, Thomas Wehler, Jürgen Alt, Helge Bischoff, Michael Geissler, Frank Griesinger, Jens Kollmeier, Alexandros Papachristofilou, Fatma Doener, Mariola Fotin-Mleczek, Madeleine Hipp, Henoch S. Hong, Karl-Josef Kallen, Ute Klinkhardt, Claudia Stosnach, Birgit Scheel, Andreas Schroeder, Tobias Seibel, Ulrike Gnad-Vogt, Alfred Zippelius, Ha-Ram Park, Yong-Oon Ahn, Tae Min Kim, Soyeon Kim, Seulki Kim, Yu Soo Lee, Bhumsuk Keam, Dong-Wan Kim, Dae Seog Heo, Shari Pilon-Thomas, Amy Weber, Jennifer Morse, Krithika Kodumudi, Hao Liu, John Mullinax, Amod A. Sarnaik, Luke Pike, Andrew Bang, Tracy Balboni, Allison Taylor, Alexander Spektor, Tyler Wilhite, Monica Krishnan, Daniel Cagney, Brian Alexander, Ayal Aizer, Elizabeth Buchbinder, Mark Awad, Leena Ghandi, Jonathan Schoenfeld, Elizabeth Lessey-Morillon, Lisa Ridnour, Neil H. Segal, Manish Sharma, Dung T. Le, Robert L. Ferris, Andrew D. Zelenetz, Ronald Levy, Izidore S. Lossos, Caron Jacobson, Radhakrishnan Ramchandren, John Godwin, A. Dimitrios Colevas, Roland Meier, Suba Krishnan, Xuemin Gu, Jaclyn Neely, John Timmerman, Claire I. Vanpouille-Box, Silvia C. Formenti, Sandra Demaria, Erik Wennerberg, Aranzazu Mediero, Bruce N. Cronstein, Michael P. Gustafson, AriCeli DiCostanzo, Courtney Wheatley, Chul-Ho Kim, Svetlana Bornschlegl, Dennis A. Gastineau, Bruce D. Johnson, Allan B. Dietz, Cameron MacDonald, Mark Bucsek, Guanxi Qiao, Bonnie Hylander, Elizabeth Repasky, William J. Turbitt, Yitong Xu, Andrea Mastro, Connie J. Rogers, Sita Withers, Ziming Wang, Lam T. Khuat, Cordelia Dunai, Bruce R. Blazar, Dan Longo, Robert Rebhun, Steven K. Grossenbacher, Arta Monjazeb, William J. Murphy, Scott Rowlinson, Giulia Agnello, Susan Alters, David Lowe, Nicole Scharping, Ashley V. Menk, Ryan Whetstone, Xue Zeng, Greg M. Delgoffe, Patricia M. Santos, Jian Shi, Greg Delgoffe, Misako Nagasaka, Ammar Sukari, Miranda Byrne-Steele, Wenjing Pan, Xiaohong Hou, Brittany Brown, Mary Eisenhower, Jian Han, Natalie Collins, Robert Manguso, Hans Pope, Yashaswi Shrestha, Jesse Boehm, W. Nicholas Haining, Kyle R. Cron, Ayelet Sivan, Keston Aquino-Michaels, Marco Orecchioni, Davide Bedognetti, Wouter Hendrickx, Claudia Fuoco, Filomena Spada, Francesco Sgarrella, Gianni Cesareni, Francesco Marincola, Kostas Kostarelos, Alberto Bianco, Lucia Delogu, Jessica Roelands, Sabri Boughorbel, Julie Decock, Scott Presnell, Ena Wang, Franco M. Marincola, Peter Kuppen, Michele Ceccarelli, Darawan Rinchai, Damien Chaussabel, Lance Miller, Andrew Nguyen, J. Zachary Sanborn, Charles Vaske, Shahrooz Rabizadeh, Kayvan Niazi, Steven Benz, Shashank Patel, Nicholas Restifo, James White, Sam Angiuoli, Mark Sausen, Sian Jones, Maria Sevdali, John Simmons, Victor Velculescu, Luis Diaz, Theresa Zhang, Jennifer S. Sims, Sunjay M. Barton, Angela Kadenhe-Chiweshe, Filemon Dela Cruz, Andrew T. Turk, Christopher F. Mazzeo, Andrew L. Kung, Jeffrey N. Bruce, Darrell J. Yamashiro, Eileen P. Connolly, Jason Baird, Marka Crittenden, David Friedman, Hong Xiao, Rom Leidner, Bryan