46 results on '"Kerur N"'
Search Results
2. Genetic characterization of Indian peste des petits ruminants virus (PPRV) by sequencing and phylogenetic analysis of fusion protein and nucleoprotein gene segments
- Author
-
Kerur, N., Jhala, M.K., and Joshi, C.G.
- Published
- 2008
- Full Text
- View/download PDF
3. Analysis of Structure, Conduct and Performance of Vegetable Seed Market: Case Study in Belagavi District of Karnataka, IndiaCucumber (Cucumis sativus L.)
- Author
-
Raj, Stephan, primary, Naik, A. D., additional, Naik, B. K., additional, and Kerur, N. M., additional
- Published
- 2020
- Full Text
- View/download PDF
4. Brand Preferences and Factors Influencing for Purchase of Branded Ready-To- Eat Food Products in Belgaum City of Karnataka State, India
- Author
-
Chaitra, G. B., primary and Kerur, N. M., additional
- Published
- 2020
- Full Text
- View/download PDF
5. To assess the brand preference in hybrid cotton seeds in North Karnataka
- Author
-
Sajjan, Srividyarani S., primary and Kerur, N. M., additional
- Published
- 2018
- Full Text
- View/download PDF
6. To identify and analyze the marketing channels for hybrid cotton seeds in North Karnataka
- Author
-
Sajjan, Srividyarani S., primary and Kerur, N. M., additional
- Published
- 2018
- Full Text
- View/download PDF
7. Pesticide purchasing behaviour of farmers in Dharwad district
- Author
-
Hosurkar, Mohasin A., primary and Kerur, N. M., additional
- Published
- 2018
- Full Text
- View/download PDF
8. DICER1/Alu RNA dysmetabolism induces Caspase-8-mediated cell death in age-related macular degeneration
- Author
-
Kim Y, Tarallo V, Kerur N, Yasuma T, Gelfand BD, Bastos-Carvalho A, Hirano Y, Yasuma R, Mizutani T, Fowler BJ, Li S, Kaneko H, Bogdanovich S, Ambati BK, Hinton DR, Hauswirth WW, Hakem R, Wright C, and Ambati J.
- Subjects
sense organs ,eye diseases - Abstract
Geographic atrophy, an advanced form of age-related macular degeneration (AMD) characterized by death of the retinal pigmented epithelium (RPE), causes untreatable blindness in millions worldwide. The RPE of human eyes with geographic atrophy accumulates toxic Alu RNA in response to a deficit in the enzyme DICER1, which in turn leads to activation of the NLRP3 inflammasome and elaboration of IL-18. Despite these recent insights, it is still unclear how RPE cells die during the course of the disease. In this study, we implicate the involvement of Caspase-8 as a critical mediator of RPE degeneration. Here we show that DICER1 deficiency, Alu RNA accumulation, and IL-18 up-regulation lead to RPE cell death via activation of Caspase-8 through a Fas ligand-dependent mechanism. Coupled with our observation of increased Caspase-8 expression in the RPE of human eyes with geographic atrophy, our findings provide a rationale for targeting this apoptotic pathway in this disease.
- Published
- 2014
9. HMGB1 is a potent inducer of choroidal angiogenesis through a MyD88-dependent manner
- Author
-
KIM, Y, primary, KERUR, N, additional, LI, S, additional, and AMBATI, J, additional
- Published
- 2013
- Full Text
- View/download PDF
10. Angiogenic role of glycerol in laser-induced choroidal neovascularization
- Author
-
YASUMA, R, primary, YASUMA, T, additional, KIM, Y, additional, GELFAND, B, additional, FOWLER, B, additional, KERUR, N, additional, BASTOS DE CARVALHO, A, additional, MIZUTANI, T, additional, and AMBATI, J, additional
- Published
- 2013
- Full Text
- View/download PDF
11. Intercellular trafficking of Alu RNAs causes geographic atrophy expansion in AMD
- Author
-
BASTOS CARVALHO, A, primary, KERUR, N, additional, KIM, Y, additional, FOWLER, B, additional, GELFAND, B, additional, and AMBATI, J, additional
- Published
- 2013
- Full Text
- View/download PDF
12. Alu retrotransposon quantification in the retina and plasma: Mechanism‐based risk assessment in age‐related macular degeneration
- Author
-
FOWLER, B, primary, KERUR, N, additional, GELFAND, B, additional, BASTOS‐CARVALHO, A, additional, and AMBATI, J, additional
- Published
- 2013
- Full Text
- View/download PDF
13. COX-2/PGE2: molecular ambassadors of Kaposi's sarcoma-associated herpes virus oncoprotein-v-FLIP
- Author
-
Sharma-Walia, N, primary, Patel, K, additional, Chandran, K, additional, Marginean, A, additional, Bottero, V, additional, Kerur, N, additional, and Paul, A G, additional
- Published
- 2012
- Full Text
- View/download PDF
14. Trend, Export Competitiveness and Direction of Trade of Indian Cigarette.
- Author
-
Patted, Nataraj, Kerur, N. M., Hebbar, Ashwini, Naik, A. D., Mokashi, Prakash, and Kumar, S.
- Subjects
- *
CIGARETTE industry , *TOBACCO , *SMOKING , *AGRICULTURAL laborers - Abstract
Tobacco is one of the important commercial crops of India and being so, it is vital in the Indian economy. It provides employment directly and indirectly to 36 millions of people. Although the cultivation of tobacco is restricted to 0.3 per cent of the total cultivated area, it provides employment to a large number of people on the one hand, it makes significant contribution to national exchequer by way of excise revenue and foreign exchange earnings to the another side. As a labour intensive crop, growing, harvesting and processing of tobacco represent the means of livelihood of a large number of agricultural laborers. Demand for cigarette has been consistently increasing. Results of the analysis depicted that trend with respect to export for cigarette except Russia and UAE all other countries are having a positive slope with increasing trend. In case of nominal protection coefficient United States of America was the most competitive market. Current major stable markets for cigarette are Russian Republic and USA. In future Malaysia and UAE will be the major importing countries. [ABSTRACT FROM AUTHOR]
- Published
- 2013
15. Dynamics of Indian FCV Tobacco in International Market.
- Author
-
Patted, Nataraj, Kerur, N. M., Kumar, S., Mokashi, Prakash, and Hebbar, Ashwini
- Subjects
- *
TOBACCO , *FOREIGN exchange , *INTERNATIONAL markets , *TOBACCO curing , *INTERNATIONAL trade , *TOBACCO farmers , *AGRICULTURAL laborers - Abstract
Tobacco is one of the important commercial crops, provides employment directly and indirectly to 36 millions of people. Although the cultivation of tobacco is restricted to 0.3 per cent of the total cultivated area but it makes significant contribution to national exchequer by way of excise revenue and foreign exchange earnings to the another side. Tobacco is a labour-intensive crop in India. Growing, harvesting and processing of tobacco represent the means of livelihood of a large number of agricultural laborers. Demand for flue cured virginia has been consistently increasing. Results of the analysis depicted that trend with respect to export for flue cured virginia are having a positive slope with increasing trend. In case of nominal protection coefficient Belgium was the most competitive market. Current major stable markets for cigarette are Russian Republic and Other minor importing countries. [ABSTRACT FROM AUTHOR]
- Published
- 2013
16. Human IgG1 antibodies suppress angiogenesis in a target-independent manner
- Author
-
Bogdanovich S, Kim Y, Mizutani T, Yasuma R, Tudisco L, Cicatiello V, Bastos-Carvalho A, Kerur N, Hirano Y, Jz, Baffi, Valeria Tarallo, Li S, Yasuma T, Arpitha P, Bj, Fowler, Cb, Wright, Apicella I, Greco A, Brunetti A, and Ruvo M
17. Intravenous immune globulin suppresses angiogenesis in mice and humans
- Author
-
Ivana Apicella, Sasha Bogdanovich, Bradley D. Gelfand, Arturo Brunetti, J. Sjef Verbeek, Charles B. Wright, Shengjian Li, Younghee Kim, Laura Tudisco, Tetsuhiro Yasuma, Jeanette H. W. Leusen, Yoshio Hirano, Ana Bastos-Carvalho, Jayakrishna Ambati, Takeshi Mizutani, Sandro De Falco, Valeria Cicatiello, Ingrid E. Lundberg, Benjamin J. Fowler, Balamurali K. Ambati, Adelaide Greco, Valeria Tarallo, Nagaraj Kerur, Sevim Barbasso Helmers, Ondrej Viklicky, Reo Yasuma, Yasuma, R, Cicatiello, V, Mizutani, T, Tudisco, L, Kim, Y, Tarallo, V, Bogdanovich, S, Hirano, Y, Kerur, N, Li, S, Yasuma, T, Fowler, Bj, Wright, Cb, Apicella, I, Greco, Adelaide, Brunetti, Arturo, Ambati, Bk, Helmers, Sb, Lundberg, Ie, Viklicky, O, Leusen, Jh, Verbeek, J, Gelfand, Bd, Bastos Carvalho, A, De Falco, S, and Ambati, J.
- Subjects
0301 basic medicine ,Genetically modified mouse ,Cancer Research ,Angiogenesis ,Article ,03 medical and health sciences ,angiogenesis ,hemic and lymphatic diseases ,Genetics ,medicine ,Journal Article ,Receptor ,biology ,immuneglobuline, angioinhibition, mice ,business.industry ,medicine.disease ,immune globulin ,Fragment crystallizable region ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Polyclonal antibodies ,Corneal neovascularization ,Immunology ,biology.protein ,Antibody ,business ,Blood vessel - Abstract
Human intravenous immune globulin (IVIg), a purified IgG fraction composed of ~60% IgG1 and obtained from the pooled plasma of thousands of donors, is clinically used for a wide range of diseases. The biological actions of IVIg are incompletely understood and have been attributed both to the polyclonal antibodies therein and also to their IgG (IgG) Fc regions. Recently, we demonstrated that multiple therapeutic human IgG1 antibodies suppress angiogenesis in a target-independent manner via FcγRI, a high-affinity receptor for IgG1. Here we show that IVIg possesses similar anti-angiogenic activity and inhibited blood vessel growth in five different mouse models of prevalent human diseases, namely, neovascular age-related macular degeneration, corneal neovascularization, colorectal cancer, fibrosarcoma and peripheral arterial ischemic disease. Angioinhibition was mediated by the Fc region of IVIg, required FcγRI and had similar potency in transgenic mice expressing human FcγRs. Finally, IVIg therapy administered to humans for the treatment of inflammatory or autoimmune diseases reduced kidney and muscle blood vessel densities. These data place IVIg, an agent approved by the US Food and Drug Administration, as a novel angioinhibitory drug in doses that are currently administered in the clinical setting. In addition, they raise the possibility of an unintended effect of IVIg on blood vessels.
