38 results on '"Bindi Patel"'
Search Results
2. Acetylated tau inhibits chaperone-mediated autophagy and promotes tau pathology propagation in mice
- Author
-
Benjamin Caballero, Mathieu Bourdenx, Enrique Luengo, Antonio Diaz, Peter Dongmin Sohn, Xu Chen, Chao Wang, Yves R. Juste, Susanne Wegmann, Bindi Patel, Zapporah T. Young, Szu Yu Kuo, Jose Antonio Rodriguez-Navarro, Hao Shao, Manuela G. Lopez, Celeste M. Karch, Alison M. Goate, Jason E. Gestwicki, Bradley T. Hyman, Li Gan, and Ana Maria Cuervo
- Subjects
Science - Abstract
The tau protein has been implicated in neurodegenerative disorders and can propagate from cell to cell. Here, the authors show that tau acetylation reduces its degradation by chaperone-mediated autophagy, causing re-routing to other autophagic pathways and increasing extracellular tau release.
- Published
- 2021
- Full Text
- View/download PDF
3. Descriptive comparison of admission characteristics between pandemic waves and multivariable analysis of the association of the Alpha variant (B.1.1.7 lineage) of SARS-CoV-2 with disease severity in inner London
- Author
-
Keith Morris, Judith Breuer, NgeeKeong Tan, Eric Witele, Sophie Hunter, Monica Panca, Aleks Marek, Paul Flowers, Gaia Nebbia, Sam Haldenby, Jacqui Prieto, Gee Yen Shin, Fiona Mapp, Andrew Copas, Mark Hopkins, Oliver Stirrup, Adam Witney, Kenneth Laing, May Rabuya, Vasa Curcin, Alison Holmes, Mohammad Raza, Wenjuan Wang, Rachel Williams, David Robertson, Julie Samuel, Rory Gunson, Helen Wheeler, Alexander J Keeley, Paul Randell, Cariad Evans, Tabassum Khan, Michelle Ramsay, Darren Smith, James Price, Sarah Francis, Shazaad Ahmad, Finola Higgins, Eleni Nastouli, Abhinav Kumar, Katie Johnson, Sharon Glaysher, Scott Elliott, Rebecca Gregory, Matthew D Parker, Helen Umpleby, Emanuela Pelosi, Emma Thomson, Anna Riddell, Yanzhong Wang, David Harrington, Alexandra Bailey, Nikunj Mahida, Charlotte Williams, Tanzina Haque, David G Partridge, Yusri Taha, Adrienn Angyal, Catherine Houlihan, James Shepherd, Hayley Colton, Chris Davis, Adela Alcolea-Medina, Themoula Charalampous, Beatrix Kele, Irene Monahan, Guy Mollett, Sunando Roy, Joshua Taylor, Sophie Weller, Eleri Wilson-Davies, Joseph Hughes, Tabitha Mahungu, Cassie Pope, Samuel Robson, Kordo Saeed, Luke Snell, James Blackstone, Leanne Hockey, Georgia Marley, Christine Peters, Flavia Flaviani, Bindi Patel, Rahul Batra, Jennifer Hart, Nadua Bayzid, Marius Cotic, Luke Green, Amy State, Alison Cope, Peijun Zhang, Max Whiteley, Marta Gallis Ramalho, Stella Christou, Paige Wolverson, Joe Heffer, Nikki Smith, Salman Goudarzi, Kate Cook, Katie Loveson, Buddhini Samaraweera, Stephen Aplin, Sarah Jeremiah, Matthew Harvey, Thea Sass, Dan Frampton, Matt Byott, Judith Heaney, Ana da Silva Filipe, Thushan de Silva, Jonathan Edgeworth, Luke B Snell, Leonardo de Jongh, Teresa Cutino-Moguel, Raghavendran Kulasegaran-Shylini, Claire E. Broad, Dola Owoyemi, Clare Coffey, Martina Cummins, Tyrra D’Souza, Emily Goldstein, Emilie Shepherd, Katherine Smollett, Alice Broos, Stephen Carmichael, Nicholas Suarez, Sreenu Vattipally, Ioulia Tsatsani, Jacqueline McTaggart, Stephanie McEnhill, Adela Medina, Jörg Saßmannshausen, Sulekha Gurung, Anu Augustine, Sid Mookerjee, Krystal Johnson, Thilipan Thaventhiran, Damien Mine, Isa Ahmad, Anitha Ramanathan, Anu Chawla, Alistair Derby, Becky Taylor, Charles Numbere, Jenifer Mason, Nicholas Machin, Julie Cawthorne, Ryan George, James Montgomery, Deborah McKew, Angela Cobb, Maria Leader, Shirelle Burton-Fanning, Lydia Taylor, Matthew Bashton, Matthew Crown, Matthew Loose, Patrick McClure, Mitch Clarke, Elaine Baxter, Carl Yates, Irfan Aslam, Vicki Fleming, Michelle Lister, Johnny Debebe, Nadine Holmes, Christopher Moore, Matt Carlile, Dianne Irish-Tavares, Mia De Mesa, Vicky Pang, Jelena Heaphy, Wendy Chatterton, Monika Pusok, Tranprit Saluja, Zahira Maqsood, Angie Williams, Debbie Devonport, Lucy Palinkas, Diane Thomlinson, Julie Booth, Ashok Dadrah, Amanda Symonds, Cassandra Craig, Benjamin B Lindsey, Benjamin H Foulkes, Stavroula F Louka, Phillip Ravencroft, Sharon Hsu, Nasar Ali, Rasha Raghei, Samantha E Hansford, Hailey R Hornsby, Phil Wade, Kay Cawthron, Maqsood Khan, Amber Ford, Imogen Wilson, Kate Harrington, Nic Tinker, Sally Nyinza, Adhyana Mahanama, Siona Silviera, Christopher Fearn, Claudia Cardosa Pereira, Vaz Malik, Gema Martinez-Garcia, Leila Hail, Ndifreke Atang, Helen Francis, Milica Rajkov, Rachel McComish, Alyson MacNeil, Alif Tamuri, and Stefan Piatek
- Subjects
Medicine - Published
- 2022
- Full Text
- View/download PDF
4. SARS-CoV-2 lineage B.1.1.7 is associated with greater disease severity among hospitalised women but not men: multicentre cohort study
- Author
-
Judith Breuer, Catherine F Houlihan, David Partridge, Gaia Nebbia, Jacqui Prieto, Gee Yen Shin, Oliver Stirrup, Kenneth Laing, Rachel Williams, Helen Wheeler, Paul Randell, Ana da Silva Filipe, Tommy Rampling, Tabassum Khan, James Price, Sharon Glaysher, Scott Elliott, Helen Umpleby, Emanuela Pelosi, Emma Thomson, Cristina Venturini, Anna Riddell, Alison Cox, Andrew C Hayward, Malin Bergström, David Harrington, Charlotte Williams, Tanzina Haque, Dianne Irish, Adrienn Angyal, Marios Margaritis, Florencia Boshier, José Afonso Guerra-Assunção, Adela Alcolea-Medina, Angela Beckett, Themoula Charalampous, Raghavendran Kulasegaran Shylini, Beatrix Kele, Irene Monahan, Guy Mollett, Matthew Parker, Sunando Roy, Joshua Taylor, Sophie Weller, Eleri Wilson-Davies, Phillip Wade, Joseph Hughes, Tabitha Mahungu, Cassie Pope, Samuel Robson, Kordo Saeed, Thushan de Silva, Luke Snell, Adam A Witney, James Blackstone, Leanne Hockey, Georgia Marley, Christine Peters, Flavia Flaviani, Bindi Patel, Tom G S Williams, Rahul Batra, Jonathan D Edgeworth, Pinglawathee Madona, Jennifer Hart, Juanita Pang, Helena Tutill, Nadua Bayzid, Marius Cotic, Luke Green, Benjamin Lindsey, Amy State, Alison Cope, Peijun Zhang, Max Whiteley, Marta Gallis Ramalho, Stella Christou, Stavroula Louka, Hailey Hornsby, Benjamin Foulkes, Paige Wolverson, Joe Heffer, Nikki Smith, Salman Goudarzi, Chris Fearn, Kate Cook, Katie Loveson, Adhyana Mahamana, Buddhini Samaraweera, Siona Silveira, Stephen Aplin, Sarah Jeremiah, Matthew Harvey, Thea Sass, Ngee Keong Tan, Claudia Cardoso Pereira, Dan Frampton, Matt Byott, Judith Heaney, Emilie Sanchez, and Stavroula M Paraskevopoulou
- Subjects
Medicine ,Diseases of the respiratory system ,RC705-779 - Published
- 2021
- Full Text
- View/download PDF
5. The impact of viral mutations on recognition by SARS-CoV-2 specific T cells
- Author
-
Thushan I. de Silva, Guihai Liu, Benjamin B. Lindsey, Danning Dong, Shona C. Moore, Nienyun Sharon Hsu, Dhruv Shah, Dannielle Wellington, Alexander J. Mentzer, Adrienn Angyal, Rebecca Brown, Matthew D. Parker, Zixi Ying, Xuan Yao, Lance Turtle, Susanna Dunachie, Mala K. Maini, Graham Ogg, Julian C. Knight, Yanchun Peng, Sarah L. Rowland-Jones, Tao Dong, David M. Aanensen, Khalil Abudahab, Helen Adams, Alexander Adams, Safiah Afifi, Dinesh Aggarwal, Shazaad S.Y. Ahmad, Louise Aigrain, Adela Alcolea-Medina, Nabil-Fareed Alikhan, Elias Allara, Roberto Amato, Tara Annett, Stephen Aplin, Cristina V. Ariani, Hibo Asad, Amy Ash, Paula Ashfield, Fiona Ashford, Laura Atkinson, Stephen W. Attwood, Cressida Auckland, Alp Aydin, David J. Baker, Paul Baker, Carlos E. Balcazar, Jonathan Ball, Jeffrey C. Barrett, Magdalena Barrow, Edward Barton, Matthew Bashton, Andrew R. Bassett, Rahul Batra, Chris Baxter, Nadua Bayzid, Charlotte Beaver, Angela H. Beckett, Shaun M. Beckwith, Luke Bedford, Robert Beer, Andrew Beggs, Katherine L. Bellis, Louise Berry, Beatrice Bertolusso, Angus Best, Emma Betteridge, David Bibby, Kelly Bicknell, Debbie Binns, Alec Birchley, Paul W. Bird, Chloe Bishop, Rachel Blacow, Victoria Blakey, Beth Blane, Frances Bolt, James Bonfield, Stephen Bonner, David Bonsall, Tim Boswell, Andrew Bosworth, Yann Bourgeois, Olivia Boyd, Declan T. Bradley, Cassie Breen, Catherine Bresner, Judith Breuer, Stephen Bridgett, Iraad F. Bronner, Ellena Brooks, Alice Broos, Julianne R. Brown, Giselda Bucca, Sarah L. Buchan, David Buck, Matthew Bull, Phillipa J. Burns, Shirelle Burton-Fanning, Timothy Byaruhanga, Matthew Byott, Sharon Campbell, Alessandro M. Carabelli, James S. Cargill, Matthew Carlile, Silvia F. Carvalho, Anna Casey, Anibolina Castigador, Jana Catalan, Vicki Chalker, Nicola J. Chaloner, Meera Chand, Joseph G. Chappell, Themoula Charalampous, Wendy Chatterton, Yasmin Chaudhry, Carol M. Churcher, Gemma Clark, Phillip Clarke, Benjamin J. Cogger, Kevin Cole, Jennifer Collins, Rachel Colquhoun, Thomas R. Connor, Kate F. Cook, Jason Coombes, Sally Corden, Claire Cormie, Nicholas Cortes, Marius Cotic, Seb Cotton, Simon Cottrell, Lindsay Coupland, MacGregor Cox, Alison Cox, Noel Craine, Liam Crawford, Aidan Cross, Matthew R. Crown, Dorian Crudgington, Nicola Cumley, Tanya Curran, Martin D. Curran, Ana da Silva Filipe, Gavin Dabrera, Alistair C. Darby, Rose K. Davidson, Alisha Davies, Robert M. Davies, Thomas Davis, Daniela de Angelis, Elen De Lacy, Leonardo de Oliveira Martins, Johnny Debebe, Rebecca Denton-Smith, Samir Dervisevic, Rebecca Dewar, Jayasree Dey, Joana Dias, Donald Dobie, Matthew J. Dorman, Fatima Downing, Megan Driscoll, Louis du Plessis, Nichola Duckworth, Jillian Durham, Kirstine Eastick, Lisa J. Easton, Richard Eccles, Jonathan Edgeworth, Sue Edwards, Kate El Bouzidi, Sahar Eldirdiri, Nicholas Ellaby, Scott Elliott, Gary Eltringham, Leah Ensell, Michelle J. Erkiert, Marina Escalera Zamudio, Sarah Essex, Johnathan M. Evans, Cariad Evans, William Everson, Derek J. Fairley, Karlie Fallon, Arezou Fanaie, Ben W. Farr, Christopher Fearn, Theresa Feltwell, Lynne Ferguson, Laia Fina, Flavia Flaviani, Vicki M. Fleming, Sally Forrest, Ebenezer Foster-Nyarko, Benjamin H. Foulkes, Luke Foulser, Mireille Fragakis, Dan Frampton, Sarah Francois, Christophe Fraser, Timothy M. Freeman, Helen Fryer, Marc Fuchs, William Fuller, Kavitha Gajee, Katerina Galai, Abbie Gallagher, Eileen Gallagher, Michael D. Gallagher, Marta Gallis, Amy Gaskin, Bree Gatica-Wilcox, Lily Geidelberg, Matthew Gemmell, Iliana Georgana, Ryan P. George, Laura Gifford, Lauren Gilbert, Sophia T. Girgis, Sharon Glaysher, Emily J. Goldstein, Tanya Golubchik, Andrea N. Gomes, Sónia Gonçalves, Ian G. Goodfellow, Scott Goodwin, Salman Goudarzi, Marina Gourtovaia, Clive Graham, Lee Graham, Paul R. Grant, Luke R. Green, Angie Green, Jane Greenaway, Richard Gregory, Martyn Guest, Rory N. Gunson, Ravi K. Gupta, Bernardo Gutierrez, Sam T. Haldenby, William L. Hamilton, Samantha E. Hansford, Tanzina Haque, Kathryn A. Harris, Ian Harrison, Ewan M. Harrison, Jennifer Hart, John A. Hartley, William T. Harvey, Matthew Harvey, Mohammed O. Hassan-Ibrahim, Judith Heaney, Thomas Helmer, John H. Henderson, Andrew R. Hesketh, Jessica Hey, David Heyburn, Ellen E. Higginson, Verity Hill, Jack D. Hill, Rachel A. Hilson, Ember Hilvers, Matthew T.G. Holden, Amy Hollis, Christopher W. Holmes, Nadine Holmes, Alison H. Holmes, Richard Hopes, Hailey R. Hornsby, Myra Hosmillo, Catherine Houlihan, Hannah C. Howson-Wells, Jonathan Hubb, Hannah Huckson, Warwick Hughes, Joseph Hughes, Margaret Hughes, Stephanie Hutchings, Giles Idle, Chris J. Illingworth, Robert Impey, Dianne Irish-Tavares, Miren Iturriza-Gomara, Rhys Izuagbe, Chris Jackson, Ben Jackson, Leigh M. Jackson, Kathryn A. Jackson, David K. Jackson, Aminu S. Jahun, Victoria James, Keith James, Christopher Jeanes, Aaron R. Jeffries, Sarah Jeremiah, Andrew Jermy, Michaela John, Rob Johnson, Kate Johnson, Ian Johnston, Owen Jones, Sophie Jones, Hannah Jones, Christopher R. Jones, Neil Jones, Amelia Joseph, Sarah Judges, Gemma L. Kay, Sally Kay, Jon-Paul Keatley, Alexander J. Keeley, Anita Kenyon, Leanne M. Kermack, Manjinder Khakh, Stephen P. Kidd, Maimuna Kimuli, Stuart Kirk, Christine Kitchen, Katie Kitchman, Bridget A. Knight, Cherian Koshy, Moritz U.G. Kraemer, Sara Kumziene-Summerhayes, Dominic Kwiatkowski, Angie