Bell, Kristina Young, Michael Gough, Zhen Bian, Koby Kidder, Yuan Liu, Emily Curran, Xiufen Chen, Leticia P. Corrales, Justin Kline, Ethan G. Aguilar, Jennifer Guerriero, Alaba Sotayo, Holly Ponichtera, Alexandra Pourzia, Sara Schad, Ruben Carrasco, Suzan Lazo, Roderick Bronson, Anthony Letai, Richard S. Kornbluth, Sachin Gupta, James Termini, Elizabeth Guirado, Geoffrey W. Stone, Christina Meyer, Laura Helming, Nicholas Wilson, Robert Hofmeister, Natalie J. Neubert, Laure Tillé, David Barras, Charlotte Soneson, Petra Baumgaertner, Donata Rimoldi, David Gfeller, Mauro Delorenzi, Silvia A. Fuertes Marraco, Daniel E. Speiser, Tara S. Abraham, Bo Xiang, Michael S. Magee, Scott A. Waldman, Adam E. Snook, Wojciech Blogowski, Ewa Zuba-Surma, Marta Budkowska, Daria Salata, Barbara Dolegowska, Teresa Starzynska, Leo Chan, Srinivas Somanchi, Kelsey McCulley, Dean Lee, Nico Buettner, Feng Shi, Paisley T. Myers, Stuart Curbishley, Sarah A. Penny, Lora Steadman, David Millar, Ellen Speers, Nicola Ruth, Gabriel Wong, Robert Thimme, David Adams, Mark Cobbold, Remy Thomas, Mariam Al-Muftah, Michael KK Wong, Michael Morse, Joseph I. Clark, Howard L. Kaufman, Gregory A. Daniels, Hong Hua, Tharak Rao, Janice P. Dutcher, Kai Kang, Yogen Saunthararajah, Vamsidhar Velcheti, Vikas Kumar, Firoz Anwar, Amita Verma, Zinal Chheda, Gary Kohanbash, John Sidney, Kaori Okada, Shruti Shrivastav, Diego A. Carrera, Shuming Liu, Naznin Jahan, Sabine Mueller, Ian F. Pollack, Angel M. Carcaboso, Alessandro Sette, Yafei Hou, Hideho Okada, Jessica J. Field, Weiping Zeng, Vincent FS Shih, Che-Leung Law, Peter D. Senter, Shyra J. Gardai, Nicole M. Okeley, Jennifer G. Abelin, Abu Z. Saeed, Stacy A. Malaker, Jeffrey Shabanowitz, Stephen T. Ward, Donald F. Hunt, Pam Profusek, Laura Wood, Dale Shepard, Petros Grivas, Kerstin Kapp, Barbara Volz, Detlef Oswald, Burghardt Wittig, Manuel Schmidt, Julian P. Sefrin, Lars Hillringhaus, Valeria Lifke, Alexander Lifke, Anna Skaletskaya, Jose Ponte, Thomas Chittenden, Yulius Setiady, Eva Sivado, Vincent Thomas, Meddy El Alaoui, Sébastien Papot, Charles Dumontet, Mike Dyson, John McCafferty, Said El Alaoui, Praveen K. Bommareddy, Andrew Zloza, Frederick Kohlhapp, Ann W. Silk, Sachin Jhawar, Tomas Paneque, Jenna Newman, Pedro Beltran, Felicia Cao, Bang-Xing Hong, Tania Rodriguez-Cruz, Xiao-Tong Song, Stephen Gottschalk, Hugo Calderon, Sam Illingworth, Alice Brown, Kerry Fisher, Len Seymour, Brian Champion, Emma Eriksson, Jessica Wenthe, Ann-Charlotte Hellström, Gabriella Paul-Wetterberg, Angelica Loskog, Ioanna Milenova, Magnus Ståhle, Justyna Jarblad-Leja, Gustav Ullenhag, Anna Dimberg, Rafael Moreno, Ramon Alemany, Sharad Goyal, Ann Silk, Janice Mehnert, Nashat Gabrail, Jennifer Bryan, Daniel Medina, Leah Mitchell, Kader Yagiz, Fernando Lopez, Daniel Mendoza, Anthony Munday, Harry Gruber, Douglas Jolly, Steven Fuhrmann, Sasa Radoja, Wei Tan, Aldo Pourchet, Alan Frey, Ian Mohr, Matthew Mulvey, Robert H. I. Andtbacka, Merrick Ross, Sanjiv Agarwala, Kenneth