- Published
- 2016
- Full Text
- View/download PDF
18. Human IgG1 antibodies suppress angiogenesis in a target-independent manner
- Author
-
Tetsuhiro Yasuma, Hiroko Terasaki, Mike Clark, Valeria Cicatiello, Laura Tudisco, Yuichiro Ogura, Wallace Y. Langdon, Judit Z. Baffi, Miho Nozaki, Pierre Bruhns, Younghee Kim, Parthasarathy Arpitha, Sasha Bogdanovich, Nagaraj Kerur, Takeshi Mizutani, Yoshio Hirano, Bradley D. Gelfand, Benjamin J. Fowler, Balamurali K. Ambati, Ivana Apicella, Kathryn L. Armour, Shengjian Li, Sandro De Falco, Ryo Ijima, Hiroki Kaneko, Ana Bastos-Carvalho, Valeria Tarallo, Jeanette H. W. Leusen, Charles B. Wright, Jayakrishna Ambati, J. Sjef Verbeek, Arturo Brunetti, Reo Yasuma, Annamaria Sandomenico, Adelaide Greco, Menotti Ruvo, Bogdanovich, S, Kim, Y, Mizutani, T, Yasuma, R, Tudisco, L, Cicatiello, V, Bastos Carvalho, A, Kerur, N, Hirano, Y, Baffi, Jz, Tarallo, V, Li, S, Yasuma, T, Arpitha, P, Fowler, Bj, Wright, Cb, Apicella, I, Greco, Adelaide, Brunetti, Arturo, Ruvo, M, Sandomenico, A, Nozaki, M, Ijima, R, Kaneko, H, Ogura, Y, Terasaki, H, Ambati, Bk, Leusen, Jh, Langdon, Wy, Clark, Mr, Armour, Kl, Bruhns, P, Verbeek, J, Gelfand, Bd, De Falco, S, Ambati, J., University of Kentucky, Nagoya City University [Nagoya, Japan], Institute of Genetics and Biophysics - 'Adriano Buzzati-Traverso' [Naples, Italy] ( IGB-CNR), BIO-KER (Multimedica Group) [Naples], University of Naples Federico II, CEINGE - Biotecnologie Avanzate, CNR – Istituto di Biostrutture e Bioimmagini, University of Utah School of Medicine [Salt Lake City], Salt Lake City Veterans Affairs Health Care System, University Medical Center [Utrecht], The University of Western Australia (UWA), University of Cambridge [UK] (CAM), Anticorps en thérapie et pathologie - Antibodies in Therapy and Pathology, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Leiden University Medical Center (LUMC), IRCCS Multimedica, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), JA was supported by NIH grants DP1GM114862, R01EY018350, R01EY018836, R01EY020672, R01EY022238, R01EY024068, R21EY019778 and RC1EY020442, Doris Duke Distinguished Clinical Scientist Award, Burroughs Wellcome Fund Clinical Scientist Award in Translational Research, Ellison Medical Foundation Senior Scholar in Aging Award, Foundation Fighting Blindness Individual Investigator Research Award, Carl Marshall Reeves Foundation, Harrington Discovery Institute Scholar-Innovator Award, John Templeton Foundation, Dr E. Vernon Smith and Eloise C. Smith Macular Degeneration Endowed Chair, and Research to Prevent Blindness departmental unrestricted grant, SDF by Associazione Italiana Ricerca sul Cancro (AIRC) grant no. IG11420 and Italian Ministry for Scientific Research, projects PON01_02342 and PON01_01434, MR and AS by Italian Ministry for Scientific Research, grants FIRB MERIT N° RBNE08NKH7_003 and PON01_01602, PON01_02342. JZB by NIH K08EY021521 and University of Kentucky Physician Scientist Award, BJF and SB by NIH T32HL091812 and UL1RR033173, YH by Alcon Research Award, AB-C by the Program for Advanced Medical Education (sponsored by Fundação Calouste Gulbenkian, Fundação Champalimaud, Ministério da Saúde and Fundação para a Ciência e Tecnologia, Portugal) and Bayer Global Ophthalmology Research Award, YH by Alcon Japan Research award, NK by Beckman Initiative for Macular Research and NIH K99/R00EY024336, TY by Fight for Sight Postdoctoral Award, CBW by International Retinal Research Foundation, BDG by American Heart Association and International Retinal Research Foundation, BKA by NIH R01EY017182 and R01EY017950, VA Merit Award, and Department of Defense., University of Kentucky (UK), University of Naples Federico II = Università degli studi di Napoli Federico II, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Universiteit Leiden
- Subjects
0301 basic medicine ,Cancer Research ,Bevacizumab ,medicine.drug_class ,Angiogenesis ,Population ,Pharmacology ,Monoclonal antibody ,Ofatumumab ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Genetics ,Journal Article ,angiogenesis, mice, antibodies ,Medicine ,education ,Gene knockdown ,education.field_of_study ,business.industry ,3. Good health ,Blockade ,Vascular endothelial growth factor A ,030104 developmental biology ,chemistry ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,business ,medicine.drug - Abstract
Aberrant angiogenesis is implicated in diseases affecting nearly 10% of the world’s population. The most widely used anti-angiogenic drug is bevacizumab, a humanized IgG1 monoclonal antibody that targets human VEGFA. Although bevacizumab does not recognize mouse Vegfa, it inhibits angiogenesis in mice. Here we show bevacizumab suppressed angiogenesis in three mouse models not via Vegfa blockade but rather Fc-mediated signaling through FcγRI (CD64) and c-Cbl, impairing macrophage migration. Other approved humanized or human IgG1 antibodies without mouse targets (adalimumab, alemtuzumab, ofatumumab, omalizumab, palivizumab and tocilizumab), mouse IgG2a, and overexpression of human IgG1-Fc or mouse IgG2a-Fc, also inhibited angiogenesis in wild-type and FcγR humanized mice. This anti-angiogenic effect was abolished by Fcgr1 ablation or knockdown, Fc cleavage, IgG-Fc inhibition, disruption of Fc-FcγR interaction, or elimination of FcRγ-initated signaling. Furthermore, bevacizumab’s Fc region potentiated its anti-angiogenic activity in humanized VEGFA mice. Finally, mice deficient in FcγRI exhibited increased developmental and pathological angiogenesis. These findings reveal an unexpected anti-angiogenic function for FcγRI and a potentially concerning off-target effect of hIgG1 therapies.
- Published
- 2016
- Full Text
- View/download PDF
19. Therapeutic targeting of telomerase ameliorates experimental choroidal neovascularization.
- Author
-
Kumar A, Nagasaka Y, Jayananthan V, Zidan A, Heisler-Taylor T, Ambati J, Tamiya S, and Kerur N
- Subjects
- Animals, Mice, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor A genetics, Mice, Inbred C57BL, Aminobenzoates pharmacology, RNA genetics, RNA metabolism, Oligonucleotides, Antisense pharmacology, Naphthalenes, Telomerase antagonists & inhibitors, Telomerase genetics, Telomerase metabolism, Choroidal Neovascularization pathology, Choroidal Neovascularization metabolism, Choroidal Neovascularization drug therapy, Disease Models, Animal
- Abstract
Choroidal neovascularization (CNV) is the principal driver of blindness in neovascular age-related macular degeneration (nvAMD). Increased activity of telomerase, has been associated with endothelial cell proliferation, survival, migration, and invasion in the context of tumor angiogenesis. Expanding on this knowledge, we investigated the role of telomerase in the development of CNV in mouse model. We observed increased gene expression and activity of telomerase in mouse CNV. Genetic deficiency of the telomerase components, telomerase reverse transcriptase (Tert) and telomerase RNA component (Terc) suppressed laser-induced CNV in mice. Similarly, a small molecule inhibitor of TERT (BIBR 1532), and antisense oligonucleotides (ASOs) targeting Tert and Terc reduced CNV growth. Bone marrow chimera studies suggested that telomerase activity in non-bone marrow-derived cells is crucial for the development of CNV. Comparison of BIBR 1532 with VEGF neutralizing therapeutic strategy in mouse revealed a comparable level of angiosuppressive activity. However, when BIBR and anti-VEGF antibodies were administered as a combination at sub-therapeutic doses, a statistically significant suppression of CNV was observed. These findings underscore the potential benefits of combining sub-therapeutic doses of BIBR and anti-VEGF antibodies for developing newer therapeutic strategies for NV-AMD. Telomerase inhibition with BIBR 1532 suppressed induction of multiple cytokines and growth factors critical for neovascularization. In conclusion, our study identifies telomerase as a promising therapeutic target for treating neovascular disease of the eye and thus provides a proof of principle for further exploration of telomerase inhibition as a novel treatment strategy for nvAMD., Competing Interests: Declaration of competing interest N.K. is named as an inventor on a patent application on non-canonical signaling activity of cGAMP filed by the University of Virginia. N.K. and J.A., are named as inventors on patent applications on macular degeneration filed by the University of Virginia or the University of Kentucky. J.A., is a co-founder of DiceRx, iVeena Holdings, iVeena Delivery Systems and Inflammasome Therapeutics, and, unrelated to this work, he has been a consultant for Abbvie/Allergan, Boehringer-Ingelheim, Janssen, Olix Pharmaceuticals, Retinal Solutions, and Saksin LifeSciences. The other authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. DDX17 is an essential mediator of sterile NLRC4 inflammasome activation by retrotransposon RNAs.