Lackenby, Kenneth G. Laing, Temi Lampejo, Cordelia F. Langford, Deborah Lavin, Andrew I. Lawton, Jack Lee, David Lee, Stefanie V. Lensing, Steven Leonard, Lisa J. Levett, Thanh Le-Viet, Jonathan Lewis, Kevin Lewis, Jennifier Liddle, Steven Liggett, Patrick J. Lillie, Michelle M. Lister, Rich Livett, Stephanie Lo, Nicholas J. Loman, Matthew W. Loose, Stavroula F. Louka, Katie F. Loveson, Sarah Lowdon, Hannah Lowe, Helen L. Lowe, Anita O. Lucaci, Catherine Ludden, Jessica Lynch, Ronan A. Lyons, Katrina Lythgoe, Nicholas W. Machin, George MacIntyre-Cockett, Andrew Mack, Ben Macklin, Alasdair Maclean, Emily Macnaughton, Pinglawathee Madona, Mailis Maes, Laurentiu Maftei, Adhyana I.K. Mahanama, Tabitha W. Mahungu, Daniel Mair, Joshua Maksimovic, Cassandra S. Malone, Daniel Maloney, Nikos Manesis, Robin Manley, Anna Mantzouratou, Angela Marchbank, Arun Mariappan, Inigo Martincorena, Rocio T. Martinez Nunez, Alison E. Mather, Patrick Maxwell, Megan Mayhew, Tamyo Mbisa, Clare M. McCann, Shane A. McCarthy, Kathryn McCluggage, Patrick C. McClure, J.T. McCrone, Martin P. McHugh, James P. McKenna, Caoimhe McKerr, Georgina M. McManus, Claire L. McMurray, Claire McMurray, Alan McNally, Lizzie Meadows, Nathan Medd, Oliver Megram, Mirko Menegazzo, Ian Merrick, Stephen L. Michell, Michelle L. Michelsen, Mariyam Mirfenderesky, Jeremy Mirza, Julia Miskelly, Emma Moles-Garcia, Robin J. Moll, Zoltan Molnar, Irene M. Monahan, Matteo Mondani, Siddharth Mookerjee, Christopher Moore, Jonathan Moore, Nathan Moore, Catherine Moore, Helen Morcrette, Sian Morgan, Mari Morgan, Matilde Mori, Arthur Morriss, Samuel Moses, Craig Mower, Peter Muir, Afrida Mukaddas, Florence Munemo, Robert Munn, Abigail Murray, Leanne J. Murray, Darren R. Murray, Manasa Mutingwende, Richard Myers, Eleni Nastouli, Gaia Nebbia, Andrew Nelson, Charlotte Nelson, Sam Nicholls, Jenna Nichols, Roberto Nicodemi, Kyriaki Nomikou, Justin O’Grady, Sarah O'Brien, Mina Odedra, Natasha Ohemeng-Kumi, Karen Oliver, Richard J. Orton, Husam Osman, xeine O'Toole, Nicole Pacchiarini, Debra Padgett, Andrew J. Page, Emily J. Park, Naomi R. Park, Surendra Parmar, David G. Partridge, David Pascall, Amita Patel, Bindi Patel, Steve Paterson, Brendan A.I. Payne, Sharon J. Peacock, Clare Pearson, Emanuela Pelosi, Benita Percival, Jon Perkins, Malorie Perry, Malte L. Pinckert, Steven Platt, Olga Podplomyk, Manoj Pohare, Marcus Pond, Cassie F. Pope, Radoslaw Poplawski, Jessica Powell, Jennifer Poyner, Liam Prestwood, Anna Price, James R. Price, Jacqui A. Prieto, David T. Pritchard, Sophie J. Prosolek, Georgia Pugh, Monika Pusok, Oliver G. Pybus, Hannah M. Pymont, Michael A. Quail, Joshua Quick, Clara Radulescu, Jayna Raghwani, Manon Ragonnet-Cronin, Lucille Rainbow, Diana Rajan, Shavanthi Rajatileka, Newara A. Ramadan, Andrew Rambaut, John Ramble, Paul A. Randell, Paul Randell, Liz Ratcliffe, Veena Raviprakash, Mohammad Raza, Nicholas M. Redshaw, Sara Rey, Nicola Reynolds, Alex Richter, David L. Robertson, Esther Robinson, Samuel C. Robson, Fiona Rogan, Stefan Rooke, Will Rowe, Sunando Roy, Steven Rudder, Chris Ruis, Steven Rushton, Felicity Ryan, Kordo Saeed, Buddhini Samaraweera, Christine M. Sambles, Roy Sanderson, Theo Sanderson, Fei Sang, Thea Sass, Emily Scher, Garren Scott, Carol Scott, Jasveen Sehmi, Sharif Shaaban, Divya Shah, Jessica Shaw, Ekaterina Shelest, James G. Shepherd, Liz A. Sheridan, Nicola Sheriff, Lesley Shirley, John Sillitoe, Siona Silviera, David A. Simpson, Aditi Singh, Dawn Singleton, Timofey Skvortsov, Tim J. Sloan, Graciela Sluga, Ken Smith, Kim S. Smith, Perminder Smith, Darren L. Smith, Louise Smith, Colin P. Smith, Nikki Smith, Katherine L. Smollett, Luke B. Snell, Thomas Somassa, Joel Southgate, Karla Spellman, Michael H. Spencer Chapman, Lewis G. Spurgin, Moira J. Spyer, Rachael Stanley, William Stanley, Thomas D. Stanton, Igor Starinskij, Joanne Stockton, Susanne Stonehouse, Nathaniel Storey, David J. Studholme, Malur Sudhanva, Emma Swindells, Yusri Taha, Ngee Keong Tan, Julian W. Tang, Miao Tang, Ben E.W. Taylor, Joshua F. Taylor, Sarah Taylor, Ben Temperton, Kate E. Templeton, Claire Thomas, Laura Thomson, Emma C. Thomson, Alicia Thornton, Scott A.J. Thurston, John A. Todd, Rachael Tomb, Lily Tong, Gerry Tonkin-Hill, M. Estee Torok, Jaime M. Tovar-Corona, Amy Trebes, Alexander J. Trotter, Ioulia Tsatsani, Robyn Turnbull, Katherine A. Twohig, Helen Umpleby, Anthony P. Underwood, Edith E. Vamos, Tetyana I. Vasylyeva, Sreenu Vattipally, Gabrielle Vernet, Barry B. Vipond, Erik M. Volz, Sarah Walsh, Dennis Wang, Ben Warne, Joanna Warwick-Dugdale, Elizabeth Wastnedge, Joanne Watkins, Louisa K. Watson, Sheila Waugh, Hermione J. Webster, Danni Weldon, Elaine Westwick, Thomas Whalley, Helen Wheeler, Mark Whitehead, Max Whiteley, Andrew Whitwham, Claudia Wierzbicki, Nicholas J. Willford, Lesley-Anne Williams, Rebecca Williams, Cheryl Williams, Chris Williams, Charlotte A. Williams, Rachel J. Williams, Thomas Williams, Catryn Williams, Kathleen A. Williamson, Eleri Wilson-Davies, Eric Witele, Karen T. Withell, Adam A. Witney, Paige Wolverson, Nick Wong, Trudy Workman, Victoria Wright, Derek W. Wright, Tim Wyatt, Sarah Wyllie, Li Xu-McCrae, Mehmet Yavus, Geraldine Yaze, Corin A. Yeats, Gonzalo Yebra, Wen C. Yew, Gregory R. Young, Jamie Young, Alex E. Zarebski, Peijun Zhang, J. Kenneth Baillie, Malcolm G. Semple, Peter J.M. Openshaw, Gail Carson, Beatrice Alex, Petros Andrikopoulos, Benjamin Bach, Wendy S. Barclay, Debby Bogaert, Kanta Chechi, Graham S. Cooke, Annemarie B. Docherty, Gonçalo dos Santos Correia, Marc-Emmanuel Dumas, Jake Dunning, Tom Fletcher, Christopher A. Green, William Greenhalf, Julian L. Griffin, Rishi K. Gupta, Ewen M. Harrison, Julian A. Hiscox, Antonia Ying Wai Ho, Peter W. Horby, Samreen Ijaz, Saye Khoo, Paul Klenerman, Andrew Law, Matthew R. Lewis, Sonia Liggi, Wei Shen Lim, Lynn Maslen, Laura Merson, Alison M. Meynert, Mahdad Noursadeghi, Michael Olanipekun, Anthonia Osagie, Massimo Palmarini, Carlo Palmieri, William A. Paxton, Georgios Pollakis, Nicholas Price, Clark D. Russell, Vanessa Sancho-Shimizu, Caroline J. Sands, Janet T. Scott, Louise Sigfrid, Tom Solomon, Shiranee Sriskandan, David Stuart, Charlotte Summers, Olivia V. Swann, Zoltan Takats, Panteleimon Takis, Richard S. Tedder, A.A. Roger Thompson, Ryan S. Thwaites, Maria Zambon, Hayley Hardwick, Chloe Donohue, Fiona Griffiths, Wilna Oosthuyzen, Cara Donegan, Rebecca G. Spencer, Jo Dalton, Michelle Girvan, Egle Saviciute, Stephanie Roberts, Janet Harrison, Laura Marsh, Marie Connor, Sophie Halpin, Clare Jackson, Carrol Gamble, Daniel Plotkin, James Lee, Gary Leeming, Murray Wham, Sara Clohisey, Ross Hendry, James Scott-Brown, Victoria Shaw, Sarah E. McDonald, Seán Keating, Katie A. Ahmed, Jane A. Armstrong, Milton Ashworth, Innocent G. Asiimwe, Siddharth Bakshi, Samantha L. Barlow, Laura Booth, Benjamin Brennan, Katie Bullock, Benjamin W.A. Catterall, Jordan J. Clark, Emily A. Clarke, Sarah Cole, Louise Cooper, Helen Cox, Christopher Davis, Oslem Dincarslan, Chris Dunn, Philip Dyer, Angela Elliott, Anthony Evans, Lorna Finch, Lewis W.S. Fisher, Terry Foster, Isabel Garcia-Dorival, Philip Gunning, Catherine Hartley, Rebecca L. Jensen, Christopher B. Jones, Trevor R. Jones, Shadia Khandaker, Katharine King, Robyn T. Kiy, Chrysa Koukorava, Annette Lake, Suzannah Lant, Diane Latawiec, Lara Lavelle-Langham, Daniella Lefteri, Lauren Lett, Lucia A. Livoti, Maria Mancini, Sarah McDonald, Laurence McEvoy, John McLauchlan, Soeren Metelmann, Nahida S. Miah, Joanna Middleton, Joyce Mitchell, Ellen G. Murphy, Rebekah Penrice-Randal, Jack Pilgrim, Tessa Prince, Will Reynolds, P. Matthew Ridley, Debby Sales, Victoria E. Shaw, Rebecca K. Shears, Benjamin Small, Krishanthi S. Subramaniam, Agnieska Szemiel, Aislynn Taggart, Jolanta Tanianis-Hughes, Jordan Thomas, Erwan Trochu, Libby van Tonder, Eve Wilcock, J. Eunice Zhang, Lisa Flaherty, Nicole Maziere, Emily Cass, Alejandra Doce Carracedo, Nicola Carlucci, Anthony Holmes, Hannah Massey, Lee Murphy, Nicola Wrobel, Sarah McCafferty, Kirstie Morrice, Alan MacLean, Kayode Adeniji, Daniel Agranoff, Ken Agwuh, Dhiraj Ail, Erin L. Aldera, Ana Alegria, Sam Allen, Brian Angus, Abdul Ashish, Dougal Atkinson, Shahedal Bari, Gavin Barlow, Stella Barnass, Nicholas Barrett, Christopher Bassford, Sneha Basude, David Baxter, Michael Beadsworth, Jolanta Bernatoniene, John Berridge, Colin Berry, Nicola Best, Pieter Bothma, David Chadwick, Robin Brittain-Long, Naomi Bulteel, Tom Burden, Andrew Burtenshaw, Vikki Caruth, Duncan Chambler, Nigel Chee, Jenny Child, Srikanth Chukkambotla, Tom Clark, Paul Collini, Catherine Cosgrove, Jason Cupitt, Maria-Teresa Cutino-Moguel, Paul Dark, Chris Dawson, Phil Donnison, Sam Douthwaite, Andrew Drummond, Ingrid DuRand, Ahilanadan Dushianthan, Tristan Dyer, Chi Eziefula, Chrisopher Fegan, Adam Finn, Duncan Fullerton, Sanjeev Garg, Atul Garg, Effrossyni Gkrania-Klotsas, Jo Godden, Arthur Goldsmith, Elaine Hardy, Stuart Hartshorn, Daniel Harvey, Peter Havalda, Daniel B. Hawcutt, Maria Hobrok, Luke Hodgson, Anil Hormis, Michael Jacobs, Susan Jain, Paul Jennings, Agilan Kaliappan, Vidya Kasipandian, Stephen Kegg, Michael Kelsey, Jason Kendall, Caroline Kerrison, Ian Kerslake, Oliver Koch, Gouri Koduri, George Koshy, Shondipon Laha, Steven Laird, Susan Larkin, Tamas Leiner, Patrick Lillie, James Limb, Vanessa Linnett, Jeff Little, Mark Lyttle, Michael MacMahon, Emily MacNaughton, Ravish Mankregod, Huw Masson, Elijah Matovu, Katherine McCullough, Ruth McEwen, Manjula Meda, Gary Mills, Jane Minton, Kavya Mohandas, Quen Mok, James Moon, Elinoor Moore, Patrick Morgan, Craig Morris, Katherine Mortimore, Mbiye Mpenge, Rohinton Mulla, Michael Murphy, Megan Nagel, Thapas Nagarajan, Mark Nelson, Lillian Norris, Matthew K. O'Shea, Igor Otahal, Marlies Ostermann, Mark Pais, Selva Panchatsharam, Danai Papakonstantinou, Hassan Paraiso, Brij Patel, Natalie Pattison, Justin Pepperell, Mark Peters, Mandeep Phull, Stefania Pintus, Jagtur Singh Pooni, Tim Planche, Frank Post, David Price, Rachel Prout, Nikolas Rae, Henrik Reschreiter, Tim Reynolds, Neil Richardson, Mark Roberts, Devender Roberts, Alistair Rose, Guy Rousseau, Bobby Ruge, Brendan Ryan, Taranprit Saluja, Matthias L. Schmid, Aarti Shah, Prad Shanmuga, Anil Sharma, Anna Shawcross, Jeremy Sizer, Manu Shankar-Hari, Richard Smith, Catherine Snelson, Nick Spittle, Nikki Staines, Tom Stambach, Richard Stewart, Pradeep Subudhi, Tamas Szakmany, Kate Tatham, Jo Thomas, Chris Thompson, Robert Thompson, Ascanio Tridente, Darell Tupper-Carey, Mary Twagira, Nick Vallotton, Rama Vancheeswaran, Lisa Vincent-Smith, Shico Visuvanathan, Alan Vuylsteke, Sam Waddy, Rachel Wake, Andrew Walden, Ingeborg Welters, Tony Whitehouse, Paul Whittaker, Ashley Whittington, Padmasayee Papineni, Meme Wijesinghe, Martin Williams, Lawrence Wilson, Stephen Winchester, Martin Wiselka, Adam Wolverson, Daniel G. Wootton, Andrew Workman, Bryan Yates, and Peter Young
- Subjects
Phylogenetics ,Molecular biology ,Immunology ,Immune response ,Virology ,Science - Abstract
Summary: We identify amino acid variants within dominant SARS-CoV-2 T cell epitopes by interrogating global sequence data. Several variants within nucleocapsid and ORF3a epitopes have arisen independently in multiple lineages and result in loss of recognition by epitope-specific T cells assessed by IFN-γ and cytotoxic killing assays. Complete loss of T cell responsiveness was seen due to Q213K in the A∗01:01-restricted CD8+ ORF3a epitope FTSDYYQLY207-215; due to P13L, P13S, and P13T in the B∗27:05-restricted CD8+ nucleocapsid epitope QRNAPRITF9-17; and due to T362I and P365S in the A∗03:01/A∗11:01-restricted CD8+ nucleocapsid epitope KTFPPTEPK361-369. CD8+ T cell lines unable to recognize variant epitopes have diverse T cell receptor repertoires. These data demonstrate the potential for T cell evasion and highlight the need for ongoing surveillance for variants capable of escaping T cell as well as humoral immunity.