Grossmann, Matthew Taylor, John Vetto, Rogerio Neves, Adil Daud, Hung Khong, Stephanie M. Meek, Richard Ungerleider, Scott Welden, Maki Tanaka, Matthew Williams, Sigrun Hallmeyer, Bernard Fox, Zipei Feng, Christopher Paustian, Carlo Bifulco, Sadia Zafar, Otto Hemminki, Simona Bramante, Lotta Vassilev, Hongjie Wang, Andre Lieber, Silvio Hemmi, Tanja de Gruijl, Anna Kanerva, Tameem Ansari, Srividya Sundararaman, Diana Roen, Paul Lehmann, Anja C. Bloom, Lewis H. Bender, Ian B. Walters, Jay A. Berzofsky, Fanny Chapelin, Eric T. Ahrens, Jeff DeFalco, Michael Harbell, Amy Manning-Bog, Alexander Scholz, Danhui Zhang, Gilson Baia, Yann Chong Tan, Jeremy Sokolove, Dongkyoon Kim, Kevin Williamson, Xiaomu Chen, Jillian Colrain, Gregg Espiritu Santo, Ngan Nguyen, Wayne Volkmuth, Norman Greenberg, William Robinson, Daniel Emerling, Charles G. Drake, Daniel P. Petrylak, Emmanuel S. Antonarakis, Adam S. Kibel, Nancy N. Chang, Tuyen Vu, Dwayne Campogan, Heather Haynes, James B. Trager, Nadeem A. Sheikh, David I. Quinn, Peter Kirk, Murali Addepalli, Thomas Chang, Ping Zhang, Marina Konakova, Katsunobu Hagihara, Steven Pai, Laurie VanderVeen, Palakshi Obalapur, Peiwen Kuo, Phi Quach, Lawrence Fong, Deborah H. Charych, Jonathan Zalevsky, John L. Langowski, Yolanda Kirksey, Ravi Nutakki, Shalini Kolarkar, Rhoneil Pena, Ute Hoch, Stephen K. Doberstein, John Cha, Zach Mallon, Myra Perez, Amanda McDaniel, Snjezana Anand, Darrin Uecker, Richard Nuccitelli, Eva Wieckowski, Ravikumar Muthuswamy, Roshni Ravindranathan, Ariana N. Renrick, Menaka Thounaojam, Portia Thomas, Samuel Pellom, Anil Shanker, Duafalia Dudimah, Alan Brooks, Yu-Lin Su, Tomasz Adamus, Qifang Zhang, Sergey Nechaev, Marcin Kortylewski, Spencer Wei, Clark Anderson, Chad Tang, Jonathan Schoenhals, Efrosini Tsouko, John Heymach, Patricia de Groot, Joe Chang, Kenneth R. Hess, Adi Diab, Padmanee Sharma, David Hong, James Welsh, Andrea J. Parsons, Jardin Leleux, Stephane Ascarateil, Marie Eve Koziol, Dina Bai, Peihong Dai, Weiyi Wang, Ning Yang, Stewart Shuman, Liang Deng, Patrick Dillon, Gina Petroni, David Brenin, Kim Bullock, Walter Olson, Mark E. Smolkin, Kelly Smith, Carmel Nail, Craig L. Slingluff, Meenu Sharma, Faisal Fa’ak, Louise Janssen, Hiep Khong, Zhilan Xiao, Yared Hailemichael, Manisha Singh, Christina Vianden, Willem W. Overwijk, Andrea Facciabene, Pierini Stefano, Fang Chongyung, Stavros Rafail, Michael Nielsen, Peter Vanderslice, Darren G. Woodside, Robert V. Market, Ronald J. Biediger, Upendra K. Marathi, Kevin Hollevoet, Nick Geukens, Paul Declerck, Nathalie Joly, Laura McIntosh, Eustache Paramithiotis, Magnus Rizell, Malin Sternby, Bengt Andersson, Alex Karlsson-Parra, Rui Kuai, Lukasz Ochyl, Anna Schwendeman, James Moon, Weiwen Deng, Thomas E. Hudson, Bill Hanson, Chris S. Rae, Joel Burrill, Justin Skoble, George Katibah, Michele deVries, Peter Lauer, Thomas W. Dubensky, Xin Chen, Li Zhou, Xiubao Ren, Charu Aggarwal, Drishty Mangrolia, Roger Cohen, Gregory Weinstein, Matthew Morrow, Joshua