- Author
-
Wang SB, Narendran S, Hirahara S, Varshney A, Pereira F, Apicella I, Ambati M, Ambati VL, Yerramothu P, Ambati K, Nagasaka Y, Argyle D, Huang P, Baker KL, Marion KM, Gupta K, Liu B, Hinton DR, Canna SW, Sallam T, Sadda SR, Kerur N, Gelfand BD, and Ambati J
- Subjects
- Animals, Apoptosis Regulatory Proteins deficiency, Calcium-Binding Proteins deficiency, Cells, Cultured, Mice, Mice, Inbred C57BL, Mice, Knockout, Apoptosis Regulatory Proteins immunology, Calcium-Binding Proteins immunology, DEAD-box RNA Helicases immunology, Inflammasomes immunology, RNA immunology, Retroelements immunology
- Abstract
Detection of microbial products by multiprotein complexes known as inflammasomes is pivotal to host defense against pathogens. Nucleotide-binding domain leucine-rich repeat (NLR) CARD domain containing 4 (NLRC4) forms an inflammasome in response to bacterial products; this requires their detection by NLR family apoptosis inhibitory proteins (NAIPs), with which NLRC4 physically associates. However, the mechanisms underlying sterile NLRC4 inflammasome activation, which is implicated in chronic noninfectious diseases, remain unknown. Here, we report that endogenous short interspersed nuclear element (SINE) RNAs, which promote atrophic macular degeneration (AMD) and systemic lupus erythematosus (SLE), induce NLRC4 inflammasome activation independent of NAIPs. We identify DDX17, a DExD/H box RNA helicase, as the sensor of SINE RNAs that licenses assembly of an inflammasome comprising NLRC4, NLR pyrin domain–containing protein 3, and apoptosis-associated speck-like protein–containing CARD and induces caspase-1 activation and cytokine release. Inhibiting DDX17-mediated NLRC4 inflammasome activation decreased interleukin-18 release in peripheral blood mononuclear cells of patients with SLE and prevented retinal degeneration in an animal model of AMD. Our findings uncover a previously unrecognized noncanonical NLRC4 inflammasome activated by endogenous retrotransposons and provide potential therapeutic targets for SINE RNA–driven diseases.
- Published
- 2021
- Full Text
- View/download PDF
21. A non-canonical, interferon-independent signaling activity of cGAMP triggers DNA damage response signaling.
- Author
-
Banerjee D, Langberg K, Abbas S, Odermatt E, Yerramothu P, Volaric M, Reidenbach MA, Krentz KJ, Rubinstein CD, Brautigan DL, Abbas T, Gelfand BD, Ambati J, and Kerur N
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins metabolism, Crassostrea genetics, Crassostrea metabolism, G1 Phase Cell Cycle Checkpoints, Humans, Immunity, Innate, Interferon Type I metabolism, Membrane Proteins metabolism, Mice, Nucleotidyltransferases metabolism, Phosphorylation, Poly ADP Ribosylation, Protein Serine-Threonine Kinases metabolism, Recombinational DNA Repair, Sea Anemones genetics, Sea Anemones metabolism, DNA Damage, Nucleotides, Cyclic metabolism, Signal Transduction
- Abstract
Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), produced by cyclic GMP-AMP synthase (cGAS), stimulates the production of type I interferons (IFN). Here we show that cGAMP activates DNA damage response (DDR) signaling independently of its canonical IFN pathways. Loss of cGAS dampens DDR signaling induced by genotoxic insults. Mechanistically, cGAS activates DDR in a STING-TBK1-dependent manner, wherein TBK1 stimulates the autophosphorylation of the DDR kinase ATM, with the consequent activation of the CHK2-p53-p21 signal transduction pathway and the induction of G1 cell cycle arrest. Despite its stimulatory activity on ATM, cGAMP suppresses homology-directed repair (HDR) through the inhibition of polyADP-ribosylation (PARylation), in which cGAMP reduces cellular levels of NAD
+ ; meanwhile, restoring NAD+ levels abrogates cGAMP-mediated suppression of PARylation and HDR. Finally, we show that cGAMP also activates DDR signaling in invertebrate species lacking IFN (Crassostrea virginica and Nematostella vectensis), suggesting that the genome surveillance mechanism of cGAS predates metazoan interferon-based immunity., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
22. Alu complementary DNA is enriched in atrophic macular degeneration and triggers retinal pigmented epithelium toxicity via cytosolic innate immunity.
- Author
-
Fukuda S, Narendran S, Varshney A, Nagasaka Y, Wang SB, Ambati K, Apicella I, Pereira F, Fowler BJ, Yasuma T, Hirahara S, Yasuma R, Huang P, Yerramothu P, Makin RD, Wang M, Baker KL, Marion KM, Huang X, Baghdasaryan E, Ambati M, Ambati VL, Banerjee D, Bonilha VL, Tolstonog GV, Held U, Ogura Y, Terasaki H, Oshika T, Bhattarai D, Kim KB, Feldman SH, Aguirre JI, Hinton DR, Kerur N, Sadda SR, Schumann GG, Gelfand BD, and Ambati J
- Abstract
Long interspersed nuclear element-1 (L1)–mediated reverse transcription (RT) of Alu RNA into cytoplasmic Alu complementary DNA (cDNA) has been implicated in retinal pigmented epithelium (RPE) degeneration. The mechanism of Alu cDNA–induced cytotoxicity and its relevance to human disease are unknown. Here we report that Alu cDNA is highly enriched in the RPE of human eyes with geographic atrophy, an untreatable form of age-related macular degeneration. We demonstrate that the DNA sensor cGAS engages Alu cDNA to induce cytosolic mitochondrial DNA escape, which amplifies cGAS activation, triggering RPE degeneration via the inflammasome. The L1-extinct rice rat was resistant to Alu RNA–induced Alu cDNA synthesis and RPE degeneration, which were enabled upon L1-RT overexpression. Nucleoside RT inhibitors (NRTIs), which inhibit both L1-RT and inflammasome activity, and NRTI derivatives (Kamuvudines) that inhibit inflammasome, but not RT, both block Alu cDNA toxicity, identifying inflammasome activation as the terminal effector of RPE degeneration.
- Published
- 2021
- Full Text
- View/download PDF
23. Cytoplasmic synthesis of endogenous Alu complementary DNA via reverse transcription and implications in age-related macular degeneration.
- Author
-
Fukuda S, Varshney A, Fowler BJ, Wang SB, Narendran S, Ambati K, Yasuma T, Magagnoli J, Leung H, Hirahara S, Nagasaka Y, Yasuma R, Apicella I, Pereira F, Makin RD, Magner E, Liu X, Sun J, Wang M, Baker K, Marion KM, Huang X, Baghdasaryan E, Ambati M, Ambati VL, Pandey A, Pandya L, Cummings T, Banerjee D, Huang P, Yerramothu P, Tolstonog GV, Held U, Erwin JA, Paquola ACM, Herdy JR, Ogura Y, Terasaki H, Oshika T, Darwish S, Singh RK, Mozaffari S, Bhattarai D, Kim KB, Hardin JW, Bennett CL, Hinton DR, Hanson TE, Röver C, Parang K, Kerur N, Liu J, Werner BC, Sutton SS, Sadda SR, Schumann GG, Gelfand BD, Gage FH, and Ambati J
- Subjects
- Animals, Cytoplasm genetics, DNA, Complementary genetics, Epithelium metabolism, Epithelium pathology, Humans, Macular Degeneration pathology, Retinal Pigments biosynthesis, Retroelements genetics, Reverse Transcription genetics, Alu Elements genetics, Long Interspersed Nucleotide Elements genetics, Macular Degeneration genetics, Retinal Pigments metabolism
- Abstract
Alu retroelements propagate via retrotransposition by hijacking long interspersed nuclear element-1 (L1) reverse transcriptase (RT) and endonuclease activities. Reverse transcription of Alu RNA into complementary DNA (cDNA) is presumed to occur exclusively in the nucleus at the genomic integration site. Whether Alu cDNA is synthesized independently of genomic integration is unknown. Alu RNA promotes retinal pigmented epithelium (RPE) death in geographic atrophy, an untreatable type of age-related macular degeneration. We report that Alu RNA-induced RPE degeneration is mediated via cytoplasmic L1-reverse-transcribed Alu cDNA independently of retrotransposition. Alu RNA did not induce cDNA production or RPE degeneration in L1-inhibited animals or human cells. Alu reverse transcription can be initiated in the cytoplasm via self-priming of Alu RNA. In four health insurance databases, use of nucleoside RT inhibitors was associated with reduced risk of developing atrophic macular degeneration (pooled adjusted hazard ratio, 0.616; 95% confidence interval, 0.493-0.770), thus identifying inhibitors of this Alu replication cycle shunt as potential therapies for a major cause of blindness., Competing Interests: Competing interest statement: J.A. is a co-founder of iVeena Holdings, iVeena Delivery Systems, and Inflammasome Therapeutics, and has been a consultant for Allergan, Biogen, Boehringer-Ingelheim, Immunovant, Janssen, Olix Pharmaceuticals, Retinal Solutions, and Saksin LifeSciences unrelated to this work. J.A., B.D.G., B.J.F., S.N., K.A., S.-b.W., I.A., M.A., F.P., N.K., and S.F. are named as inventors on patent applications on macular degeneration filed by the University of Virginia or the University of Kentucky. J.W.H. has received consulting fees from Celgene Corporation unrelated to this work. S.S.S. has received research grants from Boehringer Ingelheim, Gilead Sciences, Portola Pharmaceuticals, and United Therapeutics unrelated to this work. J.A. and B.D.G. are co-founders of DiceRx.
- Published
- 2021
- Full Text
- View/download PDF
24. Emerging Nano-Formulations and Nanomedicines Applications for Ocular Drug Delivery.
- Author
-
Khiev D, Mohamed ZA, Vichare R, Paulson R, Bhatia S, Mohapatra S, Lobo GP, Valapala M, Kerur N, Passaglia CL, Mohapatra SS, and Biswal MR
- Abstract
Ocular diseases can deteriorate vision to the point of blindness and thus can have a major impact on the daily life of an individual. Conventional therapies are unable to provide absolute therapy for all ocular diseases due to the several limitations during drug delivery across the blood-retinal barrier, making it a major clinical challenge. With recent developments, the vast number of publications undergird the need for nanotechnology-based drug delivery systems in treating ocular diseases. The tool of nanotechnology provides several essential advantages, including sustained drug release and specific tissue targeting. Additionally, comprehensive in vitro and in vivo studies have suggested a better uptake of nanoparticles across ocular barriers. Nanoparticles can overcome the blood-retinal barrier and consequently increase ocular penetration and improve the bioavailability of the drug. In this review, we aim to summarize the development of organic and inorganic nanoparticles for ophthalmic applications. We highlight the potential nanoformulations in clinical trials as well as the products that have become a commercial reality.