- Published
- 2021
- Full Text
- View/download PDF
6. Comparative performance of SARS-CoV-2 lateral flow antigen tests and association with detection of infectious virus in clinical specimens: a single-centre laboratory evaluation study
- Author
-
Suzanne Pickering, PhD, Rahul Batra, MD, Blair Merrick, MBChB, Luke B Snell, MBBS, Gaia Nebbia, FRCPath, Sam Douthwaite, MRCP, Fiona Reid, MSc, Amita Patel, BSc, Mark Tan Kia Ik, BSc, Bindi Patel, BSc, Themoula Charalampous, PhD, Adela Alcolea-Medina, MPhil, Maria Jose Lista, PhD, Penelope R Cliff, PhD, Emma Cunningham, MSc, Jane Mullen, BSc, Katie J Doores, PhD, Jonathan D Edgeworth, ProfFRCPath, Michael H Malim, ProfDPhil, Stuart J D Neil, ProfPhD, and Rui Pedro Galão, PhD
- Subjects
Medicine (General) ,R5-920 ,Microbiology ,QR1-502 - Abstract
Summary: Background: Lateral flow devices (LFDs) for rapid antigen testing are set to become a cornerstone of SARS-CoV-2 mass community testing, although their reduced sensitivity compared with PCR has raised questions of how well they identify infectious cases. Understanding their capabilities and limitations is, therefore, essential for successful implementation. We evaluated six commercial LFDs and assessed their correlation with infectious virus culture and PCR cycle threshold (Ct) values. Methods: In a single-centre, laboratory evaluation study, we did a head-to-head comparison of six LFDs commercially available in the UK: Innova Rapid SARS-CoV-2 Antigen Test, Spring Healthcare SARS-CoV-2 Antigen Rapid Test Cassette, E25Bio Rapid Diagnostic Test, Encode SARS-CoV-2 Antigen Rapid Test Device, SureScreen COVID-19 Rapid Antigen Test Cassette, and SureScreen COVID-19 Rapid Fluorescence Antigen Test. We estimated the specificities and sensitivities of the LFDs using stored naso-oropharyngeal swabs collected at St Thomas' Hospital (London, UK) for routine diagnostic SARS-CoV-2 testing by real-time RT-PCR (RT-rtPCR). Swabs were from inpatients and outpatients from all departments of St Thomas' Hospital, and from health-care staff (all departments) and their household contacts. SARS-CoV-2-negative swabs from the same population (confirmed by RT-rtPCR) were used for comparative specificity determinations. All samples were collected between March 23 and Oct 27, 2020. We determined the limit of detection (LOD) for each test using viral plaque-forming units (PFUs) and viral RNA copy numbers of laboratory-grown SARS-CoV-2. Additionally, LFDs were selected to assess the correlation of antigen test result with RT-rtPCR Ct values and positive viral culture in Vero E6 cells. This analysis included longitudinal swabs from five infected inpatients with varying disease severities. Furthermore, the sensitivities of available LFDs were assessed in swabs (n=23; collected from Dec 4, 2020, to Jan 12, 2021) confirmed to be positive (RT-rtPCR and whole-genome sequencing) for the B.1.1.7 variant, which was the dominant genotype in the UK at the time of study completion. Findings: All LFDs showed high specificity (≥98·0%), except for the E25Bio test (86·0% [95% CI 77·9–99·9]), and most tests reliably detected 50 PFU/test (equivalent SARS-CoV-2 N gene Ct value of 23·7, or RNA copy number of 3 × 106/mL). Sensitivities of the LFDs on clinical samples ranged from 65·0% (55·2–73·6) to 89·0% (81·4–93·8). These sensitivities increased to greater than 90% for samples with Ct values of lower than 25 for all tests except the SureScreen fluorescence (SureScreen-F) test. Positive virus culture was identified in 57 (40·4%) of 141 samples; 54 (94·7%) of the positive cultures were from swabs with Ct values lower than 25. Among the three LFDs selected for detailed comparisons (the tests with highest sensitivity [Innova], highest specificity [Encode], and alternative technology [SureScreen-F]), sensitivity of the LFDs increased to at least 94·7% when only including samples with detected viral growth. Longitudinal studies of RT-rtPCR-positive samples (tested with Innova, Encode, and both SureScreen-F and the SureScreen visual [SureScreen-V] test) showed that most of the tests identified all infectious samples as positive. Test performance (assessed for Innova and SureScreen-V) was not affected when reassessed on swabs positive for the UK variant B.1.1.7. Interpretation: In this comprehensive comparison of antigen LFDs and virus infectivity, we found a clear relationship between Ct values, quantitative culture of infectious virus, and antigen LFD positivity in clinical samples. Our data support regular testing of target groups with LFDs to supplement the current PCR testing capacity, which would help to rapidly identify infected individuals in situations in which they would otherwise go undetected. Funding: King's Together Rapid COVID-19, Medical Research Council, Wellcome Trust, Huo Family Foundation, UK Department of Health, National Institute for Health Research Comprehensive Biomedical Research Centre.
- Published
- 2021
- Full Text
- View/download PDF
7. Study on Juvenile Delinquency among Adolescents in Secondary Schools
- Author
-
null Bindi Patel, null Mr. Parashram, and null Ishwar Das Vairagi
- Subjects
General Earth and Planetary Sciences ,General Environmental Science - Abstract
Today, more than ever, the problem of juvenile delinquency pricks at the conscience of many societies, and India is not an exception. In this study, the researcher discusses the experiences and views of teachers of secondary schools regarding juvenile delinquency and the apparent moral decline among their learners. The problem was investigated from the perspective of socio-education, a field of research which, amongst others, studies the role of social factors that feature in the development of children and youth during their growth to adulthood. In Indian schools, the problem of juvenile delinquency is a issue. In his article on discipline in the Free State township schools, Masitsa (2008: 234- 236), cites a number of newspaper reports of juvenile delinquency among school children. This problem is not unique to South Africa, but affects many industrialised countries today (Rossouw, 2003: 416). De Wet’s study (2004: 206) on school vandalism, which is an example of antisocial behaviour, reveals that the problem also affects Britain, USA, Canada, France, the Netherlands and Australia, amongst others. MATERIALS AND METHODS: The research design for this study was a combination of quantitative and mainly qualitative, descriptive and interpretive enquiry, thus a mixed method research (MMR) approach. The study conducted on 91 samples. Data was collected using questionnaire and focus group interview. RESULTS: The delinquency rates in selected schools are very high. In addition, it was found that male learners, more than female learners, exhibit the most antisocial behaviour. In general, both genders, at varying rates disrespect authority, have the habit of lying, engage in theft, vandalism, truancy, substance and alcohol abuse, inappropriate sexual behaviour resulting in teenage pregnancy, and gangsterism, amongst others. The variable of location indicates that schools in formerly disadvantaged areas such as townships, squatter and rural areas experience the most antisocial behaviours by learners, as compared to their counterparts in formerly advantaged areas. Most teachers’ personal encounters of the problem in city schools seemed to be minimal. Some teachers of schools that are situated in middle class areas in townships shared similar sentiments to those of their city counterparts. Although the rate of learner misconduct is alarming, it can be contained as evidenced by behavioural changes witnessed in those learners who are remorseful. This suggests that intervention strategies do play a positive role in curbing unacceptable behaviour. Conclusion: The findings of this study suggested that the majority of teachers in secondary schools are aware of the huge challenge posed by delinquent learners with regard to teaching and learning. The major perceptions the study gathered were that the dawn of a democratic dispensation in 1994 worsened adolescent delinquency and rendered many previously disadvantaged schools dysfunctional
- Published
- 2023
8. Investigation of hospital discharge cases and SARS-CoV-2 introduction into Lothian care homes
- Author
-
S. Cotton, M.P. McHugh, R. Dewar, J.G. Haas, K. Templeton, Samuel C. Robson, Thomas R. Connor, Nicholas J. Loman, Tanya Golubchik, Rocio T. Martinez Nunez, David Bonsall, Andrew Rambaut, Luke B. Snell, Rich Livett, Catherine Ludden, Sally Corden, Eleni Nastouli, Gaia Nebbia, Ian Johnston, Jacqui A. Prieto, Kordo Saeed, David K. Jackson, Catherine Houlihan, Dan Frampton, William L. Hamilton, Adam A. Witney, Giselda Bucca, Cassie F. Pope, Catherine Moore, Emma C. Thomson, Teresa Cutino-Moguel, Ewan M. Harrison, Colin P. Smith, Fiona Rogan, Shaun M. Beckwith, Abigail Murray, Dawn Singleton, Kirstine Eastick, Liz A. Sheridan, Paul Randell, Leigh M. Jackson, Cristina V. Ariani, Sónia Gonçalves, Derek J. Fairley, Matthew W. Loose, Joanne Watkins, Samuel Moses, Sam Nicholls, Matthew Bull, Roberto Amato, Darren L. Smith, David M. Aanensen, Jeffrey C. Barrett, Beatrix Kele, Dinesh Aggarwal, James G. Shepherd, Martin D. Curran, Surendra Parmar, Matthew D. Parker, Catryn Williams, Sharon Glaysher, Anthony P. Underwood, Matthew Bashton, Nicole Pacchiarini, Katie F. Loveson, Matthew Byott, Alessandro M. Carabelli, Kate E. Templeton, Sharon J. Peacock, Thushan I. de Silva, Dennis Wang, Cordelia F. Langford, John Sillitoe, Rory N. Gunson, Simon Cottrell, Justin O’Grady, Dominic Kwiatkowski, Patrick J. Lillie, Nicholas Cortes, Nathan Moore, Claire Thomas, Phillipa J. Burns, Tabitha W. Mahungu, Steven Liggett, Angela H. Beckett, Matthew TG. Holden, Lisa J. Levett, Husam Osman, Mohammed O. Hassan-Ibrahim, David A. Simpson, Meera Chand, Ravi K. Gupta, Alistair C. Darby, Steve Paterson, Oliver G. Pybus, Erik M. Volz, Daniela de Angelis, David L. Robertson, Andrew J. Page, Inigo Martincorena, Louise Aigrain, Andrew R. Bassett, Nick Wong, Yusri Taha, Michelle J. Erkiert, Michael H. Spencer Chapman, Rebecca Dewar, Martin P. McHugh, Siddharth Mookerjee, Stephen Aplin, Matthew Harvey, Thea Sass, Helen Umpleby, Helen Wheeler, James P. McKenna, Ben Warne, Joshua F. Taylor, Yasmin Chaudhry, Rhys Izuagbe, Aminu S. Jahun, Gregory R. Young, Claire McMurray, Clare M. McCann, Andrew Nelson, Scott Elliott, Hannah Lowe, Anna Price, Matthew R. Crown, Sara Rey, Sunando Roy, Ben Temperton, Sharif Shaaban, Andrew R. Hesketh, Kenneth G. Laing, Irene M. Monahan, Judith Heaney, Emanuela Pelosi, Siona Silviera, Eleri Wilson-Davies, Helen Fryer, Helen Adams, Louis du Plessis, Rob Johnson, William T. Harvey, Joseph Hughes, Richard J. Orton, Lewis G. Spurgin, Yann Bourgeois, Chris Ruis, Áine O'Toole, Marina Gourtovaia, Theo Sanderson, Christophe Fraser, Jonathan Edgeworth, Judith Breuer, Stephen L. Michell, John A. Todd, Michaela John, David Buck, Kavitha Gajee, Gemma L. Kay, David Heyburn, Themoula Charalampous, Adela Alcolea-Medina, Katie Kitchman, Alan McNal, David T. Pritch, Samir Dervisevic, Peter Muir, Esther Robinson, Barry B. Vipond, Newara A. Ramadan, Christopher Jeanes, Danni Weldon, Jana Catalan, Neil Jones, Ana da Silva Filipe, Chris Williams, Marc Fuchs, Julia Miskelly, Aaron R. Jeffries, Karen Oliver, Naomi R. Park, Amy Ash, Cherian Koshy, Magdalena Barrow, Sarah L. Buchan, Anna Mantzouratou, Gemma Clark, Christopher W. Holmes, Sharon Campbell, Thomas Davis, Ngee Keong Tan, Julianne R. Brown, Kathryn A. Harris, Stephen P. Kidd, Paul R. Grant, Li Xu-McCrae, Alison Cox, Pinglawathee Madona, Marcus Pond, Paul A. Randell, Karen T. Withell, Cheryl Williams, Clive Graham, Rebecca Denton-Smith, Emma Swindells, Robyn Turnbull, Tim J. Sloan, Andrew Bosworth, Stephanie Hutchings, Hannah M. Pymont, Anna Casey, Liz Ratcliffe, Christopher R. Jones, Bridget A. Knight, Tanzina Haque, Jennifer Hart, Dianne Irish-Tavares, Eric Witele, Craig Mower, Louisa K. Watson DipHE, Jennifer Collins, Gary Eltringham, Dorian Crudgington, Ben Macklin, Miren Iturriza-Gomara, Anita O. Lucaci, Patrick C. McClure, Matthew Carlile, Nadine Holmes, Christopher Moore, Nathaniel Storey, Stefan Rooke, Gonzalo Yebra, Noel Craine, Malorie Perry, Nabil-Fareed Alikhan, Stephen Bridgett, Kate F. Cook, Christopher Fearn, Salman Goudarzi, Ronan A. Lyons, Thomas Williams, Sam T. Haldenby, Jillian Durham, Steven Leonard, Robert M. Davies, Rahul Batra, Beth Blane, Moira J. Spyer, Perminder Smith, Mehmet Yavus, Rachel J. Williams, Adhyana IK. Mahanama, Buddhini Samaraweera, Sophia T. Girgis, Samantha E. Hansford, Angie Green, Charlotte Beaver, Katherine L. Bellis, Matthew J. Dorman, Sally Kay, Liam Prestwood, Shavanthi Rajatileka, Joshua Quick, Radoslaw Poplawski, Nicola Reynolds, Andrew Mack, Arthur Morriss, Thomas Whalley, Bindi Patel, Iliana Georgana, Myra Hosmillo, Malte L. Pinckert, Joanne Stockton, John H. Henderson, Amy Hollis, William Stanley, Wen C. Yew, Richard Myers, Alicia Thornton, Alexander Adams, Tara Annett, Hibo Asad, Alec Birchley, Jason Coombes, Johnathan M. Evans, Laia Fina, Bree Gatica-Wilcox, Lauren Gilbert, Lee Graham, Jessica Hey, Ember Hilvers, Sophie Jones, Hannah