Bauml, Kim Kraynyak, Jean Boyer, Jian Yan, Jessica Lee, Laurent Humeau, Sandra Oyola, Susan Duff, David Weiner, Zane Yang, Mark Bagarazzi, Douglas G. McNeel, Jens Eickhoff, Robert Jeraj, Mary Jane Staab, Jane Straus, Brian Rekoske, Glenn Liu, Marit Melssen, William Grosh, Nikole Varhegyi, Nadejda Galeassi, Donna H. Deacon, Elizabeth Gaughan, Maurizio Ghisoli, Minal Barve, Robert Mennel, Gladice Wallraven, Luisa Manning, Neil Senzer, John Nemunaitis, Masahiro Ogasawara, Shuichi Ota, Kaitlin M. Peace, Diane F. Hale, Timothy J. Vreeland, Doreen O. Jackson, John S. Berry, Alfred F. Trappey, Garth S. Herbert, Guy T. Clifton, Mark O. Hardin, Anne Toms, Na Qiao, Jennifer Litton, George E. Peoples, Elizabeth A. Mittendorf, Lila Ghamsari, Emilio Flano, Judy Jacques, Biao Liu, Jonathan Havel, Vladimir Makarov, Timothy A. Chan, Jessica B. Flechtner, John Facciponte, Stefano Ugel, Francesco De Sanctis, George Coukos, Sébastien Paris, Agnes Pottier, Laurent Levy, Bo Lu, Federica Cappuccini, Emily Pollock, Richard Bryant, Freddie Hamdy, Adrian Hill, Irina Redchenko, Hussein Sultan, Takumi Kumai, Valentyna Fesenkova, Esteban Celis, Ingrid Fernando, Claudia Palena, Justin M. David, Elizabeth Gabitzsch, Frank Jones, James L. Gulley, Mireia Uribe Herranz, Hiroshi Wada, Atsushi Shimizu, Toshihiro Osada, Satoshi Fukaya, Eiji Sasaki, Milad Abolhalaj, David Askmyr, Kristina Lundberg, Ann-Sofie Albrekt, Lennart Greiff, Malin Lindstedt, Dallas B. Flies, Tomoe Higuchi, Wojciech Ornatowski, Jaryse Harris, Sarah F. Adams, Todd Aguilera, Marjan Rafat, Laura Castellini, Hussein Shehade, Mihalis Kariolis, Dadi Jang, Rie vonEbyen, Edward Graves, Lesley Ellies, Erinn Rankin, Albert Koong, Amato Giaccia, Reham Ajina, Shangzi Wang, Jill Smith, Mariaelena Pierobon, Sandra Jablonski, Emanuel Petricoin, Louis M. Weiner, Lorcan Sherry, John Waller, Mark Anderson, Alison Bigley, Chantale Bernatchez, Cara Haymaker, Harriet Kluger, Michael Tetzlaff, Natalie Jackson, Ivan Gergel, Mary Tagliaferri, Patrick Hwu, Mario Snzol, Michael Hurwitz, Theresa Barberi, Allison Martin, Rahul Suresh, David Barakat, Sarah Harris-Bookman, Charles Drake, Alan Friedman, Sara Berkey, Stephanie Downs-Canner, Robert P. Edwards, Tyler Curiel, Kunle Odunsi, Tullia C. Bruno, Brandon Moore, Olivia Squalls, Peggy Ebner, Katherine Waugh, John Mitchell, Wilbur Franklin, Daniel Merrick, Martin McCarter, Brent Palmer, Jeffrey Kern, Dario Vignali, Jill Slansky, Anissa S. H. Chan, Xiaohong Qiu, Kathryn Fraser, Adria Jonas, Nadine Ottoson, Keith Gordon, Takashi O. Kangas, Steven Leonardo, Kathleen Ertelt, Richard Walsh, Mark Uhlik, Jeremy Graff, Nandita Bose, Ravi Gupta, Nitin Mandloi, Kiran Paul, Ashwini Patil, Rekha Sathian, Aparna Mohan, Malini Manoharan, Amitabha Chaudhuri, Yu Chen, Jing Lin, Yun-bin Ye, Chun-wei Xu, Gang Chen, Zeng-qing Guo, Andrey Komarov, Alex Chenchik, Michael Makhanov, Costa Frangou, Yi Zheng, Carla Coltharp, Darryn Unfricht, Ryan Dilworth, Leticia Fridman, Linying Liu, Milind Rajopadhye, Peter