- Published
- 2021
- Full Text
- View/download PDF
25. Repurposing anti-inflammasome NRTIs for improving insulin sensitivity and reducing type 2 diabetes development.
- Author
-
Ambati J, Magagnoli J, Leung H, Wang SB, Andrews CA, Fu D, Pandey A, Sahu S, Narendran S, Hirahara S, Fukuda S, Sun J, Pandya L, Ambati M, Pereira F, Varshney A, Cummings T, Hardin JW, Edun B, Bennett CL, Ambati K, Fowler BJ, Kerur N, Röver C, Leitinger N, Werner BC, Stein JD, Sutton SS, and Gelfand BD
- Subjects
- Adipocytes metabolism, Animals, Cell Survival, DEAD-box RNA Helicases metabolism, Diabetes Mellitus, Type 2 prevention & control, Diet, High-Fat adverse effects, HIV-1 drug effects, Hepatitis B, Humans, Male, Mice, Mice, Inbred C57BL, Muscle Cells metabolism, Ribonuclease III metabolism, Diabetes Mellitus, Type 2 drug therapy, Drug Repositioning, Inflammasomes drug effects, Insulin Resistance, Reverse Transcriptase Inhibitors pharmacology
- Abstract
Innate immune signaling through the NLRP3 inflammasome is activated by multiple diabetes-related stressors, but whether targeting the inflammasome is beneficial for diabetes is still unclear. Nucleoside reverse-transcriptase inhibitors (NRTI), drugs approved to treat HIV-1 and hepatitis B infections, also block inflammasome activation. Here, we show, by analyzing five health insurance databases, that the adjusted risk of incident diabetes is 33% lower in patients with NRTI exposure among 128,861 patients with HIV-1 or hepatitis B (adjusted hazard ratio for NRTI exposure, 0.673; 95% confidence interval, 0.638 to 0.710; P < 0.0001; 95% prediction interval, 0.618 to 0.734). Meanwhile, an NRTI, lamivudine, improves insulin sensitivity and reduces inflammasome activation in diabetic and insulin resistance-induced human cells, as well as in mice fed with high-fat chow; mechanistically, inflammasome-activating short interspersed nuclear element (SINE) transcripts are elevated, whereas SINE-catabolizing DICER1 is reduced, in diabetic cells and mice. These data suggest the possibility of repurposing an approved class of drugs for prevention of diabetes.
- Published
- 2020
- Full Text
- View/download PDF
26. Voluntary Exercise Suppresses Choroidal Neovascularization in Mice.
- Author
-
Makin RD, Argyle D, Hirahara S, Nagasaka Y, Zhang M, Yan Z, Kerur N, Ambati J, and Gelfand BD
- Subjects
- Animals, Female, Male, Mice, Mice, Inbred C57BL, Choroidal Neovascularization prevention & control, Motor Activity
- Abstract
Purpose: To determine the effect of voluntary exercise on choroidal neovascularization (CNV) in mice., Methods: Age-matched wild-type C57BL/6J mice were housed in cages equipped with or without running wheels. After four weeks of voluntary running or sedentariness, mice were subjected to laser injury to induce CNV. After surgical recovery, mice were placed back in cages with or without exercise wheels for seven days. CNV lesion volumes were measured by confocal microscopy. The effect of wheel running only in the seven days after injury was also evaluated. Macrophage abundance and cytokine expression were quantified., Results: In the first study, exercise-trained mice exhibited a 45% reduction in CNV volume compared to sedentary mice. In the replication study, a 32% reduction in CNV volume in exercise-trained mice was observed (P = 0.029). Combining these two studies, voluntary exercise was found to reduce CNV by 41% (P = 0.0005). Exercise-trained male and female mice had similar CNV volumes (P = 0.99). The daily running distance did not correlate with CNV lesion size. Exercise only after the laser injury without a preconditioning period did not reduce CNV size (P = 0.41). CNV lesions of exercise-trained mice also exhibited significantly lower F4/80+ macrophage staining and Vegfa and Ccl2 mRNA expression., Conclusions: These findings provide the first experimental evidence that voluntary exercise improves CNV outcomes. These studies indicate that exercise before laser treatment is required to improve CNV outcomes.
- Published
- 2020
- Full Text
- View/download PDF
27. Chronic Dicer1 deficiency promotes atrophic and neovascular outer retinal pathologies in mice.
- Author
-
Wright CB, Uehara H, Kim Y, Yasuma T, Yasuma R, Hirahara S, Makin RD, Apicella I, Pereira F, Nagasaka Y, Narendran S, Fukuda S, Albuquerque R, Fowler BJ, Bastos-Carvalho A, Georgel P, Hatada I, Chang B, Kerur N, Ambati BK, Ambati J, and Gelfand BD
- Subjects
- Animals, Choroidal Neovascularization genetics, Choroidal Neovascularization metabolism, Choroidal Neovascularization pathology, Choroidal Neovascularization physiopathology, DEAD-box RNA Helicases genetics, Humans, Macular Degeneration genetics, Macular Degeneration pathology, Macular Degeneration physiopathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Retinal Degeneration genetics, Retinal Degeneration metabolism, Retinal Degeneration pathology, Retinal Degeneration physiopathology, Retinal Neovascularization genetics, Retinal Neovascularization metabolism, Retinal Neovascularization parasitology, Retinal Neovascularization physiopathology, Retinal Pigment Epithelium metabolism, Retinal Pigment Epithelium pathology, Ribonuclease III genetics, DEAD-box RNA Helicases metabolism, Macular Degeneration metabolism, Retinal Pigment Epithelium blood supply, Ribonuclease III metabolism
- Abstract
Degeneration of the retinal pigmented epithelium (RPE) and aberrant blood vessel growth in the eye are advanced-stage processes in blinding diseases such as age-related macular degeneration (AMD), which affect hundreds of millions of people worldwide. Loss of the RNase DICER1, an essential factor in micro-RNA biogenesis, is implicated in RPE atrophy. However, the functional implications of DICER1 loss in choroidal and retinal neovascularization are unknown. Here, we report that two independent hypomorphic mouse strains, as well as a separate model of postnatal RPE-specific DICER1 ablation, all presented with spontaneous RPE degeneration and choroidal and retinal neovascularization. DICER1 hypomorphic mice lacking critical inflammasome components or the innate immune adaptor MyD88 developed less severe RPE atrophy and pathological neovascularization. DICER1 abundance was also reduced in retinas of the JR5558 mouse model of spontaneous choroidal neovascularization. Finally, adenoassociated vector-mediated gene delivery of a truncated DICER1 variant (OptiDicer) reduced spontaneous choroidal neovascularization in JR5558 mice. Collectively, these findings significantly expand the repertoire of DICER1 in preserving retinal homeostasis by preventing both RPE degeneration and pathological neovascularization., Competing Interests: Competing interest statement: J.A. is a co-founder of iVeena Holdings, iVeena Delivery Systems, and Inflammasome Therapeutics, and has been a consultant for Allergan, Biogen, Boehringer-Ingelheim, Immunovant, Janssen, Olix Pharmaceuticals, Retinal Solutions, and Saksin LifeSciences unrelated to this work. B.K.A. is a co-founder of iVeena Holdings, iVeena Delivery Systems, and Inflammasome Therapeutics, and has been a consultant to Alcon, Genentech, and Johnson & Johnson unrelated to this work. H.U., S.F., B.J.F., N.K., B.K.A., J.A., and B.D.G. are named as inventors on patent applications related to the intellectual property described in this manuscript that have been filed by the University of Virginia, the University of Kentucky, or the University of Utah., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
28. RF/6A Chorioretinal Cells Do Not Display Key Endothelial Phenotypes.
- Author
-
Makin RD, Apicella I, Nagasaka Y, Kaneko H, Turner SD, Kerur N, Ambati J, and Gelfand BD
- Subjects
- Animals, Biomarkers, Blotting, Western, Cell Line, E-Selectin genetics, Endothelial Cells metabolism, Gene Expression Profiling, Genetic Markers genetics, Humans, Immunohistochemistry, Macaca mulatta, Mice, Mice, Inbred C57BL, Microscopy, Fluorescence, Phenotype, Real-Time Polymerase Chain Reaction, Transcriptome genetics, Choroid blood supply, Endothelial Cells cytology, Retinal Vessels physiology
- Abstract
Purpose: The misuse of inauthentic cell lines is widely recognized as a major threat to the integrity of biomedical science. Whereas the majority of efforts to address this have focused on DNA profiling, we sought to anatomically, transcriptionally, and functionally authenticate the RF/6A chorioretinal cell line, which is widely used as an endothelial cell line to model retinal and choroidal angiogenesis., Methods: Multiple vials of RF/6A cells obtained from different commercial distributors were studied to validate their genetic, transcriptomic, anatomic, and functional fidelity to bona fide endothelial cells., Results: Transcriptomic profiles of RF/6A cells obtained either de novo or from a public data repository did not correspond to endothelial gene expression signatures. Expression of established endothelial markers were very low or undetectable in RF/6A compared to primary human endothelial cells. Importantly, RF/6A cells also did not display functional characteristics of endothelial cells such as uptake of acetylated LDL, expression of E-selectin in response to TNF-α exposure, alignment in the direction of shear stress, and AKT and ERK1/2 phosphorylation following VEGFA stimulation., Conclusions: Multiple independent sources of RF/6A do not exhibit key endothelial cell phenotypes. Therefore, these cells appear unsuitable as surrogates for choroidal or retinal endothelial cells. Further, cell line authentication methods should extend beyond genomic profiling to include anatomic, transcriptional, and functional assessments.
- Published
- 2018
- Full Text
- View/download PDF
29. cGAS drives noncanonical-inflammasome activation in age-related macular degeneration.