Jones, Sara Kumziene-Summerhayes, Caoimhe McKerr, Jessica Powell, Georgia Pugh, Sarah Taylor, Alexander J. Trotter, Charlotte A. Williams, Leanne M. Kermack, Benjamin H. Foulkes, Marta Gallis, Hailey R. Hornsby, Stavroula F. Louka, Manoj Pohare, Paige Wolverson, Peijun Zhang, George MacIntyre-Cockett, Amy Trebes, Robin J. Moll, Lynne Ferguson, Emily J. Goldstein, Alasdair Maclean, Rachael Tomb, Igor Starinskij, Laura Thomson, Joel Southgate, Moritz UG. Kraemer, Jayna Raghwani, Alex E. Zarebski, Olivia Boyd, Lily Geidelberg, Chris J. Illingworth, Chris Jackson, David Pascall, Sreenu Vattipally, Timothy M. Freeman, Sharon N. Hsu, Benjamin B. Lindsey, Keith James, Kevin Lewis, Gerry Tonkin-Hill, Jaime M. Tovar-Corona, MacGregor Cox, Khalil Abudahab, Mirko Menegazzo, Ben EW. Taylor, Corin A. Yeats, Afrida Mukaddas, Derek W. Wright, Leonardo de Oliveira Martins, Rachel Colquhoun, Verity Hill, Ben Jackson, J.T. McCrone, Nathan Medd, Emily Scher, Jon-Paul Keatley, Tanya Curran, Sian Morgan, Patrick Maxwell, Ken Smith, Sahar Eldirdiri, Anita Kenyon, Alison H. Holmes, James R. Price, Tim Wyatt, Alison E. Mather, Timofey Skvortsov, John A. Hartley, Martyn Guest, Christine Kitchen, Ian Merrick, Robert Munn, Beatrice Bertolusso, Jessica Lynch, Gabrielle Vernet, Stuart Kirk, Elizabeth Wastnedge, Rachael Stanley, Giles Idle, Declan T. Bradley, Nicholas F. Killough, Jennifer Poyner, Matilde Mori, Owen Jones, Victoria Wright, Ellena Brooks, Carol M. Churcher, Laia Delgado Callico, Mireille Fragakis, Katerina Galai, Andrew Jermy, Sarah Judges, Anna Markov, Georgina M. McManus, Kim S. Smith, Peter MD. Thomas-McEwen, Elaine Westwick, Stephen W. Attwood, Frances Bolt, Alisha Davies, Elen De Lacy, Fatima Downing, Sue Edwards, Lizzie Meadows, Sarah Jeremiah, Nikki Smith, Luke Foulser, Amita Patel, Louise Berry, Tim Boswell, Vicki M. Fleming, Hannah C. Howson-Wells, Amelia Joseph, Manjinder Khakh, Michelle M. Lister, Paul W. Bird, Karlie Fallon, Thomas Helmer, Claire L. McMurray, Mina Odedra, Jessica Shaw, Julian W. Tang, Nicholas J. Willford, Victoria Blakey, Veena Raviprakash, Nicola Sheriff, Lesley-Anne Williams, Theresa Feltwell, Luke Bedford, James S. Cargill, Warwick Hughes, Jonathan Moore, Susanne Stonehouse, Laura Atkinson, Jack CD. Lee, Divya Shah, Natasha Ohemeng-Kumi, John Ramble, Jasveen Sehmi, Rebecca Williams, Wendy Chatterton, Monika Pusok, William Everson, Anibolina Castigador, Emily Macnaughton, Kate El Bouzidi, Temi Lampejo, Malur Sudhanva, Cassie Breen, Graciela Sluga, Shazaad SY. Ahmad, Ryan P. George, Nicholas W. Machin, Debbie Binns, Victoria James, Rachel Blacow, Lindsay Coupland, Louise Smith, Edward Barton, Debra Padgett, Garren Scott, Aidan Cross, Mariyam Mirfenderesky, Jane Greenaway, Kevin Cole, Phillip Clarke, Nichola Duckworth, Sarah Walsh, Kelly Bicknell, Robert Impey, Sarah Wyllie, Richard Hopes, Chloe Bishop, Vicki Chalker, Ian Harrison, Laura Gifford, Zoltan Molnar, Cressida Auckland, Cariad Evans, Kate Johnson, David G. Partridge, Mohammad Raza, Paul Baker, Stephen Bonner, Sarah Essex, Leanne J. Murray, Andrew I. Lawton, Shirelle Burton-Fanning, Brendan AI. Payne, Sheila Waugh, Andrea N. Gomes, Maimuna Kimuli, Darren R. Murray, Paula Ashfield, Donald Dobie, Fiona Ashford, Angus Best, Liam Crawford, Nicola Cumley, Megan Mayhew, Oliver Megram, Jeremy Mirza, Emma Moles-Garcia, Benita Percival, Megan Driscoll, Leah Ensell, Helen L. Lowe, Laurentiu Maftei, Matteo Mondani, Nicola J. Chaloner, Benjamin J. Cogger, Lisa J. Easton, Hannah Huckson, Jonathan Lewis, Sarah Lowdon, Cassandra S. Malone, Florence Munemo, Manasa Mutingwende, Roberto Nicodemi, Olga Podplomyk FD, Thomas Somassa, Andrew Beggs, Alex Richter, Claire Cormie, Joana Dias, Sally Forrest, Ellen E. Higginson, Mailis Maes, Jamie Young, Rose K. Davidson, Kathryn A. Jackson, Alexander J. Keeley, Jonathan Ball, Timothy Byaruhanga, Joseph G. Chappell, Jayasree Dey, Jack D. Hill, Emily J. Park, Arezou Fanaie, Rachel A. Hilson, Geraldine Yaze, Stephanie Lo, Safiah Afifi, Robert Beer, Joshua Maksimovic, Kathryn McCluggage, Karla Spellman, Catherine Bresner, William Fuller, Angela Marchbank, Trudy Workma, Ekaterina Shelest, Johnny Debebe, Fei Sang, Sarah Francois, Bernardo Gutierrez, Tetyana I. Vasylyeva, Flavia Flaviani, Manon Ragonnet-Cronin, Katherine L. Smollett, Alice Broos, Daniel Mair, Jenna Nichols, Kyriaki Nomikou, Lily Tong, Ioulia Tsatsani, Sarah O'Brien, Steven Rushton, Roy Sanderson, Jon Perkins, Seb Cotton, Abbie Gallagher, Elias Allara, Clare Pearson, David Bibby, Gavin Dabrer, Nicholas Ellaby, Eileen Gallagher, Jonathan Hubb, Angie Lackenby, David Lee, Nikos Manesis, Tamyo Mbisa, Steven Platt, Katherine A. Twohig, Mari Morgan, Alp Aydin, David J. Baker, Ebenezer Foster-Nyarko, Sophie J. Prosolek, Steven Rudder, Chris Baxter, Sílvia F. Carvalho, Deborah Lavin, Arun Mariappan, Clara Radulescu, Aditi Singh, Miao Tang, Helen Morcrette, Nadua Bayzid, Marius Cotic, Carlos E. Balcazar, Michael D. Gallagher, Daniel Maloney, Thomas D. Stanton, Kathleen A. Williamson, Robin Manley, Michelle L. Michelsen, Christine M. Sambles, David J. Studholme, Joanna Warwick-Dugdale, Richard Eccles, Matthew Gemmell, Richard Gregory, Margaret Hughes, Charlotte Nelson, Lucille Rainbow, Edith E. Vamos, Hermione J. Webster, Mark Whitehead, Claudia Wierzbicki, Adrienn Angyal, Luke R. Green, Max Whiteley, Emma Betteridge, Iraad F. Bronner, Ben W. Farr, Scott Goodwin, Stefanie V. Lensing, Shane A. McCarthy, Michael A. Quail, Diana Rajan, Nicholas M. Redshaw, Carol Scott, Lesley Shirley, Scott AJ. Thurston, Will Rowe, Amy Gaskin, Thanh Le-Viet, James Bonfield, Jennifier Liddle, Andrew Whitwham, University of St Andrews. School of Medicine, University of St Andrews. Infection and Global Health Division, and Apollo - University of Cambridge Repository
- Subjects
Microbiology (medical) ,MCC ,Introduction ,SARS-CoV-2 ,NDAS ,COVID-19 ,Care homes ,General Medicine ,NIS ,Patient Discharge ,Hospitals ,Hospitalization ,Infectious Diseases ,SDG 3 - Good Health and Well-being ,RA0421 ,RA0421 Public health. Hygiene. Preventive Medicine ,Humans ,Hospital discharge - Abstract
COG-UK is supported by funding from the MRC part of UK Research & Innovation, the National Institute of Health Research (Grant code MC_PC_19027), and Genome Research Limited, operating as the Welcome Sanger Institute. Background The first epidemic wave of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in Scotland resulted in high case numbers and mortality in care homes. In Lothian, over one-third of care homes reported an outbreak, while there was limited testing of hospital patients discharged to care homes. Aim To investigate patients discharged from hospitals as a source of SARS-CoV-2 introduction into care homes during the first epidemic wave. Methods A clinical review was performed for all patients discharges from hospitals to care homes from 1st March 2020 to 31st May 2020. Episodes were ruled out based on coronavirus disease 2019 (COVID-19) test history, clinical assessment at discharge, whole-genome sequencing (WGS) data and an infectious period of 14 days. Clinical samples were processed for WGS, and consensus genomes generated were used for analysis using Cluster Investigation and Virus Epidemiological Tool software. Patient timelines were obtained using electronic hospital records. Findings In total, 787 patients discharged from hospitals to care homes were identified. Of these, 776 (99%) were ruled out for subsequent introduction of SARS-CoV-2 into care homes. However, for 10 episodes, the results were inconclusive as there was low genomic diversity in consensus genomes or no sequencing data were available. Only one discharge episode had a genomic, time and location link to positive cases during hospital admission, leading to 10 positive cases in their care home. Conclusion The majority of patients discharged from hospitals were ruled out for introduction of SARS-CoV-2 into care homes, highlighting the importance of screening all new admissions when faced with a novel emerging virus and no available vaccine. Publisher PDF
- Published
- 2023
9. 854 HPK1 inhibition relieves suppression downstream of TCR activation to drive enhanced cytokine production and antigen-specific killing, an effect that is further enhanced by immune checkpoint blockade
- Author
-
Rajesh Singh, Bindi Patel, Rameshwari Rayaji, Sachie Marubayashi, Sean Cho, Stefan Garrido-Shaqfeh, Joseph Kulusich, Cesar Meleza, Nidhi Tibrewal, Joice Thomas, Pradeep Nareddy, Ehesan Sharif, Sharon Zhao, Dave Green, Manmohan Leleti, Jay Powers, Matt Walters, and Daniel DiRenzo
- Published
- 2022
10. Cowpea Mosaic Virus (CPMV)-Based Cancer Testis Antigen NY-ESO-1 Vaccine Elicits an Antigen-Specific Cytotoxic T Cell Response
- Author
-
Alan D. Levine, Sourabh Shukla, Chao Wang, Braulio Lorens, Bindi Patel, and Nicole F. Steinmetz
- Subjects
biology ,Biochemistry (medical) ,Cowpea mosaic virus ,Biomedical Engineering ,Cancer ,General Chemistry ,medicine.disease ,biology.organism_classification ,Virology ,Article ,Biomaterials ,Immune system ,Antigen ,medicine ,Cytotoxic T cell ,Cancer/testis antigens ,Cancer vaccine ,NY-ESO-1 - Abstract
Cancer vaccines are promising adjuvant immunotherapies that can stimulate the immune system to recognize tumor-associated antigens and eliminate the residual or recurring disease. The aberrant and restricted expression of highly immunogenic cancer testis antigen NY-ESO-1 in several malignancies, including triple-negative breast cancer, melanoma, myelomas, and ovarian cancer, makes NY-ESO-1 an attractive antigenic target for cancer vaccines. This study describes a NY-ESO-1 vaccine based on a bio-inspired nanomaterial platform technology, specifically a plant virus nanoparticle. The 30 nm icosahedral plant virus cowpea mosaic virus (CPMV) displaying multiple copies of human HLA-A2 restricted peptide antigen NY-ESO-1(157–165) exhibited enhanced uptake and activation of antigen-presenting cells and stimulated a potent CD8(+) T cell response in transgenic human HLA-A2 expressing mice. CD8(+) T cells from immunized mice exhibited antigen-specific proliferation and cancer cell cytotoxicity, highlighting the potential application of a CPMV-NY-ESO-1 vaccine against NY-ESO-1(+) malignancies.
- Published
- 2020
11. Proposed Best Practice Guidelines for Scientific Response Documents: A Consensus Statement from phactMI
- Author
-
Ahsan Jamil, Renee M Johnson, Anne Flanigan-Minnick, Hamza Sarwar, Bindi Patel, Tiffany Huang, Evelyn R. Hermes-DeSantis, Susan Wnorowski, Sonia Kaur Sandhu, Mabel M Cortes, Patrick Reilly, Alysa Redlich, Elissa Vine, Chang Woo Han, and Rita A. Haydar
- Subjects
Statement (computer science) ,Product Labeling ,Consensus ,Evidence-based practice ,Computer science ,Health Personnel ,Best practice ,Section (typography) ,Public Health, Environmental and Occupational Health ,030226 pharmacology & pharmacy ,01 natural sciences ,Transparency (behavior) ,Data science ,010104 statistics & probability ,03 medical and health sciences ,0302 clinical medicine ,Systematic review ,Pharmaceutical Preparations ,Humans ,Pharmacology (medical) ,0101 mathematics ,Adaptation (computer science) ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) - Abstract
The Medical Information Department of a pharmaceutical manufacturer provides written scientific responses to unsolicited requests from healthcare providers for information on products that extends beyond the product labeling (off-label). These scientific response documents are non-promotional, evidence-based, and scientifically balanced, conforming with internal pharmaceutical manufacturer's procedures and the Food and Drug Administration (FDA) Draft Guidance on Responding to Unsolicited Requests for Off-Label Information. Members of phactMI™ developed this proposal to offer best practices for content generation of scientific response documents. Scientific response documents review available literature to respond to an unsolicited request; therefore, they are similar in nature to systematic reviews. The sections and elements identified in this proposed best practice guidelines for scientific response documents are based on an adaptation of the sections and elements of systematic reviews. The sections of a scientific response document should include a restatement of the unsolicited request (title); a structured summary (abstract); approved indications, black box warnings, and background information when appropriate (introduction); the literature search information and study selection (methods); summation of data from clinical trials, meta-analysis, case reports, and/or real world evidence, as appropriate (results); treatment guidelines, if applicable and available (discussion); and references. Elements for each section should be included in a scientific response document as appropriate, as some elements are not necessary in some documents, based on the question. These elements were selected for inclusion to address any potential concerns of bias and transparency and reflect the intent that scientific response documents should be non-promotional, accurate, truthful, free of commercial bias, scientifically balanced, and evidence based.