Miller, Fernando Concha-Benavente, Julie Bauman, Sumita Trivedi, Raghvendra Srivastava, James Ohr, Dwight Heron, Uma Duvvuri, Seungwon Kim, Heather Torrey, Toshi Mera, Yoshiaki Okubo, Eva Vanamee, Rosemary Foster, Denise Faustman, Edward Stack, Daisuke Izaki, Kristen Beck, Dan Tong Jia, Paul Armenta, Ashley White-Stern, Douglas Marks, Bret Taback, Basil Horst, Laura Hix Glickman, David B. Kanne, Kelsey S. Gauthier, Anthony L. Desbien, Brian Francica, Justin L. Leong, Leonard Sung, Ken Metchette, Shailaja Kasibhatla, Anne Marie Pferdekamper, Lianxing Zheng, Charles Cho, Yan Feng, Jeffery M. McKenna, John Tallarico, Steven Bender, Chudi Ndubaku, Sarah M. McWhirter, Elena Gonzalez Gugel, Charles J. M. Bell, Adiel Munk, Luciana Muniz, Nina Bhardwaj, Fei Zhao, Kathy Evans, Christine Xiao, Alisha Holtzhausen, Brent A. Hanks, Nathalie Scholler, Catherine Yin, Pien Van der Meijs, Andrew M. Prantner, Cecile M. Krejsa, Leia Smith, Brian Johnson, Daniel Branstetter, Paul L. Stein, Juan C. Jaen, Joanne BL Tan, Ada Chen, Timothy Park, Jay P. Powers, Holly Sexton, Guifen Xu, Steve W. Young, Ulrike Schindler, Wentao Deng, David John Klinke, Hannah M. Komar, Gregory Serpa, Omar Elnaggar, Philip Hart, Carl Schmidt, Mary Dillhoff, Ming Jin, Michael C. Ostrowski, Madhuri Koti, Katrina Au, Nichole Peterson, Peter Truesdell, Gillian Reid-Schachter, Charles Graham, Andrew Craig, Julie-Ann Francis, Beatrix Kotlan, Timea Balatoni, Emil Farkas, Laszlo Toth, Mihaly Ujhelyi, Akos Savolt, Zoltan Doleschall, Szabolcs Horvath, Klara Eles, Judit Olasz, Orsolya Csuka, Miklos Kasler, Gabriella Liszkay, Eytan Barnea, Collin Blakely, Patrick Flynn, Reid Goodman, Raphael Bueno, David Sugarbaker, David Jablons, V. Courtney Broaddus, Brian West, Paul R. Kunk, Joseph M. Obeid, Kevin Winters, Patcharin Pramoonjago, Edward B. Stelow, Todd W. Bauer, Osama E. Rahma, Adam Lamble, Yoko Kosaka, Fei Huang, Kate A. Saser, Homer Adams, Christina E. Tognon, Ted Laderas, Shannon McWeeney, Marc Loriaux, Jeffery W. Tyner, Brian J. Druker, Evan F. Lind, Zhuqing Liu, Shanhong Lu, Lawrence P. Kane, Gulidanna Shayan, Julia Femel, Ryan Lane, Jamie Booth, Amanda W. Lund, Anthony Rodriguez, Victor H. Engelhard, Alessandra Metelli, Bill X. Wu, Caroline W. Fugle, Rachidi Saleh, Shaoli Sun, Jennifer Wu, Bei Liu, Zihai Li, Zachary S. Morris, Emily I. Guy, Clinton Heinze, Jasdeep Kler, Monica M. Gressett, Lauryn R. Werner, Stephen D. Gillies, Alan J. Korman, Hans Loibner, Jacquelyn A. Hank, Alexander L. Rakhmilevich, Paul M. Harari, Paul M. Sondel, Erica Huelsmann, Joseph Broucek, Dorothee Brech, Tobias Straub, Martin Irmler, Johannes Beckers, Florian Buettner, Elke Schaeffeler, Matthias Schwab, Elfriede Noessner, Alison Wolfreys, Andre Da Costa, John Silva, Andrea Crosby, Ludovicus Staelens, Graham Craggs, Annick Cauvin, Sean Mason, Alison M. Paterson, Andrew C. Lake, Caroline M. Armet, Rachel W. O’Connor, Jonathan A. Hill, Emmanuel Normant, Ammar Adam, Detlev M. Biniszkiewicz, Scott C. Chappel, Vito J. Palombella, Pamela