- Author
-
Kerur N, Fukuda S, Banerjee D, Kim Y, Fu D, Apicella I, Varshney A, Yasuma R, Fowler BJ, Baghdasaryan E, Marion KM, Huang X, Yasuma T, Hirano Y, Serbulea V, Ambati M, Ambati VL, Kajiwara Y, Ambati K, Hirahara S, Bastos-Carvalho A, Ogura Y, Terasaki H, Oshika T, Kim KB, Hinton DR, Leitinger N, Cambier JC, Buxbaum JD, Kenney MC, Jazwinski SM, Nagai H, Hara I, West AP, Fitzgerald KA, Sadda SR, Gelfand BD, and Ambati J
- Subjects
- Animals, DEAD-box RNA Helicases genetics, Humans, Interferon Type I metabolism, Mice, Retinal Pigment Epithelium metabolism, Ribonuclease III genetics, Signal Transduction, Geographic Atrophy enzymology, Inflammasomes metabolism, Nucleotidyltransferases metabolism
- Abstract
Geographic atrophy is a blinding form of age-related macular degeneration characterized by retinal pigmented epithelium (RPE) death; the RPE also exhibits DICER1 deficiency, resultant accumulation of endogenous Alu-retroelement RNA, and NLRP3-inflammasome activation. How the inflammasome is activated in this untreatable disease is largely unknown. Here we demonstrate that RPE degeneration in human-cell-culture and mouse models is driven by a noncanonical-inflammasome pathway that activates caspase-4 (caspase-11 in mice) and caspase-1, and requires cyclic GMP-AMP synthase (cGAS)-dependent interferon-β production and gasdermin D-dependent interleukin-18 secretion. Decreased DICER1 levels or Alu-RNA accumulation triggers cytosolic escape of mitochondrial DNA, which engages cGAS. Moreover, caspase-4, gasdermin D, interferon-β, and cGAS levels were elevated in the RPE in human eyes with geographic atrophy. Collectively, these data highlight an unexpected role of cGAS in responding to mobile-element transcripts, reveal cGAS-driven interferon signaling as a conduit for mitochondrial-damage-induced inflammasome activation, expand the immune-sensing repertoire of cGAS and caspase-4 to noninfectious human disease, and identify new potential targets for treatment of a major cause of blindness.
- Published
- 2018
- Full Text
- View/download PDF
30. NLRP3 inflammasome activation by mycobacterial ESAT-6 and dsRNA in intraocular tuberculosis.
- Author
-
Basu S, Fowler BJ, Kerur N, Arnvig KB, and Rao NA
- Subjects
- Animals, Antigens, Bacterial genetics, Bacterial Proteins genetics, Caspase 1 genetics, Caspase 1 immunology, Host-Parasite Interactions, Humans, Inflammasomes genetics, Mice, Mice, Inbred C57BL, Mycobacterium tuberculosis genetics, NLR Family, Pyrin Domain-Containing 3 Protein genetics, RNA, Bacterial genetics, RNA, Double-Stranded genetics, Retinal Pigment Epithelium immunology, Retinal Pigment Epithelium microbiology, Tuberculosis, Ocular genetics, Tuberculosis, Ocular microbiology, Antigens, Bacterial immunology, Bacterial Proteins immunology, Inflammasomes immunology, Mycobacterium tuberculosis immunology, NLR Family, Pyrin Domain-Containing 3 Protein immunology, RNA, Bacterial immunology, RNA, Double-Stranded immunology, Tuberculosis, Ocular immunology
- Abstract
The molecular basis of intraocular tuberculosis (TB) is not well understood. In this study, we investigated the role of two constituents of viable Mycobacterium tuberculosis - Early Secreted Antigenic Target-6 (ESAT-6), and mycobacterial RNA- in inflammasome activation in the retinal pigment epithelium (RPE), a key site of inflammation in intraocular TB. We found that ESAT-6 induced caspase-1 activation and inflammasome priming in mouse RPE cells, substantially more in wild-type than in Tlr2/3/4/7/9
-/- , Myd88-/- or Nlrp3-/- RPE cells. Sub-retinal ESAT-6 injection resulted in greater RPE degeneration in wild-type than in Nlrp3-/- mice. In human ocular TB tissue sections, NLRP3 staining was noted in retina as well as RPE. Mycobacterial RNA, specifically its double stranded component, also induced caspase-1 activation, and the double stranded RNA was immunolocalized to human ocular TB sections. Our observations suggest that inflammasome activation in RPE by viable M. tuberculosis could potentially contribute to human intraocular TB., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
31. Human IgG1 antibodies suppress angiogenesis in a target-independent manner.
- Author
-
Bogdanovich S, Kim Y, Mizutani T, Yasuma R, Tudisco L, Cicatiello V, Bastos-Carvalho A, Kerur N, Hirano Y, Baffi JZ, Tarallo V, Li S, Yasuma T, Arpitha P, Fowler BJ, Wright CB, Apicella I, Greco A, Brunetti A, Ruvo M, Sandomenico A, Nozaki M, Ijima R, Kaneko H, Ogura Y, Terasaki H, Ambati BK, Leusen JH, Langdon WY, Clark MR, Armour KL, Bruhns P, Verbeek JS, Gelfand BD, De Falco S, and Ambati J
- Abstract
Aberrant angiogenesis is implicated in diseases affecting nearly 10% of the world's population. The most widely used anti-angiogenic drug is bevacizumab, a humanized IgG1 monoclonal antibody that targets human VEGFA. Although bevacizumab does not recognize mouse Vegfa, it inhibits angiogenesis in mice. Here we show bevacizumab suppressed angiogenesis in three mouse models not via Vegfa blockade but rather Fc-mediated signaling through FcγRI (CD64) and c-Cbl, impairing macrophage migration. Other approved humanized or human IgG1 antibodies without mouse targets (adalimumab, alemtuzumab, ofatumumab, omalizumab, palivizumab and tocilizumab), mouse IgG2a, and overexpression of human IgG1-Fc or mouse IgG2a-Fc, also inhibited angiogenesis in wild-type and FcγR humanized mice. This anti-angiogenic effect was abolished by Fcgr1 ablation or knockdown, Fc cleavage, IgG-Fc inhibition, disruption of Fc-FcγR interaction, or elimination of FcRγ-initated signaling. Furthermore, bevacizumab's Fc region potentiated its anti-angiogenic activity in humanized VEGFA mice. Finally, mice deficient in FcγRI exhibited increased developmental and pathological angiogenesis. These findings reveal an unexpected anti-angiogenic function for FcγRI and a potentially concerning off-target effect of hIgG1 therapies.
- Published
- 2016
- Full Text
- View/download PDF
32. Intravenous immune globulin suppresses angiogenesis in mice and humans.
- Author
-
Yasuma R, Cicatiello V, Mizutani T, Tudisco L, Kim Y, Tarallo V, Bogdanovich S, Hirano Y, Kerur N, Li S, Yasuma T, Fowler BJ, Wright CB, Apicella I, Greco A, Brunetti A, Ambati BK, Helmers SB, Lundberg IE, Viklicky O, Leusen JH, Verbeek JS, Gelfand BD, Bastos-Carvalho A, De Falco S, and Ambati J
- Abstract
Human intravenous immune globulin (IVIg), a purified IgG fraction composed of ~ 60% IgG1 and obtained from the pooled plasma of thousands of donors, is clinically used for a wide range of diseases. The biological actions of IVIg are incompletely understood and have been attributed both to the polyclonal antibodies therein and also to their IgG (IgG) Fc regions. Recently, we demonstrated that multiple therapeutic human IgG1 antibodies suppress angiogenesis in a target-independent manner via FcγRI, a high-affinity receptor for IgG1. Here we show that IVIg possesses similar anti-angiogenic activity and inhibited blood vessel growth in five different mouse models of prevalent human diseases, namely, neovascular age-related macular degeneration, corneal neovascularization, colorectal cancer, fibrosarcoma and peripheral arterial ischemic disease. Angioinhibition was mediated by the Fc region of IVIg, required FcγRI and had similar potency in transgenic mice expressing human FcγRs. Finally, IVIg therapy administered to humans for the treatment of inflammatory or autoimmune diseases reduced kidney and muscle blood vessel densities. These data place IVIg, an agent approved by the US Food and Drug Administration, as a novel angioinhibitory drug in doses that are currently administered in the clinical setting. In addition, they raise the possibility of an unintended effect of IVIg on blood vessels.
- Published
- 2016
- Full Text
- View/download PDF
33. Iron Toxicity in the Retina Requires Alu RNA and the NLRP3 Inflammasome.
- Author
-
Gelfand BD, Wright CB, Kim Y, Yasuma T, Yasuma R, Li S, Fowler BJ, Bastos-Carvalho A, Kerur N, Uittenbogaard A, Han YS, Lou D, Kleinman ME, McDonald WH, Núñez G, Georgel P, Dunaief JL, and Ambati J
- Subjects
- Animals, Carrier Proteins genetics, Caspase 1 genetics, Caspase 1 metabolism, DEAD-box RNA Helicases genetics, DEAD-box RNA Helicases metabolism, Inflammasomes metabolism, Iron pharmacology, Mice, Mice, Inbred C57BL, NLR Family, Pyrin Domain-Containing 3 Protein, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Retinal Pigment Epithelium drug effects, Ribonuclease III genetics, Ribonuclease III metabolism, Alu Elements, Carrier Proteins metabolism, Iron toxicity, Retinal Pigment Epithelium metabolism
- Abstract
Excess iron induces tissue damage and is implicated in age-related macular degeneration (AMD). Iron toxicity is widely attributed to hydroxyl radical formation through Fenton's reaction. We report that excess iron, but not other Fenton catalytic metals, induces activation of the NLRP3 inflammasome, a pathway also implicated in AMD. Additionally, iron-induced degeneration of the retinal pigmented epithelium (RPE) is suppressed in mice lacking inflammasome components caspase-1/11 or Nlrp3 or by inhibition of caspase-1. Iron overload increases abundance of RNAs transcribed from short interspersed nuclear elements (SINEs): Alu RNAs and the rodent equivalent B1 and B2 RNAs, which are inflammasome agonists. Targeting Alu or B2 RNA prevents iron-induced inflammasome activation and RPE degeneration. Iron-induced SINE RNA accumulation is due to suppression of DICER1 via sequestration of the co-factor poly(C)-binding protein 2 (PCBP2). These findings reveal an unexpected mechanism of iron toxicity, with implications for AMD and neurodegenerative diseases associated with excess iron., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