- Published
- 2020
12. Comparative performance of SARS-CoV-2 lateral flow antigen tests and association with detection of infectious virus in clinical specimens: a single-centre laboratory evaluation study
- Author
-
Sam Douthwaite, Gaia Nebbia, Fiona Reid, Bindi Patel, J Mullen, Themoula Charalampous, P. R. Cliff, Stuart J. D. Neil, Maria Jose Lista, Blair Merrick, Adela Alcolea-Medina, Amita Patel, Emma Cunningham, Jonathan D. Edgeworth, Rahul Batra, Suzanne Pickering, Michael H. Malim, Mark Tan Kia Ik, Luke B Snell, Rui Pedro Galão, and Katie J. Doores
- Subjects
Microbiology (medical) ,Medicine (General) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Population ,Microbiology ,Corrections ,Virus ,R5-920 ,COVID-19 Testing ,Antigen ,Virology ,Genotype ,Medicine ,Humans ,education ,Rapid diagnostic test ,education.field_of_study ,Viral culture ,business.industry ,SARS-CoV-2 ,COVID-19 ,QR1-502 ,Infectious Diseases ,Rapid antigen test ,RNA, Viral ,business - Abstract
Summary Background Lateral flow devices (LFDs) for rapid antigen testing are set to become a cornerstone of SARS-CoV-2 mass community testing, although their reduced sensitivity compared with PCR has raised questions of how well they identify infectious cases. Understanding their capabilities and limitations is, therefore, essential for successful implementation. We evaluated six commercial LFDs and assessed their correlation with infectious virus culture and PCR cycle threshold (Ct) values. Methods In a single-centre, laboratory evaluation study, we did a head-to-head comparison of six LFDs commercially available in the UK: Innova Rapid SARS-CoV-2 Antigen Test, Spring Healthcare SARS-CoV-2 Antigen Rapid Test Cassette, E25Bio Rapid Diagnostic Test, Encode SARS-CoV-2 Antigen Rapid Test Device, SureScreen COVID-19 Rapid Antigen Test Cassette, and SureScreen COVID-19 Rapid Fluorescence Antigen Test. We estimated the specificities and sensitivities of the LFDs using stored naso-oropharyngeal swabs collected at St Thomas' Hospital (London, UK) for routine diagnostic SARS-CoV-2 testing by real-time RT-PCR (RT-rtPCR). Swabs were from inpatients and outpatients from all departments of St Thomas' Hospital, and from health-care staff (all departments) and their household contacts. SARS-CoV-2-negative swabs from the same population (confirmed by RT-rtPCR) were used for comparative specificity determinations. All samples were collected between March 23 and Oct 27, 2020. We determined the limit of detection (LOD) for each test using viral plaque-forming units (PFUs) and viral RNA copy numbers of laboratory-grown SARS-CoV-2. Additionally, LFDs were selected to assess the correlation of antigen test result with RT-rtPCR Ct values and positive viral culture in Vero E6 cells. This analysis included longitudinal swabs from five infected inpatients with varying disease severities. Furthermore, the sensitivities of available LFDs were assessed in swabs (n=23; collected from Dec 4, 2020, to Jan 12, 2021) confirmed to be positive (RT-rtPCR and whole-genome sequencing) for the B.1.1.7 variant, which was the dominant genotype in the UK at the time of study completion. Findings All LFDs showed high specificity (≥98·0%), except for the E25Bio test (86·0% [95% CI 77·9–99·9]), and most tests reliably detected 50 PFU/test (equivalent SARS-CoV-2 N gene Ct value of 23·7, or RNA copy number of 3 × 106/mL). Sensitivities of the LFDs on clinical samples ranged from 65·0% (55·2–73·6) to 89·0% (81·4–93·8). These sensitivities increased to greater than 90% for samples with Ct values of lower than 25 for all tests except the SureScreen fluorescence (SureScreen-F) test. Positive virus culture was identified in 57 (40·4%) of 141 samples; 54 (94·7%) of the positive cultures were from swabs with Ct values lower than 25. Among the three LFDs selected for detailed comparisons (the tests with highest sensitivity [Innova], highest specificity [Encode], and alternative technology [SureScreen-F]), sensitivity of the LFDs increased to at least 94·7% when only including samples with detected viral growth. Longitudinal studies of RT-rtPCR-positive samples (tested with Innova, Encode, and both SureScreen-F and the SureScreen visual [SureScreen-V] test) showed that most of the tests identified all infectious samples as positive. Test performance (assessed for Innova and SureScreen-V) was not affected when reassessed on swabs positive for the UK variant B.1.1.7. Interpretation In this comprehensive comparison of antigen LFDs and virus infectivity, we found a clear relationship between Ct values, quantitative culture of infectious virus, and antigen LFD positivity in clinical samples. Our data support regular testing of target groups with LFDs to supplement the current PCR testing capacity, which would help to rapidly identify infected individuals in situations in which they would otherwise go undetected. Funding King's Together Rapid COVID-19, Medical Research Council, Wellcome Trust, Huo Family Foundation, UK Department of Health, National Institute for Health Research Comprehensive Biomedical Research Centre.
- Published
- 2021
13. Abstract 256: Dual A2aR/A2bR antagonism with etrumadenant (AB928) eliminates the suppressive effects of adenosine on immune and cancer cells in the tumor microenvironment
- Author
-
Sachie Marubayashi, Bindi Patel, Livia Yamashiro, Dana Piovesan, Sean Cho, Jenna Jeffrey, Manmohan Leleti, Jay Powers, Matt Walters, and Daniel DiRenzo
- Subjects
Cancer Research ,Oncology - Abstract
INTRODUCTION: Tumors employ many strategies to attenuate immune responses. High levels of extracellular adenosine generated in the tumor microenvironment engage A2a and A2b adenosine receptors on immune cells, resulting in immunosuppression. Expression of A2aR and A2bR can vary by cell type, with T cells predominantly expressing A2aR while myeloid cells express both A2aR and A2bR. We have previously shown that etrumadenant, a dual A2aR/A2bR antagonist, blocks the immunosuppressive effects of adenosine in immune cells and enhances anti-tumor immune responses in mouse syngeneic tumors. Using dual and selective adenosine receptor antagonists, we assessed the contribution of these receptors to adenosine-mediated phenotypes in immune and cancer cells. METHODS: Human CD8+ T cells were isolated from healthy human blood and activated using CD3/CD28/CD2 stimulation and cytokines were analyzed by cytokine bead array at 72 hours. Primary human dendritic cells (DC) were isolated from healthy blood and matured with LPS/IFN-γ for 24 hours. Cancer cell lines were purchased from ATCC. The adenosine analogue NECA was used to stimulate A2aR/A2bR-mediated signaling. RESULTS: Activated human CD8+ T cells stimulated in the presence of NECA showed suppression of activation markers (CD69) and cytokine production (IFN-γ, IL-2 and granzyme B). As expected, we observed similar rescue of this phenotype with both etrumadenant and an A2aR-specific antagonist owing to the sole expression of A2aR on T cells. In contrast, primary DC have comparable expression of A2aR and A2bR, suggesting that dual blockade may provide greater resistance to adenosine-mediated suppression than A2aR antagonism. Indeed, etrumadenant was able to attenuate the adenosine-mediated upregulation of IL-10 and enhance IL-12p70 production, whereas a comparable A2aR-specific antagonist showed no significant rescue versus NECA-stimulated controls. These observations may be extended to suppressive myeloid populations as well as tumor-resident macrophages and myeloid-derived suppressor cells isolated from mouse syngeneic tumors, which have very high expression of both A2aR and A2bR. RNA-sequencing identified a cassette of genes regulated by adenosine-signaling in these cells, which were largely reversed by etrumadenant. Finally, cultured human cancer cell lines have high expression of A2bR, which has been implicated in driving their tumorigenesis. Etrumadenant reversed adenosine-stimulated gene expression changes in non-small cell lung cancer cell lines, restoring enriched pathways driven by adenosine signaling. CONCLUSIONS: Taken together, these results show an important role for A2aR/A2bR in adenosine-mediated immunosuppression and provide a mechanistic rationale for stimulation of anti-tumor immune responses with the dual adenosine receptor antagonist etrumadenant. Citation Format: Sachie Marubayashi, Bindi Patel, Livia Yamashiro, Dana Piovesan, Sean Cho, Jenna Jeffrey, Manmohan Leleti, Jay Powers, Matt Walters, Daniel DiRenzo. Dual A2aR/A2bR antagonism with etrumadenant (AB928) eliminates the suppressive effects of adenosine on immune and cancer cells in the tumor microenvironment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 256.
- Published
- 2022
14. Acetylated tau inhibits chaperone-mediated autophagy and promotes tau pathology propagation in mice
- Author
-
Peter Dongmin Sohn, Bindi Patel, Yves R. Juste, Chao Wang, Alison Goate, Bradley T. Hyman, Mathieu Bourdenx, Celeste M. Karch, Jose A. Rodriguez-Navarro, Susanne Wegmann, Antonio Diaz, Enrique Luengo, Jason E. Gestwicki, Li Gan, Hao Shao, Zapporah T. Young, Ana Maria Cuervo, Manuela G. López, Szu Yu Kuo, Xu Chen, and Benjamin Caballero
- Subjects
pathology [Tauopathies] ,0301 basic medicine ,Male ,Aging ,physiopathology [Tauopathies] ,General Physics and Astronomy ,Chaperone-Mediated Autophagy ,Neurodegenerative ,Inbred C57BL ,Alzheimer's Disease ,Mice ,0302 clinical medicine ,Chaperone-mediated autophagy ,2.1 Biological and endogenous factors ,Aetiology ,Alzheimer's Disease Related Dementias (ADRD) ,health care economics and organizations ,Neurons ,Multidisciplinary ,Chemistry ,Neurodegeneration ,Brain ,Acetylation ,Neural ageing ,humanities ,Cell biology ,Frontotemporal Dementia (FTD) ,Tauopathies ,metabolism [Neurons] ,Neurological ,Female ,ddc:500 ,Tauopathy ,Endosome ,Science ,tau Proteins ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cellular neuroscience ,mental disorders ,medicine ,Acquired Cognitive Impairment ,Animals ,Humans ,Microautophagy ,Autophagy ,genetics [Tauopathies] ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,General Chemistry ,medicine.disease ,metabolism [tau Proteins] ,Brain Disorders ,Mice, Inbred C57BL ,genetics [tau Proteins] ,030104 developmental biology ,metabolism [Brain] ,Dementia ,metabolism [Tauopathies] ,030217 neurology & neurosurgery - Abstract
Disrupted homeostasis of the microtubule binding protein tau is a shared feature of a set of neurodegenerative disorders known as tauopathies. Acetylation of soluble tau is an early pathological event in neurodegeneration. In this work, we find that a large fraction of neuronal tau is degraded by chaperone-mediated autophagy (CMA) whereas, upon acetylation, tau is preferentially degraded by macroautophagy and endosomal microautophagy. Rerouting of acetylated tau to these other autophagic pathways originates, in part, from the inhibitory effect that acetylated tau exerts on CMA and results in its extracellular release. In fact, experimental blockage of CMA enhances cell-to-cell propagation of pathogenic tau in a mouse model of tauopathy. Furthermore, analysis of lysosomes isolated from brains of patients with tauopathies demonstrates similar molecular mechanisms leading to CMA dysfunction. This study reveals that CMA failure in tauopathy brains alters tau homeostasis and could contribute to aggravate disease progression., The tau protein has been implicated in neurodegenerative disorders and can propagate from cell to cell. Here, the authors show that tau acetylation reduces its degradation by chaperone-mediated autophagy, causing re-routing to other autophagic pathways and increasing extracellular tau release.
- Published
- 2021
15. Targeting retinoic acid receptor alpha-corepressor interaction activates chaperone-mediated autophagy and protects against retinal degeneration
- Author
-
Raquel Gomez-Sintes, Qisheng Xin, Juan Ignacio Jimenez-Loygorri, Mericka McCabe, Antonio Diaz, Thomas P. Garner, Xiomaris M. Cotto-Rios, Yang Wu, Shuxian Dong, Cara A. Reynolds, Bindi Patel, Pedro de la Villa, Fernando Macian, Patricia Boya, Evripidis Gavathiotis, Ana Maria Cuervo, National Institutes of Health (US), JPB Foundation, Michael J. Fox Foundation for Parkinson's Research, Rainwater Charitable Foundation, Backus Foundation, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), European Commission, Gómez-Sintes, Raquel, Jiménez-Loygorri, Juan Ignacio, McCabe, Mericka, Díaz, Antonio, Cotto-Rios, Xiomaris M., Reynolds, Cara A., De la Villa, Pedro, Macian, Fernando, Boya, Patricia, Gavathiotis, Evripidis, and Cuervo, Ana Maria
- Subjects
Mice ,Multidisciplinary ,Retinoic Acid Receptor alpha ,Retinal Degeneration ,Autophagy ,General Physics and Astronomy ,Animals ,Chaperone-Mediated Autophagy ,General Chemistry ,Co-Repressor Proteins ,General Biochemistry, Genetics and Molecular Biology ,Retinitis Pigmentosa - Abstract
18 p.-8 fig., Chaperone-mediated autophagy activity, essential in the cellular defense against proteotoxicity, declines with age, and preventing this decline in experimental genetic models has proven beneficial. Here, we have identified the mechanism of action of selective chaperone-mediated autophagy activators previously developed by our group and have leveraged that information to generate orally bioavailable chaperone-mediated autophagy activators with favorable brain exposure. Chaperone-mediated autophagy activating molecules stabilize the interaction between retinoic acid receptor alpha - a known endogenous inhibitor of chaperone-mediated autophagy - and its co-repressor, nuclear receptor corepressor 1, resulting in changes of a discrete subset of the retinoic acid receptor alpha transcriptional program that leads to selective chaperone-mediated autophagy activation. Chaperone-mediated autophagy activators molecules activate this pathway in vivo and ameliorate retinal degeneration in a retinitis pigmentosa mouse model. Our findings reveal a mechanism for pharmacological targeting of chaperone-mediated autophagy activation and suggest a therapeutic strategy for retinal degeneration., This work was supported by National Institutes of Health grants AG054108, AG021904, and AG017617 (to AMC) and AG038072 (to AMC, FM, and EG), the JPB Foundation (to AMC), the Michael J. Fox Foundation (to AMC and EG), the Rainwater Charitable Foundation (to AMC and EG) and the Backus Foundation (to AMC). Chemical synthesis, NMR and MS data were in part supported by NIH awards P30 CA013330 and 1S10OD016305. Research in PB and RGS lab is funded by Ministerio de Ciencia, Innovación y Universidades (MCIU), Agencia Estatal de Investigación (AEI) and Fondo Europeo de Desarrollo Regional (FEDER) PGC2018-098557-B-I00 to PB and RTI2018-098990-J-I00 to RGS. CAR was supported by T32 GM007491 and MM by T32 AG023475.
- Published
- 2021
16. The negative effect of lipid challenge on autophagy inhibits T cell responses
- Author
-
Cristina C. Clement, Cara A. Reynolds, Ana Maria Cuervo, Bindi Patel, Ignacio Guerrero-Ros, Laura Santambrogio, and Fernando Macian
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,T cell ,MAPK8 ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,Down-Regulation ,Biology ,Diet, High-Fat ,Lymphocyte Activation ,03 medical and health sciences ,medicine ,Autophagy ,Animals ,Homeostasis ,Humans ,Obesity ,Protein kinase A ,Molecular Biology ,Mechanistic target of rapamycin ,Cell Proliferation ,030102 biochemistry & molecular biology ,Cell Biology ,T helper cell ,Lipids ,Cell biology ,Calcineurin ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Cytokines ,Female ,MAP1LC3B ,Oleic Acid ,Research Paper - Abstract
Obesity is associated with changes in the immune system that significantly hinder its ability to mount efficient immune responses. Previous studies have reported a dysregulation of immune responses caused by lipid challenge; however, the mechanisms underlying that dysregulation are still not completely understood. Autophagy is an essential catabolic process through which cellular components are degraded by the lysosomal machinery. In T cells, autophagy is an actively regulated process necessary to sustain homeostasis and activation. Here, we report that CD4(+) T cell responses are inhibited when cells are challenged with increasing concentrations of fatty acids. Furthermore, analysis of T cells from diet-induced obese mice confirms that high lipid load inhibits activation-induced responses in T cells. We have found that autophagy is inhibited in CD4(+) T cells exposed in vitro or in vivo to lipid stress, which causes decreased autophagosome formation and degradation. Supporting that inhibition of autophagy caused by high lipid load is a key mechanism that accounts for the effects on T cell function of lipid stress, we found that ATG7 (autophagy-related 7)-deficient T cells, unable to activate autophagy, did not show additional inhibitory effects on their responses to activation when subjected to lipid challenge. Our results indicate, thus, that increased lipid load can dysregulate autophagy and cause defective T cell responses, and suggest that inhibition of autophagy may underlie some of the characteristic obesity-associated defects in the T cell compartment. Abbreviations: ACTB: actin, beta; ATG: autophagy-related; CDKN1B: cyclin-dependent kinase inhibitor 1B; HFD: high-fat diet; IFNG: interferon gamma; IL: interleukin; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; MAPK8/JNK: mitogen-activated protein kinase 8; LC3-I: non-conjugated form of MAP1LC3B; LC3-II: phosphatidylethanolamine-conjugated form of MAP1LC3B; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; MS: mass spectrometry; MTOR: mechanistic target of rapamycin kinase; NFATC2: nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 2; NLRP3: NLR family, pyrin domain containing 3; OA: oleic acid; PI: propidium iodide; ROS: reactive oxygen species; STAT5A: signal transducer and activator of transcription 5A; TCR: T cell receptor; T(H)1: T helper cell type 1
- Published
- 2019
17. Selective endosomal microautophagy is starvation-inducible inDrosophila
- Author
-
Ana Maria Cuervo, Anindita Mukherjee, Bindi Patel, Andreas Jenny, and Hiroshi Koga
- Subjects
0301 basic medicine ,Atg1 ,Amino Acid Motifs ,ATG5 ,Cellular homeostasis ,Endosomes ,Biology ,ATG12 ,03 medical and health sciences ,0302 clinical medicine ,Chaperone-mediated autophagy ,Autophagy ,Animals ,Drosophila Proteins ,Microautophagy ,Molecular Biology ,fungi ,Multivesicular Bodies ,Cell Biology ,Autophagy-related protein 13 ,Basic Research Paper ,Cell biology ,Drosophila melanogaster ,030104 developmental biology ,Starvation ,Lysosomes ,Biomarkers ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Autophagy delivers cytosolic components to lysosomes for degradation and is thus essential for cellular homeostasis and to cope with different stressors. As such, autophagy counteracts various human diseases and its reduction leads to aging-like phenotypes. Macroautophagy (MA) can selectively degrade organelles or aggregated proteins, whereas selective degradation of single proteins has only been described for chaperone-mediated autophagy (CMA) and endosomal microautophagy (eMI). These 2 autophagic pathways are specific for proteins containing KFERQ-related targeting motifs. Using a KFERQ-tagged fluorescent biosensor, we have identified an eMI-like pathway in Drosophila melanogaster. We show that this biosensor localizes to late endosomes and lysosomes upon prolonged starvation in a KFERQ- and Hsc70-4- dependent manner. Furthermore, fly eMI requires endosomal multivesicular body formation mediated by ESCRT complex components. Importantly, induction of Drosophila eMI requires longer starvation than the induction of MA and is independent of the critical MA genes atg5, atg7, and atg12. Furthermore, inhibition of Tor signaling induces eMI in flies under nutrient rich conditions, and, as eMI in Drosophila also requires atg1 and atg13, our data suggest that these genes may have a novel, additional role in regulating eMI in flies. Overall, our data provide the first evidence for a novel, starvation-inducible, catabolic process resembling endosomal microautophagy in the Drosophila fat body.