M. Holland, Annette Becker, Manmohan R. Leleti, Eric Newcomb, Joanne B. L. Tan, Suthee Rapisuwon, Arash Radfar, Kellie Gardner, Geoffrey Gibney, Michael Atkins, Keith R. Rennier, Robert Crowder, Ping Wang, Russell K. Pachynski, Rosa M. Santana Carrero, Sarai Rivas, Figen Beceren-Braun, Scott Anthony, Kimberly S. Schluns, Deepali Sawant, Maria Chikina, Hiroshi Yano, Creg Workman, Elise Salerno, Ileana Mauldin, Donna Deacon, Sofia Shea, Joel Pinczewski, Thomas Gajewski, Stefani Spranger, Brendan Horton, Akiko Suzuki, Pamela Leland, Bharat H. Joshi, Raj K. Puri, Randy F. Sweis, Riyue Bao, Jason Luke, Marie-Nicole Theodoraki, Frances-Mary Mogundo, Haejung Won, Dayson Moreira, Chan Gao, Xingli Zhao, Priyanka Duttagupta, Jeremy Jones, Massimo D’Apuzzo, and Sumanta Pal
- Subjects
0301 basic medicine ,Pharmacology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Cancer ,Immunotherapy ,medicine.disease ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Family medicine ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business - Abstract
O1 IL-15 primes an mTOR-regulated gene-expression program to prolong anti-tumor capacity of human natural killer cells #### Andreas Lundqvist1, Vincent van Hoef1, Xiaonan Zhang1, Erik Wennerberg2, Julie Lorent1, Kristina Witt1, Laia Masvidal Sanz1, Shuo Liang1, Shannon Murray3, Ola Larsson1
- Published
- 2016
- Full Text
- View/download PDF
149. Vasopressin stimulates the proliferation and differentiation of red blood cell precursors and improves recovery from anemia
- Author
-
Heather Rogers, Lynn Vitale-Cross, Kenn Holmbeck, Krisztián Németh, Jerome H. Pagani, Balázs Mayer, Miklos Krepuska, W. Scott Young, Eva Mezey, Constance Tom Noguchi, Ildiko Szalayova, Vamsee D. Myneni, Dragan Maric, Naoya Uchida, Arne Hansen, Michael J. Nemeth, Jeffrey Hong, Károly Markó, John F. Tisdale, Sharon Key, Michael J. Brownstein, Heon-Jin Lee, Soumyadeep Dey, Ian Chow, Matthew M. Hsieh, and Ian McClain-Caldwell
- Subjects
0301 basic medicine ,Vasopressin ,medicine.medical_specialty ,endocrine system ,Receptors, Vasopressin ,Erythrocytes ,Vasopressins ,030204 cardiovascular system & hematology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Humans ,Peripheral blood cell ,Progenitor cell ,Cell Proliferation ,business.industry ,Anemia ,Cell Differentiation ,General Medicine ,Rats ,Red blood cell ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Erythropoietin ,Erythropoiesis ,business ,hormones, hormone substitutes, and hormone antagonists ,Antidiuretic ,medicine.drug - Abstract
Arginine vasopressin (AVP) made by hypothalamic neurons is released into the circulation to stimulate water resorption by the kidneys and restore water balance after blood loss. Patients who lack this antidiuretic hormone suffer from central diabetes insipidus. We observed that many of these patients were anemic and asked whether AVP might play a role in red blood cell (RBC) production. We found that all three AVP receptors are expressed in human and mouse