34. IL-18 is not therapeutic for neovascular age-related macular degeneration.
- Author
-
Hirano Y, Yasuma T, Mizutani T, Fowler BJ, Tarallo V, Yasuma R, Kim Y, Bastos-Carvalho A, Kerur N, Gelfand BD, Bogdanovich S, He S, Zhang X, Nozaki M, Ijima R, Kaneko H, Ogura Y, Terasaki H, Nagai H, Haro I, Núñez G, Ambati BK, Hinton DR, and Ambati J
- Subjects
- Animals, Humans, Choroidal Neovascularization drug therapy, Choroidal Neovascularization prevention & control, Interleukin-18 therapeutic use, Macular Degeneration drug therapy
- Published
- 2014
- Full Text
- View/download PDF
35. Nucleoside reverse transcriptase inhibitors possess intrinsic anti-inflammatory activity.
- Author
-
Fowler BJ, Gelfand BD, Kim Y, Kerur N, Tarallo V, Hirano Y, Amarnath S, Fowler DH, Radwan M, Young MT, Pittman K, Kubes P, Agarwal HK, Parang K, Hinton DR, Bastos-Carvalho A, Li S, Yasuma T, Mizutani T, Yasuma R, Wright C, and Ambati J
- Subjects
- Alu Elements, Animals, Anti-Inflammatory Agents, Non-Steroidal therapeutic use, Apoptosis drug effects, Carrier Proteins metabolism, Caspase 1 metabolism, Choroidal Neovascularization drug therapy, Disease Models, Animal, Geographic Atrophy drug therapy, Graft vs Host Disease drug therapy, Hepatitis drug therapy, Liver drug effects, Mice, NLR Family, Pyrin Domain-Containing 3 Protein, Receptors, Purinergic P2X7 metabolism, Retinal Pigment Epithelium drug effects, Retinal Pigment Epithelium metabolism, Retinal Pigment Epithelium physiology, Reverse Transcriptase Inhibitors therapeutic use, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Inflammasomes drug effects, Reverse Transcriptase Inhibitors pharmacology
- Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs) are mainstay therapeutics for HIV that block retrovirus replication. Alu (an endogenous retroelement that also requires reverse transcriptase for its life cycle)-derived RNAs activate P2X7 and the NLRP3 inflammasome to cause cell death of the retinal pigment epithelium in geographic atrophy, a type of age-related macular degeneration. We found that NRTIs inhibit P2X7-mediated NLRP3 inflammasome activation independent of reverse transcriptase inhibition. Multiple approved and clinically relevant NRTIs prevented caspase-1 activation, the effector of the NLRP3 inflammasome, induced by Alu RNA. NRTIs were efficacious in mouse models of geographic atrophy, choroidal neovascularization, graft-versus-host disease, and sterile liver inflammation. Our findings suggest that NRTIs are ripe for drug repurposing in P2X7-driven diseases., (Copyright © 2014, American Association for the Advancement of Science.)
- Published
- 2014
- Full Text
- View/download PDF
36. TLR-independent and P2X7-dependent signaling mediate Alu RNA-induced NLRP3 inflammasome activation in geographic atrophy.
- Author
-
Kerur N, Hirano Y, Tarallo V, Fowler BJ, Bastos-Carvalho A, Yasuma T, Yasuma R, Kim Y, Hinton DR, Kirschning CJ, Gelfand BD, and Ambati J
- Subjects
- Animals, Carrier Proteins antagonists & inhibitors, Carrier Proteins metabolism, Caspase 1 metabolism, Disease Models, Animal, Glyburide pharmacology, Humans, Interleukin-18 metabolism, Mice, NF-kappa B antagonists & inhibitors, NF-kappa B metabolism, Nitriles pharmacology, Purinergic P2X Receptor Antagonists pharmacology, RNA, Messenger metabolism, Retinal Pigment Epithelium cytology, Sulfones pharmacology, Alu Elements, Geographic Atrophy physiopathology, Inflammasomes physiology, Receptors, Purinergic P2X7 physiology, Signal Transduction physiology, Toll-Like Receptors physiology
- Abstract
Purpose: Accumulation of Alu RNA transcripts due to DICER1 deficiency in the retinal pigmented epithelium (RPE) promotes geographic atrophy. Recently we showed that Alu RNA activated the NLRP3 inflammasome, leading to RPE cell death via interleukin-18 (IL-18)-mediated MyD88 signaling. However, the molecular basis for NLRP3 inflammasome activation by Alu RNA is not well understood. We sought to decipher the key signaling events triggered by Alu RNA that lead to priming and activation of the NLRP3 inflammasome and, ultimately, to RPE degeneration by investigating the roles of the purinoreceptor P2X7, the transcription factor NF-κB, and the Toll-like receptors (TLRs) in these processes., Methods: Human and mouse RPE cells were transfected with a plasmid encoding an Alu element (pAlu) or an in vitro-transcribed Alu RNA. Inflammasome priming was assessed by measuring NLRP3 and IL18 mRNA levels by real-time quantitative PCR. Using immunoblotting, we assessed NF-κB activation by monitoring phosphorylation of its p65 subunit, and inflammasome activation by monitoring caspase-1 cleavage into its active form. RPE degeneration was induced in mice by subretinal transfection of pAlu or Alu RNA. The NF-κB inhibitor BAY 11-7082, the P2X7 receptor antagonist A-740003, and the NLRP3 inflammasome inhibitor glyburide were delivered by intravitreous injections. We studied wild-type (WT) C57Bl/6J, P2rx7(-/-), Nfkb1(-/-), and Tlr23479(-/-) mice. RPE degeneration was assessed by fundus photography and zonula occludens-1 (ZO-1) staining of mouse RPE., Results: Alu RNA-induced NF-κB activation, independent of TLR-1, -2, -3, -4, -6, -7, and -9 signaling, was required for priming the NLRP3 inflammasome. Nfkb1(-/-) and P2rx7(-/-) mice and WT mice treated with the pharmacological inhibitors of NF-κB, P2X7, or NLRP3, were protected against Alu RNA-induced RPE degeneration., Conclusions: NF-κB and P2X7 are critical signaling intermediates in Alu RNA-induced inflammasome priming and RPE degeneration. These molecules are novel targets for rational drug development for geographic atrophy.
- Published
- 2013
- Full Text
- View/download PDF
37. Retinal angiogenesis suppression through small molecule activation of p53.
- Author
-
Chavala SH, Kim Y, Tudisco L, Cicatiello V, Milde T, Kerur N, Claros N, Yanni S, Guaiquil VH, Hauswirth WW, Penn JS, Rafii S, De Falco S, Lee TC, and Ambati J
- Subjects
- Animals, Apoptosis, Cell Proliferation, Cell Survival, Cells, Cultured, Hindlimb blood supply, Human Umbilical Vein Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells physiology, Humans, Ischemia drug therapy, Macular Degeneration drug therapy, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle physiology, Rats, Retinal Vessels drug effects, Transcriptional Activation drug effects, Tumor Suppressor Protein p53 genetics, Angiogenesis Inhibitors pharmacology, Imidazoles pharmacology, Neovascularization, Physiologic drug effects, Piperazines pharmacology, Retinal Vessels physiology, Tumor Suppressor Protein p53 metabolism
- Abstract
Neovascular age-related macular degeneration is a leading cause of irreversible vision loss in the Western world. Cytokine-targeted therapies (such as anti-vascular endothelial growth factor) are effective in treating pathologic ocular angiogenesis, but have not led to a durable effect and often require indefinite treatment. Here, we show that Nutlin-3, a small molecule antagonist of the E3 ubiquitin protein ligase MDM2, inhibited angiogenesis in several model systems. We found that a functional p53 pathway was essential for Nutlin-3-mediated retinal antiangiogenesis and disruption of the p53 transcriptional network abolished the antiangiogenic activity of Nutlin-3. Nutlin-3 did not inhibit established, mature blood vessels in the adult mouse retina, suggesting that only proliferating retinal vessels are sensitive to Nutlin-3. Furthermore, Nutlin-3 inhibited angiogenesis in nonretinal models such as the hind limb ischemia model. Our work demonstrates that Nutlin-3 functions through an antiproliferative pathway with conceivable advantages over existing cytokine-targeted antiangiogenesis therapies.
- Published
- 2013
- Full Text
- View/download PDF
38. Kaposi's sarcoma-associated herpesvirus latency in endothelial and B cells activates gamma interferon-inducible protein 16-mediated inflammasomes.
- Author
-
Singh VV, Kerur N, Bottero V, Dutta S, Chakraborty S, Ansari MA, Paudel N, Chikoti L, and Chandran B
- Subjects
- Blotting, Western, Gene Expression Profiling, Host-Pathogen Interactions, Humans, B-Lymphocytes virology, Endothelial Cells virology, Herpesvirus 8, Human pathogenicity, Inflammasomes metabolism, Nuclear Proteins metabolism, Phosphoproteins metabolism, Virus Latency
- Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) infections of endothelial and B cells are etiologically linked with Kaposi's sarcoma (KS) and primary effusion B-cell lymphoma (PEL), respectively. KS endothelial and PEL B cells carry multiple copies of the nuclear episomal latent KSHV genome and secrete a variety of inflammatory cytokines, including interleukin-1β (IL-1β) and IL-18. The maturation of IL-1β and IL-18 depends upon active caspase-1, which is regulated by a multiprotein inflammasome complex induced by sensing of danger signals. During primary KSHV infection of endothelial cells, acting as a nuclear pattern recognition receptor, gamma interferon-inducible protein 16 (IFI16) colocalized with the KSHV genome in the nuclei and interacted with ASC and procaspase-1 to form a functional inflammasome (Kerur N et al., Cell Host Microbe 9:363-375, 2011). Here, we demonstrate that endothelial telomerase-immortalized human umbilical cells (TIVE) supporting KSHV stable latency (TIVE-LTC cells) and PEL (cavity-based B-cell lymphoma 1 [BCBL-1]) cells show evidence of inflammasome activation, such as the activation of caspase-1 and cleavage of pro-IL-1β and pro-IL-18. Interaction of ASC with IFI16 but not with AIM2 or NOD-like receptor P3 (NLRP3) was detected. The KSHV latency-associated viral FLIP (vFLIP) gene induced the expression of IL-1β, IL-18, and caspase-1 mRNAs in an NF-κB-dependent manner. IFI16 and cleaved IL-1β were detected in the exosomes released from BCBL-1 cells. Exosomal release could be a KSHV-mediated strategy to subvert IL-1β functions. In fluorescent in situ hybridization analyses, IFI16 colocalized with multiple copies of the KSHV genome in BCBL-1 cells. IFI16 colocalization with ASC was also detected in lung PEL sections from patients. Taken together, these findings demonstrated the constant sensing of the latent KSHV genome by IFI16-mediated innate defense and unraveled a potential mechanism of inflammation induction associated with KS and PEL lesions.