- Published
- 2016
18. Physician-reported comorbidities and treatment management in patients with non-metastatic castration-resistant prostate cancer
- Author
-
Jacqueline Parkin, Dena H. Jaffe, Jonathon Wright, Bindi Patel, Annissa Cyhaniuk, and Sreevalsa Appukkuttan
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Castration resistant ,medicine.disease ,Treatment management ,Prostate cancer ,Internal medicine ,medicine ,Non metastatic ,In patient ,business - Abstract
149 Background: With the emergence of potent therapies in non-metastatic castration-resistant prostate cancer (nmCRPC) there is a need to understand the impact of nmCRPC treatments on patient comorbidities and concomitant medications. The goal of this study was to understand treatment management of nmCRPC in patients with pre-existing comorbidities from a physician perspective. Methods: Physicians who treated nmCRPC patients with systemic therapy were recruited from a US physician panel for an online survey (Sept-Oct 2019). Physician responses included physician treatment practice, demographic characteristics, and their 'typical' nmCRPC patient profile from the past 6 months (e.g., health profile, disease management, and quality of life [QOL]). Results: Fifty US physicians (56% urologists, 44% oncologists) with 21±6 years in practice, treated on average 30 nmCRPC patients in the past 6 months. The most common nmCRPC treatments were leuprolide acetate (82%), enzalutamide (80%) and apalutamide (70%). The most common comorbidities reported were hypertension (96%), sexual dysfunction (94%), diabetes (92%), myocardial infarction (88%) and urinary issues (88%). 78% of the physicians reported taking comorbidities and medications for comorbidities into consideration when prescribing nmCRPC treatments. Between 15%-28% of physicians reported a change in nmCRPC prostate treatment and 19%-26% reported a dose change in nmCRPC treatment for up to 1/3 of their patients due to comorbidities (Table). For QOL, urologists versus oncologists indicated more days with poor health status among nmCRPC patients (e.g., median poor mental health days 30-days prior to treatment: urologists=15 days vs oncologists=5 days). Conclusions: Many physicians take into account pre-existing comorbidities and their medications when prescribing nmCRPC treatments. Differences in perceived QOL were observed between physician specialty. These findings highlight the importance of considering therapies that lessen the treatment burden in nmCRPC. [Table: see text]
- Published
- 2020
19. Defective recruitment of motor proteins to autophagic compartments contributes to autophagic failure in aging
- Author
-
David Yin, Allan W. Wolkoff, Eloy Bejarano, Xintao Wang, Olatz Pampliega, Andrea Yuste, John W. Murray, Bindi Patel, and Ana Maria Cuervo
- Subjects
0301 basic medicine ,Male ,autophagy ,Aging ,Dynein ,Kinesins ,Biology ,molecular motors ,Motor protein ,03 medical and health sciences ,Mice ,lysosomes ,Microtubule ,Molecular motor ,Animals ,Loss function ,Cellular Senescence ,vesicles ,dynein ,Autophagy ,Cell Biology ,Original Articles ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Proteostasis ,Original Article ,intracellular traffic ,autophagosomes ,Intracellular - Abstract
Summary Inability to preserve proteostasis with age contributes to the gradual loss of function that characterizes old organisms. Defective autophagy, a component of the proteostasis network for delivery and degradation of intracellular materials in lysosomes, has been described in multiple old organisms, while a robust autophagy response has been linked to longevity. The molecular mechanisms responsible for defective autophagic function with age remain, for the most part, poorly characterized. In this work, we have identified differences between young and old cells in the intracellular trafficking of the vesicular compartments that participate in autophagy. Failure to reposition autophagosomes and lysosomes toward the perinuclear region with age reduces the efficiency of their fusion and the subsequent degradation of the sequestered cargo. Hepatocytes from old mice display lower association of two microtubule‐based minus‐end‐directed motor proteins, the well‐characterized dynein, and the less‐studied KIFC3, with autophagosomes and lysosomes, respectively. Using genetic approaches to mimic the lower levels of KIFC3 observed in old cells, we confirmed that reduced content of this motor protein in fibroblasts leads to failed lysosomal repositioning and diminished autophagic flux. Our study connects defects in intracellular trafficking with insufficient autophagy in old organisms and identifies motor proteins as a novel target for future interventions aiming at correcting autophagic activity with anti‐aging purposes.
- Published
- 2018
20. Common γ-chain cytokine signaling is required for macroautophagy induction during CD4+ T-cell activation
- Author
-
Fernando Macian, Bindi Patel, and Yair Botbol
- Subjects
CD4-Positive T-Lymphocytes ,Interleukin 2 ,Cell signaling ,medicine.medical_treatment ,T cell ,Cell Communication ,Biology ,Lymphocyte Activation ,Downregulation and upregulation ,Lysosome ,Autophagy ,medicine ,Animals ,Phosphorylation ,Molecular Biology ,Cell Biology ,Basic Research Paper ,Cell biology ,Mice, Inbred C57BL ,Cytokine ,medicine.anatomical_structure ,Cytokines ,Female ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
Macroautophagy is a cellular process that mediates degradation in the lysosome of cytoplasmic components including proteins and organelles. Previous studies have shown that macroautophagy is induced in activated T cells to regulate organelle homeostasis and the cell's energy metabolism. However, the signaling pathways that initiate and regulate activation-induced macroautophagy in T cells have not been identified. Here, we show that activation-induced macroautophagy in T cells depends on signaling from common γ-chain cytokines. Consequently, inhibition of signaling through JAK3, induced downstream of cytokine receptors containing the common γ-chain, prevents full induction of macroautophagy in activated T cells. Moreover, we found that common γ-chain cytokines are not only required for macroautophagy upregulation during T cell activation but can themselves induce macroautophagy. Our data also show that macroautophagy induction in T cells is associated with an increase of LC3 expression that is mediated by a post-transcriptional mechanism. Overall, our findings unveiled a new role for common γ-chain cytokines as a molecular link between autophagy induction and T-cell activation.
- Published
- 2015
21. Lysosomal mTORC2/PHLPP1/Akt Regulate Chaperone-Mediated Autophagy
- Author
-
Hiroshi Koga, Ana Maria Cuervo, Bindi Patel, Esperanza Arias, Enric Mocholi, and Antonio Diaz
- Subjects
Male ,animal structures ,Phosphatase ,Mechanistic Target of Rapamycin Complex 2 ,Biology ,mTORC2 ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Chaperone-mediated autophagy ,Cell Line, Tumor ,Autophagy ,Phosphoprotein Phosphatases ,Animals ,Humans ,Rats, Wistar ,Molecular Biology ,Protein kinase B ,health care economics and organizations ,030304 developmental biology ,0303 health sciences ,TOR Serine-Threonine Kinases ,Nuclear Proteins ,Cell Biology ,humanities ,Rats ,3. Good health ,Mice, Inbred C57BL ,Cytosol ,Proteostasis ,Liver ,Biochemistry ,Gene Knockdown Techniques ,Multiprotein Complexes ,030220 oncology & carcinogenesis ,Chaperone (protein) ,Proteolysis ,NIH 3T3 Cells ,biology.protein ,Lysosomes ,Proto-Oncogene Proteins c-akt ,Molecular Chaperones - Abstract
Chaperone-mediated autophagy (CMA), a selective form of degradation of cytosolic proteins in lysosomes, contributes to maintenance of proteostasis and to the cellular adaptation to stress. CMA substrates are delivered by a cytosolic chaperone to the lysosomal surface, where, upon unfolding, they are internalized through a membrane translocation complex. The molecular components that participate in CMA substrate targeting and translocation are well characterized, but those involved in CMA regulation remain mostly unknown. In this study, we have identified that CMA is under the positive control of the phosphatase PHLPP1 that associates with the lysosomal membrane and counteracts the inhibitory effect of mTORC2 on CMA. Lysosomal Akt, a target of the mTORC2/PHLPP1 kinase-phosphatase pair, modulates CMA activity by controlling the dynamics of assembly and disassembly of the CMA translocation complex at the lysosomal membrane. The lysosomal mTORC2/PHLPP1/Akt axis could become a target to restore CMA dysfunction in aging and disease.
- Published
- 2015
22. Methods to study chaperone-mediated autophagy
- Author
-
Ana Maria Cuervo and Bindi Patel
- Subjects
medicine.diagnostic_test ,Proteolysis ,Organelle lumen ,Neurodegeneration ,Autophagy ,Biology ,medicine.disease ,Article ,humanities ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Cytosol ,Chaperone-mediated autophagy ,Biochemistry ,Proteome ,medicine ,Humans ,Lysosomes ,Receptor ,Molecular Biology ,health care economics and organizations ,Molecular Chaperones - Abstract
Chaperone-mediated autophagy (CMA) is a multistep process that involves selective degradation and digestion of a pool of soluble cytosolic proteins in lysosomes. Cytosolic substrates are selectively identified and targeted by chaperones to lysosomes where they are subsequently translocated into the organelle lumen through a dedicated CMA-associated lysosomal membrane receptor/translocation complex. CMA contributes to maintaining a functional proteome, through elimination of altered proteins, and participates in the cellular energetic balance through amino acid recycling. Defective or dysfunctional CMA has been associated with human pathologies such as neurodegeneration, cancer, immunodeficiency or diabetes, increasing the overall interest in methods to monitor this selective autophagic pathway. Here, we describe approaches used to study CMA in different experimental models.
- Published
- 2015
23. Huntingtin functions as a scaffold for selective macroautophagy
- Author
-
Zhen Xu, Erin Furr Stimming, Gabriela David, Zhihua Chen, Hugo J. Bellen, Bindi Patel, Yamin Sun, Antonio Tito, Dongsheng Chen, Ana Maria Cuervo, Sheng Zhang, and Yanning Rui
- Subjects
Autophagosome ,Scaffold protein ,congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Nerve Tissue Proteins ,Protein Serine-Threonine Kinases ,Biology ,BAG3 ,03 medical and health sciences ,0302 clinical medicine ,Phagosomes ,mental disorders ,Autophagy ,Huntingtin Protein ,Animals ,Autophagy-Related Protein-1 Homolog ,Drosophila Proteins ,Humans ,030304 developmental biology ,0303 health sciences ,Ubiquitin ,TOR Serine-Threonine Kinases ,Intracellular Signaling Peptides and Proteins ,Ubiquitination ,RNA-Binding Proteins ,Cell Biology ,ULK1 ,nervous system diseases ,Cell biology ,Drosophila melanogaster ,HEK293 Cells ,Gene Expression Regulation ,Microtubule-Associated Proteins ,030217 neurology & neurosurgery ,HeLa Cells ,Protein Binding ,Signal Transduction - Abstract
Selective macroautophagy is an important protective mechanism against diverse cellular stresses. In contrast to the well-characterized starvation-induced autophagy, the regulation of selective autophagy is largely unknown. Here, we demonstrate that Huntingtin, the Huntington disease gene product, functions as a scaffold protein for selective macroautophagy but it is dispensable for non-selective macroautophagy. In Drosophila, Huntingtin genetically interacts with autophagy pathway components. In mammalian cells, Huntingtin physically interacts with the autophagy cargo receptor p62 to facilitate its association with the integral autophagosome component LC3 and with Lys-63-linked ubiquitin-modified substrates. Maximal activation of selective autophagy during stress is attained by the ability of Huntingtin to bind ULK1, a kinase that initiates autophagy, which releases ULK1 from negative regulation by mTOR. Our data uncover an important physiological function of Huntingtin and provide a missing link in the activation of selective macroautophagy in metazoans.
- Published
- 2015
24. Loss of hepatic chaperone-mediated autophagy accelerates proteostasis failure in aging
- Author
-
Aleksandra M. Urbanska, Joan Villarroya, Jaime L. Schneider, Bindi Patel, Ana Maria Cuervo, Mia M. Thi, Francesc Villarroya, Laura Santambrogio, and Antonio Diaz-Carretero
- Subjects
Male ,autophagy ,Aging ,Protein aggregation ,Biology ,medicine.disease_cause ,Protein Aggregation, Pathological ,protein aggregation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Chaperone-mediated autophagy ,medicine ,Animals ,Homeostasis ,Humans ,lysosomal protein degradation ,health care economics and organizations ,030304 developmental biology ,0303 health sciences ,Autophagy ,proteotoxicity ,Cell Biology ,Original Articles ,humanities ,Cell biology ,Mice, Inbred C57BL ,macroautophagy ,Oxidative Stress ,Proteostasis ,Proteotoxicity ,Proteasome ,Liver ,ubiquitin-proteasome system ,Lysosomes ,030217 neurology & neurosurgery ,Oxidative stress ,Molecular Chaperones - Abstract
Chaperone-mediated autophagy (CMA), a cellular process that contributes to protein quality control through targeting of a subset of cytosolic proteins to lysosomes for degradation, undergoes a functional decline with age. We have used a mouse model with liver-specific defective CMA to identify changes in proteostasis attributable to reduced CMA activity in this organ with age. We have found that other proteolytic systems compensate for CMA loss in young mice which helps to preserve proteostasis. However, these compensatory responses are not sufficient for protection against proteotoxicity induced by stress (oxidative stress, lipid challenges) or associated with aging. Livers from old mice with CMA blockage exhibit altered protein homeostasis, enhanced susceptibility to oxidative stress and hepatic dysfunction manifested by a diminished ability to metabolize drugs, and a worsening of the metabolic dysregulation identified in young mice. Our study reveals that while the regulatory function of CMA cannot be compensated for in young organisms, its contribution to protein homeostasis can be handled by other proteolytic systems. However, the decline in the compensatory ability identified with age explains the more severe consequences of CMA impairment in older organisms and the contribution of CMA malfunction to the gradual decline in proteostasis and stress resistance observed during aging.