hematopoietic stem and progenitor cells. The AVPR1B appears to play the most important role in regulating erythropoiesis in both human and mouse cells. AVP increases phosphorylation of signal transducer and activator of transcription 5, as erythropoietin (EPO) does. After sublethal irradiation, AVP-deficient Brattleboro rats showed delayed recovery of RBC numbers compared to control rats. In mouse models of anemia (induced by bleeding, irradiation, or increased destruction of circulating RBCs), AVP increased the number of circulating RBCs independently of EPO. In these models, AVP appears to jump-start peripheral blood cell replenishment until EPO can take over. We suggest that specific AVPR1B agonists might be used to induce fast RBC production after bleeding, drug toxicity, or chemotherapy.
- Published
- 2016
150. The polyglutamine-expanded androgen receptor responsible for spinal and bulbar muscular atrophy inhibits the APC/CCdh1 ubiquitin ligase complex
- Author
-
Yuhong Liu, Diane E. Merry, Laura C. Bott, Florian A. Salomons, Kenneth H. Fischbeck, Nico P. Dantuma, and Dragan Maric
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,Neurite ,Bulbo-Spinal Atrophy, X-Linked ,PC12 Cells ,Anaphase-Promoting Complex-Cyclosome ,Article ,APC/C activator protein CDH1 ,03 medical and health sciences ,0302 clinical medicine ,Antigens, CD ,Neurites ,medicine ,Animals ,Genetics ,Multidisciplinary ,biology ,Cell Cycle ,Cell cycle ,Cadherins ,medicine.disease ,Rats ,Ubiquitin ligase ,Cell biology ,Androgen receptor ,Spinal and bulbar muscular atrophy ,030104 developmental biology ,Receptors, Androgen ,Ubiquitin ligase complex ,Mutation ,Proteolysis ,biology.protein ,Carrier Proteins ,030217 neurology & neurosurgery - Abstract
Polyglutamine expansion in the androgen receptor (AR) causes spinal and bulbar muscular atrophy (SBMA), an X-linked neuromuscular disease that is fully manifest only in males. It has been suggested that proteins with expanded polyglutamine tracts impair ubiquitin-dependent proteolysis due to their propensity to aggregate, but recent studies indicate that the overall activity of the ubiquitin-proteasome system is preserved in SBMA models. Here we report that AR selectively interferes with the function of the ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C), which, together with its substrate adaptor Cdh1, is critical for cell cycle arrest and neuronal architecture. We show that both wild-type and mutant AR physically interact with the APC/CCdh1 complex in a ligand-dependent fashion without being targeted for proteasomal degradation. Inhibition of APC/CCdh1 by mutant but not wild-type AR in PC12 cells results in enhanced neurite outgrowth which is typically followed by rapid neurite retraction and mitotic entry. Our data indicate a role of AR in neuronal differentiation through regulation of APC/CCdh1 and suggest abnormal cell cycle reactivation as a pathogenic mechanism in SBMA.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.