- Published
- 2013
- Full Text
- View/download PDF
39. DICER1 loss and Alu RNA induce age-related macular degeneration via the NLRP3 inflammasome and MyD88.
- Author
-
Tarallo V, Hirano Y, Gelfand BD, Dridi S, Kerur N, Kim Y, Cho WG, Kaneko H, Fowler BJ, Bogdanovich S, Albuquerque RJ, Hauswirth WW, Chiodo VA, Kugel JF, Goodrich JA, Ponicsan SL, Chaudhuri G, Murphy MP, Dunaief JL, Ambati BK, Ogura Y, Yoo JW, Lee DK, Provost P, Hinton DR, Núñez G, Baffi JZ, Kleinman ME, and Ambati J
- Subjects
- Animals, Carrier Proteins metabolism, Geographic Atrophy metabolism, Humans, Inflammasomes metabolism, Mice, NLR Family, Pyrin Domain-Containing 3 Protein, Retinal Pigment Epithelium pathology, Toll-Like Receptors metabolism, Alu Elements, DEAD-box RNA Helicases metabolism, Geographic Atrophy immunology, Geographic Atrophy pathology, Inflammasomes immunology, Myeloid Differentiation Factor 88 metabolism, Retinal Pigment Epithelium metabolism, Ribonuclease III metabolism
- Abstract
Alu RNA accumulation due to DICER1 deficiency in the retinal pigmented epithelium (RPE) is implicated in geographic atrophy (GA), an advanced form of age-related macular degeneration that causes blindness in millions of individuals. The mechanism of Alu RNA-induced cytotoxicity is unknown. Here we show that DICER1 deficit or Alu RNA exposure activates the NLRP3 inflammasome and triggers TLR-independent MyD88 signaling via IL18 in the RPE. Genetic or pharmacological inhibition of inflammasome components (NLRP3, Pycard, Caspase-1), MyD88, or IL18 prevents RPE degeneration induced by DICER1 loss or Alu RNA exposure. These findings, coupled with our observation that human GA RPE contains elevated amounts of NLRP3, PYCARD, and IL18 and evidence of increased Caspase-1 and MyD88 activation, provide a rationale for targeting this pathway in GA. Our findings also reveal a function of the inflammasome outside the immune system and an immunomodulatory action of mobile elements., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
40. IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi Sarcoma-associated herpesvirus infection.
- Author
-
Kerur N, Veettil MV, Sharma-Walia N, Bottero V, Sadagopan S, Otageri P, and Chandran B
- Subjects
- Cells, Cultured, Endothelial Cells immunology, Endothelial Cells virology, Humans, Protein Binding, Protein Interaction Mapping, Herpesvirus 8, Human immunology, Inflammasomes biosynthesis, Inflammasomes immunology, Nuclear Proteins metabolism, Phosphoproteins metabolism
- Abstract
Inflammasomes are cytoplasmic sensors of foreign molecules, including pathogens, and function to induce caspase-1 activation and IL-1β cytokine maturation. Whether such a mechanism exists in the nucleus and is effective against nuclear replicating pathogens is unknown. Nuclear replicating herpesvirus KSHV is associated with Kaposi Sarcoma, an angioproliferative tumor characterized by an inflammatory microenvironment including IL-1β. We demonstrate that during KSHV infection of endothelial cells, interferon gamma-inducible protein 16 (IFI16) interacts with the adaptor molecule ASC and procaspase-1 to form a functional inflammasome. This complex was initially detected in the nucleus and subsequently in the perinuclear area. KSHV gene expression and/or latent KSHV genome is required for inflammasome activation and IFI16 colocalizes with the KSHV genome in the infected cell nucleus. Caspase-1 activation by KSHV was reduced by IFI16 and ASC silencing. Our studies reveal IFI16 as a nuclear pathogen sensor and demonstrate that the inflammasome also functions in the nucleus., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
41. Phosphorylation and polyubiquitination of transforming growth factor beta-activated kinase 1 are necessary for activation of NF-kappaB by the Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor.
- Author
-
Bottero V, Kerur N, Sadagopan S, Patel K, Sharma-Walia N, and Chandran B
- Subjects
- HEK293 Cells, Herpesviridae Infections genetics, Herpesviridae Infections metabolism, Herpesviridae Infections virology, Herpesvirus 8, Human genetics, Humans, MAP Kinase Kinase Kinases genetics, NF-kappa B genetics, Phosphorylation, Protein Binding, Receptors, G-Protein-Coupled genetics, Signal Transduction, Ubiquitination, Viral Proteins genetics, Herpesviridae Infections enzymology, Herpesvirus 8, Human metabolism, MAP Kinase Kinase Kinases metabolism, NF-kappa B metabolism, Receptors, G-Protein-Coupled metabolism, Viral Proteins metabolism
- Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) G protein-coupled receptor (vGPCR) protein has been shown to induce several signaling pathways leading to the modulation of host gene expression. The hijacking of these pathways facilitates the viral life cycle and leads to tumorigenesis. In the present work, we show that transforming growth factor β (TGF-β)-activated kinase 1 (TAK1) is an important player in NF-κB activation induced by vGPCR. We observed that the expression of an inactive TAK1 kinase mutant (TAK1M) reduces vGPCR-induced NF-κB nuclear translocation and transcriptional activity. Consequently, the expression of several NF-κB target genes normally induced by vGPCR was blocked by TAK1M expression, including interleukin 8 (IL-8), Gro1, IκBα, COX-2, cIAP2, and Bcl2 genes. Similar results were obtained after downregulation of TAK1 by small interfering RNA (siRNA) technology. The expression of vGPCR recruited TAK1 to the plasma membrane, and vGPCR interacts with TAK1. vGPCR expression also induced TAK1 phosphorylation and lysine 63-linked polyubiquitination, the two markers of the kinase's activation. Finally, inhibition of TAK1 by celastrol inhibited vGPCR-induced NF-κB activation, indicating this natural compound could be used as a potential therapeutic drug against KSHV malignancies involving vGPCR.
- Published
- 2011
- Full Text
- View/download PDF
42. Interaction of c-Cbl with myosin IIA regulates Bleb associated macropinocytosis of Kaposi's sarcoma-associated herpesvirus.
- Author
-
Valiya Veettil M, Sadagopan S, Kerur N, Chakraborty S, and Chandran B
- Subjects
- Cells, Cultured, Endothelium, Vascular cytology, Humans, Protein Binding, Signal Transduction, Virus Internalization, Cell Membrane Structures virology, Endothelial Cells virology, Herpesvirus 8, Human physiology, Nonmuscle Myosin Type IIA metabolism, Pinocytosis, Proto-Oncogene Proteins c-cbl metabolism
- Abstract
KSHV is etiologically associated with Kaposi's sarcoma (KS), an angioproliferative endothelial cell malignancy. Macropinocytosis is the predominant mode of in vitro entry of KSHV into its natural target cells, human dermal microvascular endothelial (HMVEC-d) cells. Although macropinocytosis is known to be a major route of entry for many viruses, the molecule(s) involved in the recruitment and integration of signaling early during macropinosome formation is less well studied. Here we demonstrate that tyrosine phosphorylation of the adaptor protein c-Cbl is required for KSHV induced membrane blebbing and macropinocytosis. KSHV induced the tyrosine phosphorylation of c-Cbl as early as 1 min post-infection and was recruited to the sites of bleb formation. Infection also led to an increase in the interaction of c-Cbl with PI3-K p85 in a time dependent manner. c-Cbl shRNA decreased the formation of KSHV induced membrane blebs and macropinocytosis as well as virus entry. Immunoprecipitation of c-Cbl followed by mass spectrometry identified the interaction of c-Cbl with a novel molecular partner, non-muscle myosin heavy chain IIA (myosin IIA), in bleb associated macropinocytosis. Phosphorylated c-Cbl colocalized with phospho-myosin light chain II in the interior of blebs of infected cells and this interaction was abolished by c-Cbl shRNA. Studies with the myosin II inhibitor blebbistatin demonstrated that myosin IIA is a biologically significant component of the c-Cbl signaling pathway and c-Cbl plays a new role in the recruitment of myosin IIA to the blebs during KSHV infection. Myosin II associates with actin in KSHV induced blebs and the absence of actin and myosin ubiquitination in c-Cbl ShRNA cells suggested that c-Cbl is also responsible for the ubiquitination of these proteins in the infected cells. This is the first study demonstrating the role of c-Cbl in viral entry as well as macropinocytosis, and provides the evidence that a signaling complex containing c-Cbl and myosin IIA plays a crucial role in blebbing and macropinocytosis during viral infection and suggests that targeting c-Cbl could lead to a block in KSHV infection.
- Published
- 2010
- Full Text
- View/download PDF
43. Characterization of entry and infection of monocytic THP-1 cells by Kaposi's sarcoma associated herpesvirus (KSHV): role of heparan sulfate, DC-SIGN, integrins and signaling.