- Published
- 2015
25. Store-operated Ca2+ entry (SOCE) controls induction of lipolysis and the transcriptional reprogramming to lipid metabolism
- Author
-
Mario Ćuk, Mate Maus, Hue Tran Hornig-Do, Bindi Patel, Kathryn J. Moore, Mireille Ouimet, Stefan Feske, Zofia M.A. Chrzanowska-Lightowlers, Ana Maria Cuervo, Ulrike Kaufmann, Rita Horvath, Jun Yang, and Jayson Lian
- Subjects
0301 basic medicine ,Transcription, Genetic ,Physiology ,Lipolysis ,Biology ,Article ,03 medical and health sciences ,Mice ,Lipid droplet ,Animals ,Humans ,PPAR alpha ,Muscle, Skeletal ,Molecular Biology ,Beta oxidation ,Fatty Acids ,Lipid metabolism ,STIM1 ,Cell Biology ,STIM2 ,Lipase ,Lipid Droplets ,Peroxisome ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Mitochondria ,Up-Regulation ,030104 developmental biology ,HEK293 Cells ,Biochemistry ,Lipotoxicity ,Calcium ,Oxidation-Reduction ,Adenylyl Cyclases ,Signal Transduction - Abstract
Ca2+ signals were reported to control lipid homeostasis, but the Ca2+ channels and pathways involved are largely unknown. Store-operated Ca2+ entry (SOCE) is a ubiquitous Ca2+ influx pathway regulated by stromal interaction molecule 1 (STIM1), STIM2, and the Ca2+ channel ORAI1. We show that SOCE-deficient mice accumulate pathological amounts of lipid droplets in the liver, heart, and skeletal muscle. Cells from patients with loss-of-function mutations in STIM1 or ORAI1 show a similar phenotype, suggesting a cell-intrinsic role for SOCE in the regulation of lipid metabolism. SOCE is crucial to induce mobilization of fatty acids from lipid droplets, lipolysis, and mitochondrial fatty acid oxidation. SOCE regulates cyclic AMP production and the expression of neutral lipases as well as the transcriptional regulators of lipid metabolism, peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α), and peroxisome proliferator-activated receptor α (PPARα). SOCE-deficient cells upregulate lipophagy, which protects them from lipotoxicity. Our data provide evidence for an important role of SOCE in lipid metabolism.
- Published
- 2017
26. Functional interaction between autophagy and ciliogenesis
- Author
-
Idil Orhon, Antonio Diaz-Carretero, Isabelle Beau, Patrice Codogno, Bindi Patel, Peter Satir, Birgit H. Satir, Sunandini Sridhar, Olatz Pampliega, and Ana Maria Cuervo
- Subjects
0303 health sciences ,Multidisciplinary ,Cilium ,Autophagy ,Nutrient sensing ,Biology ,BAG3 ,Hedgehog signaling pathway ,Article ,Cell biology ,Transport protein ,03 medical and health sciences ,0302 clinical medicine ,primary cilia ,lysosomes ,Intraflagellar transport ,Ciliogenesis ,autophagosomes ,vesicular trafficking ,030217 neurology & neurosurgery ,030304 developmental biology ,intraflagellar transport proteins - Abstract
Summary Nutrient deprivation is a stimulus shared by both autophagy and the formation of primary cilia. The recently discovered role of primary cilia in nutrient sensing and signaling motivated us to explore the possible functional interactions between this signaling hub and autophagy. Here we show that part of the molecular machinery involved in ciliogenesis also participates in the early steps of the autophagic process. Signaling from the cilia, such as that from the Hedgehog pathway, induces autophagy by acting directly on essential autophagy-related proteins strategically located in the base of the cilium by ciliary trafficking proteins. While abrogation of ciliogenesis partially inhibits autophagy, blockage of autophagy enhances primary cilia growth and cilia-associated signaling during normal nutritional conditions. We propose that basal autophagy regulates ciliary growth through the degradation of proteins required for intraflagellar transport. Compromised ability to activate the autophagic response may underlie the basis of some common ciliopathies.
- Published
- 2013
27. Effects of Sex, Strain, and Energy Intake on Hallmarks of Aging in Mice
- Author
-
Ahmed Ali, Miguel Calvo-Rubio, R. Michael Anson, T. Mark Beasley, Vincent Guiterrez, John Dawson, Vilhelm A. Bohr, Pinchas Cohen, Devin Wahl, Dan L. Longo, Ana Maria Cuervo, David W. Frederick, Rafael de Cabo, M. I. Burón, Yongqing Zhang, Joseph A. Baur, Donald K. Ingram, Kevin J. Pearson, James R. Mitchell, José A. González-Reyes, Evandro Fei Fang, José M. Villalba, Christopher Hine, Julio Madrigal-Matute, Kevin G. Becker, Morten Scheibye-Knudsen, Gene B. Hubbard, Yuji Ikeno, Marta Gonzalez-Freire, Michel Bernier, Sarah J. Mitchell, Frank Madeo, Lukas Habering, David B. Allison, Susmita Kaushik, Junxiang Wan, Theresa M. Ward, Josephine M. Egan, Bindi Patel, Miguel A. Aon, Sonia Cortassa, Huan Cai, Plácido Navas, Luigi Ferrucci, David A. Sinclair, Hector H. Palacios, Filomena Broeskamp, Ministerio de Economía y Competitividad (España), Consejo Superior de Investigaciones Científicas (España), National Institutes of Health (US), American Diabetes Association, Research Foundation - Flanders, Federal Ministry of Education and Research (Germany), and Universidad de Córdoba (España)
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Aging ,Proteasome Endopeptidase Complex ,Physiology ,Calorie restriction ,Longevity ,Context (language use) ,Mice, Inbred Strains ,Biology ,Carbohydrate metabolism ,Mitochondrion ,Article ,03 medical and health sciences ,Islets of Langerhans ,Mice ,Internal medicine ,medicine ,Autophagy ,Animals ,Cluster Analysis ,Homeostasis ,Metabolomics ,Hydrogen Sulfide ,Molecular Biology ,Caloric Restriction ,Sex Characteristics ,Ubiquitin ,Gene Expression Profiling ,Cell Biology ,Mitochondria ,030104 developmental biology ,Endocrinology ,Proteostasis ,Glucose ,Phenotype ,Gene Expression Regulation ,Liver ,Immunology ,Metabolome ,Female ,Energy Intake ,Biomarkers ,Sex characteristics - Abstract
Author manuscript; available in PMC 2017 June 14.-- et al., Calorie restriction (CR) is the most robust non-genetic intervention to delay aging. However, there are a number of emerging experimental variables that alter CR responses. We investigated the role of sex, strain, and level of CR on health and survival in mice. CR did not always correlate with lifespan extension, although it consistently improved health across strains and sexes. Transcriptional and metabolomics changes driven by CR in liver indicated anaplerotic filling of the Krebs cycle together with fatty acid fueling of mitochondria. CR prevented age-associated decline in the liver proteostasis network while increasing mitochondrial number, preserving mitochondrial ultrastructure and function with age. Abrogation of mitochondrial function negated life-prolonging effects of CR in yeast and worms. Our data illustrate the complexity of CR in the context of aging, with a clear separation of outcomes related to health and survival, highlighting complexities of translation of CR into human interventions., This work was supported in part by the Intramural Research Program of the National Institute on Aging, NIH, and by NIH grants R01 AG043483 and R01 DK098656 (J.A.B.), NIH grant AG031782 (A.M.C.), the Proteostasis of Aging Core AG038072 (A.M.C.). J.M.M. was supported by a postdoctoral fellowship from the American Diabetes Association, grant 1-15-MI-03. P.C. was supported by NIH grants (1P01AG034906, 1R01GM090311, 1R01ES 020812). F.M. is grateful to the FWF for grants LIPOTOX, I1000, P 27893, P 29203 and P24381-B20 and the BMWFW for grants “Unconventional research” and «Flysleep (80.109/0001 -WF/V/3b/2015). JMV was supported by the Spanish Ministerio de Economía y Competitividad (grants BFU2011-23578 and BFU2015-64630-R). The authors thank the personnel from the Servicio Centralizado de Apoyo a la Investigación (SCAI; University of Córdoba) for technical support.
- Published
- 2016
28. Obatoclax kills anaplastic thyroid cancer cells by inducing lysosome neutralization and necrosis
- Author
-
Devora Champa, Bindi Patel, Michela Ranieri, Antonio Di Cristofano, Vladislav V. Verkhusha, Anton A. Shemetov, Arturo Orlacchio, and Ana Maria Cuervo
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Programmed cell death ,autophagy ,Indoles ,Antineoplastic Agents ,Apoptosis ,Biology ,Thyroid Carcinoma, Anaplastic ,Autophagy-Related Protein 7 ,Autophagy-Related Protein 5 ,necrosis ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,lysosomes ,Internal medicine ,Lysosome ,Spheroids, Cellular ,medicine ,Tumor Cells, Cultured ,thyroid cancer ,Animals ,Humans ,Pyrroles ,Thyroid Neoplasms ,Anaplastic thyroid cancer ,Enzyme Inhibitors ,RNA, Small Interfering ,Thyroid cancer ,Cell Proliferation ,Mice, Knockout ,Autophagy ,medicine.disease ,obatoclax ,3. Good health ,Mefloquine ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Oncology ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,Cancer cell ,Cancer research ,RNA Interference ,Obatoclax ,Research Paper - Abstract
Poorly differentiated and anaplastic thyroid carcinomas are very aggressive, almost invariably lethal neoplasms for which no effective treatment exists. These tumors are intrinsically resistant to cell death, even when their driver oncogenic signaling pathways are inhibited. We have undertaken a detailed analysis, in mouse and human thyroid cancer cells, of the mechanism through which Obatoclax, a pan-inhibitor of the anti-apoptotic proteins of the BCL2 family, effectively reduces tumor growth in vitro and in vivo. We demonstrate that Obatoclax does not induce apoptosis, but rather necrosis of thyroid cancer cells, and that non-transformed thyroid cells are significantly less affected by this compound. Surprisingly, we show that Obatoclax rapidly localizes to the lysosomes and induces loss of acidification, block of lysosomal fusion with autophagic vacuoles, and subsequent lysosomal permeabilization. Notably, prior lysosome neutralization using different V-ATPase inhibitors partially protects cancer cells from the toxic effects of Obatoclax. Although inhibition of autophagy does not affect Obatoclax-induced cell death, selective down-regulation of ATG7, but not of ATG5, partially impairs Obatoclax effects, suggesting the existence of autophagy-independent functions for ATG7. Strikingly, Obatoclax killing activity depends only on its accumulation in the lysosomes, and not on its interaction with BCL2 family members. Finally, we show that also other lysosome-targeting compounds, Mefloquine and LLOMe, readily induce necrosis in thyroid cancer cells, and that Mefloquine significantly impairs tumor growth in vivo, highlighting a clear vulnerability of these aggressive, apoptosis-resistant tumors that can be therapeutically exploited.
- Published
- 2016
29. Selective endosomal microautophagy is starvation-inducible in Drosophila
- Author
-
Anindita Mukherjee, Bindi Patel, Koga, Hiroshi, Cuervo, Ana Maria, and Jenny, Andreas
- Subjects
fungi - Abstract
Autophagy delivers cytosolic components to lysosomes for degradation and is thus essential for cellular homeostasis and to cope with different stressors. As such, autophagy counteracts various human diseases and its reduction leads to aging-like phenotypes. Macroautophagy (MA) can selectively degrade organelles or aggregated proteins, whereas selective degradation of single proteins has only been described for chaperone-mediated autophagy (CMA) and endosomal microautophagy (eMI). These 2 autophagic pathways are specific for proteins containing KFERQ-related targeting motifs. Using a KFERQ-tagged fluorescent biosensor, we have identified an eMI-like pathway in Drosophila melanogaster. We show that this biosensor localizes to late endosomes and lysosomes upon prolonged starvation in a KFERQ- and Hsc70-4- dependent manner. Furthermore, fly eMI requires endosomal multivesicular body formation mediated by ESCRT complex components. Importantly, induction of Drosophila eMI requires longer starvation than the induction of MA and is independent of the critical MA genes atg5, atg7, and atg12. Furthermore, inhibition of Tor signaling induces eMI in flies under nutrient rich conditions, and, as eMI in Drosophila also requires atg1 and atg13, our data suggest that these genes may have a novel, additional role in regulating eMI in flies. Overall, our data provide the first evidence for a novel, starvation-inducible, catabolic process resembling endosomal microautophagy in the Drosophila fat body.
- Published
- 2016
- Full Text
- View/download PDF
30. Autophagy modulates dynamics of connexins at the plasma membrane in a ubiquitin-dependent manner
- Author
-
David C. Spray, Ana Maria Cuervo, Eloy Bejarano, Henrique Girão, Bindi Patel, Andrea Yuste, Paulo Pereira, and Carla Marques
- Subjects
Male ,Nedd4 Ubiquitin Protein Ligases ,Ubiquitin-Protein Ligases ,media_common.quotation_subject ,Connexin ,Cell Communication ,Biology ,Endocytosis ,Models, Biological ,Connexins ,Cell membrane ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Phagosomes ,Chlorocebus aethiops ,Autophagy ,otorhinolaryngologic diseases ,medicine ,Animals ,Internalization ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,media_common ,0303 health sciences ,Endosomal Sorting Complexes Required for Transport ,Cell Membrane ,Ubiquitination ,Gap Junctions ,Signal transducing adaptor protein ,Articles ,Cell Biology ,Cell Compartmentation ,Cell biology ,medicine.anatomical_structure ,Membrane Trafficking ,COS Cells ,Proteolysis ,NIH 3T3 Cells ,biology.protein ,sense organs ,030217 neurology & neurosurgery - Abstract
Connexins modulate intercellular communication when assembled in gap junctions. Compromised macroautophagy increases cellular communication due to failure to degrade connexins at gap junctions. Nedd4-mediated ubiquitinylation of the connexin molecule is required to trigger its autophagy-dependent internalization and degradation., Different pathways contribute to the turnover of connexins, the main structural components of gap junctions (GJs). The cellular pool of connexins targeted to each pathway and the functional consequences of degradation through these degradative pathways are unknown. In this work, we focused on the contribution of macroautophagy to connexin degradation. Using pharmacological and genetic blockage of macroautophagy both in vitro and in vivo, we found that the cellular pool targeted by this autophagic system is primarily the one organized into GJs. Interruption of connexins' macroautophagy resulted in their retention at the plasma membrane in the form of functional GJs and subsequent increased GJ-mediated intercellular diffusion. Up-regulation of macroautophagy alone is not sufficient to induce connexin internalization and degradation. To better understand what factors determine the autophagic degradation of GJ connexins, we analyzed the changes undergone by the fraction of plasma membrane connexin 43 targeted for macroautophagy and the sequence of events that trigger this process. We found that Nedd4-mediated ubiquitinylation of the connexin molecule is required to recruit the adaptor protein Eps15 to the GJ and to initiate the autophagy-dependent internalization and degradation of connexin 43. This study reveals a novel regulatory role for macroautophagy in GJ function that is directly dependent on the ubiquitinylation of plasma membrane connexins.