- Author
-
Kerur N, Veettil MV, Sharma-Walia N, Sadagopan S, Bottero V, Paul AG, and Chandran B
- Subjects
- Antigens, Viral genetics, Cell Line, Cell Nucleus virology, DNA, Viral genetics, DNA, Viral metabolism, Endocytosis physiology, Gene Expression, Herpesvirus 8, Human genetics, Host-Pathogen Interactions physiology, Humans, Immediate-Early Proteins genetics, Kinetics, Monocytes physiology, Nuclear Proteins genetics, Signal Transduction physiology, Trans-Activators genetics, Virus Internalization, Cell Adhesion Molecules physiology, Heparitin Sulfate physiology, Herpesvirus 8, Human pathogenicity, Herpesvirus 8, Human physiology, Integrins physiology, Lectins, C-Type physiology, Monocytes virology, Receptors, Cell Surface physiology
- Abstract
KSHV effectively binds, enters and establishes infection in THP-1 cells with initial concurrent expression of latent ORF73 and lytic ORF50 genes and subsequent persistence of ORF73. KSHV genome persisted for 30 days and lytic cycle could be activated. KSHV utilized heparan sulfate for binding to THP-1 cells and primary monocytes. Blocking DC-SIGN did not inhibit KSHV binding; however, virus entry in THP-1 cells and in primary monocytes was reduced. In addition to the previously identified integrins alpha3beta1, alphavbeta3 and alphavbeta5, integrin alpha5beta1 was also utilized for infection. KSHV entered THP-1 cells via clathrin and caveolin mediated endocytosis and did not utilize macropinocytosis as in human dermal endothelial cells, and required an endosomal acidification. Infection also induced phosphorylation of FAK, Src, PI3K, NF-kappaB and ERK1/2 signaling molecules, and entry was blocked by tyrosine kinase inhibitors. These findings suggest that THP-1 cells are highly useful model for studying KSHV infection of monocytes., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
44. Piracy of prostaglandin E2/EP receptor-mediated signaling by Kaposi's sarcoma-associated herpes virus (HHV-8) for latency gene expression: strategy of a successful pathogen.
- Author
-
George Paul A, Sharma-Walia N, Kerur N, White C, and Chandran B
- Subjects
- Antigens, Viral biosynthesis, Antigens, Viral genetics, Calcium metabolism, Cyclooxygenase 2 biosynthesis, Cyclooxygenase 2 genetics, Cyclooxygenase 2 metabolism, Dinoprostone biosynthesis, Dinoprostone genetics, Down-Regulation, Endothelial Cells virology, Gene Expression, Herpesvirus 8, Human genetics, Herpesvirus 8, Human metabolism, Humans, NF-kappa B metabolism, Nuclear Proteins biosynthesis, Nuclear Proteins genetics, Phosphatidylinositol 3-Kinases metabolism, Promoter Regions, Genetic, Protein Kinase C metabolism, Receptors, Prostaglandin E antagonists & inhibitors, Receptors, Prostaglandin E biosynthesis, Receptors, Prostaglandin E genetics, Sarcoma, Kaposi metabolism, Sarcoma, Kaposi pathology, Sarcoma, Kaposi virology, Signal Transduction, Up-Regulation, Virus Latency, src-Family Kinases metabolism, Dinoprostone metabolism, Herpesvirus 8, Human physiology, Receptors, Prostaglandin E metabolism
- Abstract
Kaposi's sarcoma-associated herpes virus (KSHV) is implicated in the pathogenesis of KS, a chronic inflammation-associated malignancy. Cyclooxygenase-2 (COX-2) and its metabolite prostaglandin E2 (PGE2), two pivotal proinflammatory/oncogeneic molecules, are proposed to play roles in the expression of major KSHV latency-associated nuclear antigen-1 (LANA-1). Microsomal PGE2 synthase, PGE2, and its receptors (EP1, EP2, EP3, and EP4) were detected in KS lesions with the distinct staining of EP2/EP4 in KS lesions. In latently infected endothelial TIVE-LTC cells, EP receptor antagonists downregulated LANA-1 expression as well as Ca(2+), p-Src, p-PI3K, p-PKCzeta/lambda, and p-NF-kappaB, which are also some of the signal molecules proposed to be important in KS pathogenesis. Exogenous PGE2 and EP receptor agonists induced the LANA-1 promoter in 293 cells, and YY1, Sp1, Oct-1, Oct-6, C/EBP, and c-Jun transcription factors seem to be involved in this induction. PGE2/EP receptor-induced LANA-1 promoter activity was downregulated significantly by the inhibition of Ca(2+), p-Src, p-PI3K, p-PKCzeta/lambda, and p-NF-kappaB. These findings implicate the inflammatory PGE2/EP receptors and the associated signal molecules in herpes virus latency and uncover a novel paradigm that shows the evolution of KSHV genome plasticity to use inflammatory response for its survival advantage of maintaining latent gene expression. These data also suggest that potential use of anti-COX-2 and anti-EP receptor therapy may not only ameliorate the chronic inflammation associated with KS but could also lead to elimination of the KSHV latent infection and the associated KS lesions., ((c)2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
45. Kaposi's sarcoma associated herpes virus (KSHV) induced COX-2: a key factor in latency, inflammation, angiogenesis, cell survival and invasion.
- Author
-
Sharma-Walia N, Paul AG, Bottero V, Sadagopan S, Veettil MV, Kerur N, and Chandran B
- Subjects
- Blotting, Western, Cell Adhesion physiology, Cell Separation, Endothelial Cells metabolism, Endothelial Cells virology, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Fluorescent Antibody Technique, Gene Expression, Gene Expression Profiling, Gene Expression Regulation, Humans, Immunohistochemistry, Inflammation enzymology, Neovascularization, Pathologic enzymology, Reverse Transcriptase Polymerase Chain Reaction, Sarcoma, Kaposi virology, Cyclooxygenase 2 metabolism, Herpesvirus 8, Human physiology, Inflammation virology, Neovascularization, Pathologic virology, Sarcoma, Kaposi enzymology, Virus Latency physiology
- Abstract
Kaposi's sarcoma (KS), an enigmatic endothelial cell vascular neoplasm, is characterized by the proliferation of spindle shaped endothelial cells, inflammatory cytokines (ICs), growth factors (GFs) and angiogenic factors. KSHV is etiologically linked to KS and expresses its latent genes in KS lesion endothelial cells. Primary infection of human micro vascular endothelial cells (HMVEC-d) results in the establishment of latent infection and reprogramming of host genes, and cyclooxygenase-2 (COX-2) is one of the highly up-regulated genes. Our previous study suggested a role for COX-2 in the establishment and maintenance of KSHV latency. Here, we examined the role of COX-2 in the induction of ICs, GFs, angiogenesis and invasive events occurring during KSHV de novo infection of endothelial cells. A significant amount of COX-2 was detected in KS tissue sections. Telomerase-immortalized human umbilical vein endothelial cells supporting KSHV stable latency (TIVE-LTC) expressed elevated levels of functional COX-2 and microsomal PGE2 synthase (m-PGES), and secreted the predominant eicosanoid inflammatory metabolite PGE2. Infected HMVEC-d and TIVE-LTC cells secreted a variety of ICs, GFs, angiogenic factors and matrix metalloproteinases (MMPs), which were significantly abrogated by COX-2 inhibition either by chemical inhibitors or by siRNA. The ability of these factors to induce tube formation of uninfected endothelial cells was also inhibited. PGE2, secreted early during KSHV infection, profoundly increased the adhesion of uninfected endothelial cells to fibronectin by activating the small G protein Rac1. COX-2 inhibition considerably reduced KSHV latent ORF73 gene expression and survival of TIVE-LTC cells. Collectively, these studies underscore the pivotal role of KSHV induced COX-2/PGE2 in creating KS lesion like microenvironment during de novo infection. Since COX-2 plays multiple roles in KSHV latent gene expression, which themselves are powerful mediators of cytokine induction, anti-apoptosis, cell survival and viral genome maintainence, effective inhibition of COX-2 via well-characterized clinically approved COX-2 inhibitors could potentially be used in treatment to control latent KSHV infection and ameliorate KS.
- Published
- 2010
- Full Text
- View/download PDF
46. Kaposi sarcoma-associated herpes virus (KSHV) G protein-coupled receptor (vGPCR) activates the ORF50 lytic switch promoter: a potential positive feedback loop for sustained ORF50 gene expression.
- Author
-
Bottero V, Sharma-Walia N, Kerur N, Paul AG, Sadagopan S, Cannon M, and Chandran B
- Subjects
- Base Sequence, Binding Sites, Cell Line, Doxycycline pharmacology, Feedback, Physiological, Gene Expression drug effects, Genes, Viral, Herpesvirus 8, Human pathogenicity, Humans, Models, Biological, Mutation, Promoter Regions, Genetic, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Small Interfering genetics, RNA, Viral genetics, RNA, Viral metabolism, Signal Transduction, Sp1 Transcription Factor metabolism, rho GTP-Binding Proteins metabolism, Herpesvirus 8, Human genetics, Herpesvirus 8, Human physiology, Immediate-Early Proteins genetics, Immediate-Early Proteins physiology, Receptors, Chemokine genetics, Receptors, Chemokine physiology, Trans-Activators genetics, Trans-Activators physiology
- Abstract
KSHV vGPCR, a lytic cycle associated protein, induces several signaling pathways leading to the activation of various transcription factors and consequently the expression of cellular and viral genes. Though the role of vGPCR in KSHV tumorigenicity has been well studied, its function related to the viral life cycle is poorly understood. Reduction in vGPCR by RNA interference also resulted in the reduction in KSHV lytic switch ORF50 gene and protein expression. Induction of vGPCR by doxycycline in BC3.14 cells also resulted in more KSHV production. When this was explored, induction of the ORF50 promoter by vGPCR expression was observed. Further examination of the molecular mechanisms by which vGPCR regulates the ORF50 promoter, using various ORF50 promoter constructs, revealed that induction of ORF50 promoter by vGPCR did not involve AP1 but was dependent on Sp1 and Sp3 transcription factors. vGPCR signaling led to an increase in Sp1 and Sp3 DNA binding activity and a decrease in histone deacetylase (HDAC) activity. These activities were pertussis toxin independent, did not involve Rho and Rac-GTPases and involved the heterotrimeric G protein subunits Galpha12 and Galphaq. Studies using pharmacologic inhibitors and dominant-negative proteins identified phospholipase C, the novel protein kinase C (novel PKC) family and protein kinase D (PKD) as part of the signaling initiated by vGPCR leading to ORF50 promoter activation. Taken together, this study suggests a role for vGPCR in the sustained expression of ORF50 which could lead to a continued activation of lytic cycle genes and ultimately to successful viral progeny formation.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.