- Published
- 2012
31. HTT/Huntingtin in selective autophagy and Huntington disease: A foe or a friend within?
- Author
-
Yanning Rui, Ana Maria Cuervo, Zhen Xu, Sheng Zhang, and Bindi Patel
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,Atg1 ,animal diseases ,Aggrephagy ,Nerve Tissue Proteins ,Biology ,Models, Biological ,Mice ,mental disorders ,Mitophagy ,Autophagy ,Animals ,Humans ,Kinase activity ,Molecular Biology ,Cell Biology ,Autophagic Punctum ,nervous system diseases ,Drosophila melanogaster ,Huntington Disease ,nervous system ,Proteotoxicity ,Cancer research ,MAP1LC3A - Abstract
Huntington disease (HD) is caused by a unique mutation, an abnormal expansion of a polyglutamine (polyQ) tract in the HTT (huntingtin) protein. It has long been hypothesized that in addition to gained toxicity from the expanded polyQ, loss of HTT's normal cellular functions also contributes to HD pathogenesis. Consistently, wild-type HTT has a well-documented neuronal protective activity and is essential for postnatal neuronal survival. However, despite its relatively simple genetic cause, the etiology of HD remains unclear, due in part to the poor understanding of the physiological cellular functions of HTT. The perplexing HTT is a large ∼3,144 amino acid-long protein with ubiquitous localization, but it lacks any known functional domain that could provide clues about its cellular activities. The phenotypes of HTT knockout (KO) mice are equally puzzling: homozygote HTT KO mice die by day 7.5, but this early embryonic lethality is due to a critical role of HTT in extraembryonic membranes, not in the embryo per se, as it can be rescued if wild-type HTT is provided in extraembryonic tissues. HTT has a vast number of reported interacting partners that have been used to infer some of the growing list of cellular pathways that HTT could participate in. However, it is still uncertain how HTT achieves its well-documented neuroprotective role. Using both Drosophila and mammalian experimental models, we recently demonstrated that HTT has an essential function in selective autophagy where it serves as a scaffold by modulating the activities of the cargo receptor SQSTM1/p62 and the autophagy initiation kinase ULK1. We previously created a null deletion mutant (htt-ko) of the single HTT homolog (htt) in Drosophila and showed that in contrast to mouse, htt-ko flies, which develop ex utero, are fully viable with only mild aging-related defects. Unexpectedly, ectopic expression of a truncated form of the mammalian MAPT induced severe defects in htt-ko flies, suggesting that htt protects against pathogenic MAPT toxicity. Further, in genetic screens using this MAPT-induced phenotype as a functional readout, we detected dosage-sensitive genetic interaction between htt-ko and several components of the autophagy pathway, including atg8a (MAP1LC3A homolog), atg1 (ULK1 homolog) and ref(2)P (SQSTM1/p62 homolog), thus unveiling a functional link between HTT and autophagy. HTT is not a foreigner to the autophagy field, although the main emphasis has been on the toxic effect of mutant HTT on this cellular clearance mechanism. In fact, our earlier work in HD demonstrated that reduced autophagy in the disease context, also reported by many other groups, originated from diminished ability to “trap” cytosolic cargo. We attributed the “empty autophagosomes” phenotype in HD cells to defective cargo recognition due to the presence of mutant polyQ-HTT in the inner part of the closing autophagosomes. However, our genetic studies ablating the wild-type HTT made us reconsider this proposed gain-of-toxicity function. In this recent study, we have found that in both htt-ko flies and HTT-depleted mammalian cells, autophagic abnormalities are present but surprisingly, starvation-induced autophagy is largely normal. However, loss of HTT compromised autophagic induction when challenged by several other stresses, including proteotoxicity, lipotoxicity, and mitochondria damage. In all these instances, mammalian cells depleted of HTT also showed an “empty autophagosomes” defect, suggesting that wild-type HTT is required for efficient cargo recognition by the autophagosome. We found that HTT and SQSTM1/p62, as well as their Drosophila counterparts Htt and Ref(2)P, physically interact and that proteotoxic stress promotes this interaction. Depletion of HTT reduces the association of SQSTM1 with MAP1LC3A and also compromises the binding of SQSTM1 with proteins with lysine-63-linked ubiquitin (K63-Ub) chains, which are preferential substrates of autophagy. However, loss of HTT does not affect self-polymerization of SQSTM1 or its association with K48-Ub-modified proteins mainly cleared by the proteasome. Collectively, these findings support the idea that HTT facilitates cargo recognition by modulating the assembly of the cargo receptors and autophagy proteins. We also found that the dependence on HTT for the initiation of stress-induced selective autophagy is related to its physical interaction with ULK1, whose kinase activity is essential for autophagy initiation. HTT depletion does not affect starvation-induced activation of ULK1 kinase, but compromises ULK1 activation upon proteotoxicity challenge, revealing a contribution of HTT to differentially regulating autophagosome biogenesis under basal or stress conditions. We found that the regulation of HTT on ULK1 is at the level of its interaction with the MTORC1 complex, another main regulator of ULK1 that binds to and inactivates ULK1 under nutrient-rich conditions. We showed that (1) ULK1 exists in 2 mutually exclusive complexes, ULK1-HTT and ULK1-MTORC1; (2) HTT does not affect the kinase activity of MTORC1; (3) HTT competes with MTORC1 for binding with ULK1; (4) stresses that induce selective autophagy (e.g., proteotoxicity, lipotoxicity, or mitophagy), but not starvation, result in an increased association of ULK1 with HTT at the expense of MTOR, thus freeing ULK1 from its inhibition by MTORC1. Collectively, HTT promotes selective autophagy by activating and bringing together SQSTM1/p62 and ULK1 to assure spatial proximity between the cargo and autophagy initiation components, thereby orchestrating 2 major autophagy steps: cargo recognition and autophagy induction (Fig. 1). Figure 1. Model of HTT in promoting selective autophagy. HTT serves as a scaffolding for selective autophagy by bringing together cargo bound through SQSTM1 and an initiator of autophagy, ULK1 kinase. Basal autophagy: under basal conditions, binding of HTT to the ... Increasing evidence supports a tangled relationship between HTT, HD, and autophagy. Thus, after the first report of altered autophagy in HD by DiFiglia, the Holzbaur lab showed that HTT facilitates axonal trafficking of autophagosomes, a function that is compromised by polyQ-HTT; Hayden's group reported an autophagy-inducing domain within HTT; Steffan with Thompson and colleagues also recently reported physical interactions between HTT and several autophagy proteins including SQSTM1 and ULK1. Our finding places HTT in the core of the autophagic process, bringing together components of the cargo recognition and autophagy initiation complexes, and raising many new interesting questions. How is HTT differentially activated upon different stress challenges? Does polyQ expansion directly compromise the endogenous activities of HTT, as implicated by the very similar “empty autophagosomes” phenotypes in both HD and HTT-depleted cells, and if so, will this effect be partially responsible for the toxicity of mutant polyQ-HTT in long-lived neurons? Last, how can we exploit HTT's role in selective autophagy to modulate this process in the fight against this devastating brain disease called HD?
- Published
- 2015
32. Common γ-chain cytokine signaling is required for macroautophagy induction during CD4+ T-cell activation
- Author
-
Yair Botbol, Bindi Patel, Fernando Macian, Yair Botbol, Bindi Patel, and Fernando Macian
- Published
- 2016
- Full Text
- View/download PDF
33. Combination Vaccines: Postlicensure Safety Evaluation
- Author
-
Vitali Pool, Bindi Patel, Hiroshi Takahashi, Robert T. Chen, and Bruce G. Weniger
- Subjects
Microbiology (medical) ,Vaccine safety ,Vaccines adverse reaction ,Clinical Trials as Topic ,Standardization ,business.industry ,Contraindications ,Data Collection ,Assessment center ,Combination vaccines ,Vaccination ,Infectious Diseases ,Immunization ,Risk analysis (engineering) ,Terminology as Topic ,Immunology ,Product Surveillance, Postmarketing ,Adverse Drug Reaction Reporting Systems ,Humans ,Medicine ,Vaccines, Combined ,business ,Adverse effect ,Drug Packaging ,Software - Abstract
The success of immunizations in nearly eliminating many vaccine-preventable diseases has resulted in an increase in the need to study risks from vaccines, combination or otherwise. The well-known limitations associated with prelicensure trials have led many to hope that postlicensure studies can address safety issues. This article reviews measures that have been or should be taken to meet this expectation: establishment of clinical immunization safety assessment centers, standardization of case definitions for vaccine adverse events, use of the Vaccine Identification Standards Initiative to improve the accuracy and efficiency with which vaccination records are transferred, integration of vaccine safety monitoring into immunization registries, establishment (and enlargement) of the Vaccine Safety Datalink project, use of innovative analytic tools for better signal detection, and implementation of various methods to overcome confounding by contraindication. Only by investing in vaccine safety infrastructure at a level commensurate with investments in vaccine development can we hope to retain the public's confidence in immunization.
- Published
- 2001
34. Common γ-chain cytokine signaling is required for macroautophagy induction during CD4+ T-cell activation
- Author
-
Yair Botbol, Bindi Patel, Fernando Macian, Yair Botbol, Bindi Patel, and Fernando Macian
- Published
- 2015
- Full Text
- View/download PDF
35. Connexins modulate autophagosome biogenesis
- Author
-
Andrea Yuste, Eloy Bejarano, Bindi Patel, David C. Spray, Ana Maria Cuervo, and Randy F. Stout
- Subjects
Autophagosome ,Male ,Time Factors ,Connexin ,Biology ,Transfection ,Article ,Connexins ,Cell membrane ,Vacuolar Sorting Protein VPS15 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,otorhinolaryngologic diseases ,medicine ,Autophagy ,Animals ,Humans ,Rats, Wistar ,Transport Vesicles ,Gap junctions ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Cell Membrane ,Gap junction ,Cell Biology ,Class III Phosphatidylinositol 3-Kinases ,Cell biology ,Rats ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Starvation ,Connexin 43 ,RNA Interference ,sense organs ,Signal transduction ,Apoptosis Regulatory Proteins ,Lysosomes ,030217 neurology & neurosurgery ,Biogenesis ,HeLa Cells ,Signal Transduction ,Plasma membrane - Abstract
The plasma membrane contributes to the formation of autophagosomes, the double-membrane vesicles that sequester cytosolic cargo and deliver it to lysosomes for degradation during autophagy. In this study, we have identified a regulatory role for connexins (Cx), the main components of plasma membrane gap junctions, in autophagosome formation. We have found that plasma-membrane-localized Cx proteins constitutively downregulate autophagy through a direct interaction with several autophagy-related proteins involved in the initial steps of autophagosome formation, such as Atg16 and components of the PI(3)K autophagy initiation complex (Vps34, Beclin-1 and Vps15). On nutrient starvation, this inhibitory effect is released by the arrival of Atg14 to the Cx-Atg complex. This promotes the internalization of Cx-Atg along with Atg9, which is also recruited to the plasma membrane in response to starvation. Maturation of the Cx-containing pre-autophagosomes into autophagosomes leads to degradation of these endogenous inhibitors, allowing for sustained activation of autophagy.
- Published
- 2013
36. The lipid kinase PI4KIIIβ preserves lysosomal identity
- Author
-
Dennis Shields, Laura Santambrogio, Sunandini Sridhar, David Aphkhazava, Fernando Macian, Bindi Patel, and Ana Maria Cuervo
- Subjects
Male ,Have You Seen...? ,Endocytic cycle ,Context (language use) ,Biology ,Real-Time Polymerase Chain Reaction ,General Biochemistry, Genetics and Molecular Biology ,Article ,symbols.namesake ,Mice ,Lysosomal-Associated Membrane Protein 1 ,Chlorocebus aethiops ,Protein Isoforms ,Animals ,Humans ,Kinase activity ,RNA, Small Interfering ,Rats, Wistar ,Molecular Biology ,1-Phosphatidylinositol 4-Kinase ,Mannose 6-phosphate receptor ,General Immunology and Microbiology ,Kinase ,General Neuroscience ,Lentivirus ,Lipid metabolism ,Biological Transport ,Golgi apparatus ,Lipid Metabolism ,Immunohistochemistry ,Cell biology ,Rats ,Mice, Inbred C57BL ,Microscopy, Electron ,HEK293 Cells ,Biochemistry ,Microscopy, Fluorescence ,Gene Knockdown Techniques ,COS Cells ,symbols ,NIH 3T3 Cells ,Lysosomes - Abstract
Lipid modifications are essential in cellular sorting and trafficking inside cells. The role of phosphoinositides in trafficking between Golgi and endocytic/lysosomal compartments has been extensively explored and the kinases responsible for these lipid changes have been identified. In contrast, the mechanisms that mediate exit and recycling from lysosomes (Lys), considered for a long time as terminal compartments, are less understood. In this work, we identify a dynamic association of the lipid kinase PI4KIIIβ with Lys and unveil its regulatory function in lysosomal export and retrieval. We have found that absence of PI4KIIIβ leads to abnormal formation of tubular structures from the lysosomal surface and loss of lysosomal constituents through these tubules. We demonstrate that the kinase activity of PI4KIIIβ is necessary to prevent this unwanted lysosomal efflux under normal conditions, and to facilitate proper sorting when recycling of lysosomal material is needed, such as in the physiological context of lysosomal reformation after prolonged starvation.
- Published
- 2012
37. Alpha-defensin expression during myelopoiesis: identification of cis and trans elements that regulate expression of NP-3 in rat promyelocytes
- Author
-
Niaz Banaiee, Nannette Y. Yount, Michael E. Selsted, Bindi Patel, and Cindy M. Yamamoto
- Subjects
alpha-Defensins ,Neutrophils ,Cellular differentiation ,Immunology ,CAAT box ,Biology ,Transfection ,Cell Line ,Gene expression ,Genes, Regulator ,Immunology and Allergy ,Animals ,Granulocyte Precursor Cells ,Northern blot ,Promoter Regions, Genetic ,Gene ,Defensin ,Regulation of gene expression ,Myelopoiesis ,Cell Biology ,Molecular biology ,Clone Cells ,Rats ,Gene Expression Regulation ,Mutation - Abstract
α-Defensins are antimicrobial peptides that contribute to innate-immune functions of neutrophils and intestinal Paneth cells. Transcription of α-defensin genes occurs early in neutrophilic myelopoeisis. To examine the mechanisms that regulate α-defensin gene expression, we analyzed transcription of rat neutrophil α-defensin NP-3 in D4 cells, a subclone of the promyelocytic cell lne IPC-81. Northern blot analysis showed that D4 cells express fivefold higher levels of α-defensin mRNA than the parental cell line in a manner relatively independent of passage number. Increased levels of steady-state mRNA in D4 cells correlated with markedly elevated peptide levels detected by immunocytochemical staining. To identify the cis-acting DNA elements involved in tissue-specific expression, D4 cells were transfected with luciferase reporter constructs containing NP-3 gene 5′-flanking sequences. Analyses of transfected D4 cells demonstrated that the proximal 87 base pair (bp) sequence contained cis-acting DNA elements necessary for optimal promoter activity. Mutational analyses within the 87-bp region suggested the involvement of the CAAT box and a putative polyoma enhancer-binding protein 2/core-binding factor (PEBP2/CBF) site in defensin gene transcription. Transient transfection analyses using tandem repeats of oligonucleotides containing these sequences demonstrated that proximity of the CAAT box and PEBP2/CBF site was important for defensin promoter activity. Electrophoretic mobility shift assays indicated that PEBP2/CBF or a PEBP2/CBF-related protein was involved in a specific protein-DNA interaction occurring within a DNA fragment containing the CAAT and PEBP2/CBF sequences. These data identify functional trans- and cis-elements that regulate rat defensin gene expression in high defensin-expressing promyelocytic cells.
- Published
- 2003
38. Plant Viral Nanoparticle-based Vaccine Targeting NY-ESO-1+ Triple Negative Breast Cancer
- Author
-
Patel, Bindi, Patel
- Subjects
- Biomedical Engineering, TNBC, immunotherapy, adjuvant, NY-ESO-1, CPMV, CTL
- Abstract
Triple-negative breast cancer (TNBC) is an aggressive, life-threatening disease afflicting 10-20% of all breast cancer patients. The absence of steroid receptors and lack of HER2 receptor overexpression renders standard targeted therapies untenable treatment options limiting postsurgical disease management to systemic chemotherapy. However, the increased likelihood of loco-regional recurrence, metastatic spread and early death (within 5 years of surgical resection) in TNBC patients, underlines an urgent need for identification of new adjuvant therapies to prevent recurrence and prolong disease-free survival. Adjuvant immunotherapies targeted to cancer antigens can prolong disease free survival by recognizing and eliminating residual or recurring disease. NY-ESO-1, a highly immunogenic antigen associated with TNBC and several other malignancies is an appealing target for such interventions. NY-ESO-1 specific cancer vaccines can potentially amplify any pre-existing weak immune response present in NY-ESO-1+ patients and mobilize the immune system to eliminate tumors and metastasis; priming of immune memory by such mechanism holds the potential to prevent recurrence of the disease. Current NY-ESO-1 vaccine strategies are limited by the inherently low immunogenicity and instability of antigenic peptides, use of poorly tolerated adjuvants, ineffective presentation of immunodominant epitopes in recombinant proteins, and costly and technically challenging dendritic cell vaccines. To overcome these shortcomings, I used a highly immunostimulatory and biocompatible viral nanoparticle (VNP) carrier derived from cowpea mosaic virus (CPMV) to stimulate an effective and sustained cytotoxic T cell (CTL) response targeting NY-ESO-1 and test its efficacy in in vitro and ex vivo models.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.