621 results on '"Georg-Speyer-Haus"'
Search Results
2. Low-Level Expression of Functional Foamy Virus Receptor on Hematopoietic Progenitor Cells
- Author
-
von Laer, Dorothee, Lindemann, Dirk, Roscher, Susanne, Herwig, Uwe, Friel, Jutta, and Herchenröder, Ottmar
- Published
- 2001
- Full Text
- View/download PDF
3. Analysis of Cellular Factors Influencing the Replication of Human Immunodeficiency Virus Type I in Human Macrophages Derived from Blood of Different Healthy Donors
- Author
-
Eisert, Veronika, Kreutz, Marina, Becker, Karin, Königs, Christoph, Alex, Uwe, Rübsamen-Waigmann, Helga, Andreesen, Reinhard, and von Briesen, Hagen
- Published
- 2001
- Full Text
- View/download PDF
4. Verbundprojekt: Zielgerichtete Killerzellen für die zelluläre Krebs-Immuntherapie : Schlussbericht für den Gesamtverbund ; Berichtszeitraum: 01.02.2008 - 31.12.2011
- Author
-
Georg-Speyer-Haus
- Subjects
Medical Technology - Abstract
Ill., graph. Darst.
- Published
- 2012
- Full Text
- View/download PDF
5. Schlussbericht zu dem Verbundprojekt 'CORUS: Co-receptor usage as a marker for specific HIV diagnostics with high sensitivity' : Teilprojekt 1: Selection of peptide ligands for V3 peptides by the phage display technology
- Author
-
Georg-Speyer-Haus
- Subjects
Chemistry - Abstract
Ill., graph. Darst.
- Published
- 2011
- Full Text
- View/download PDF
6. Funktionale Systeme auf Nanopartikel-Basis zur Behandlung von Gehirn-Tumoren, TP 2: Transportmechanismus- und Toxizitätsuntersuchungen in Zellkulturmodellen : Schlussbericht ... ; Laufzeit des Vorhabens: 01.02.2002 - 31.12.2005 (verlängert)
- Author
-
Georg-Speyer-Haus
- Subjects
Blut-Hirn-Schranke ,Nanopartikel ,Medicine ,Onkologie ,Hirntumor - Abstract
Ill., graph. Darst.
- Published
- 2006
- Full Text
- View/download PDF
7. Supramolekulare Drug-Delivery-Systeme zum gezielten Transport von Nucleinsäure-Analoga über biologische Barrieren : Schlussbericht ; Berichtszeitraum: 01.06.2000 - 31.08.2003 ; Laufzeit des Vorhabens: 01.06.2000 - 31.05.2003 (verlängert bis 31.08.2003)
- Author
-
Georg-Speyer-Haus
- Subjects
Chemistry ,Chemical and environmental engineering - Published
- 2003
- Full Text
- View/download PDF
8. Secretable HIV entry inhibitory peptides for therapy of HIV infection
- Author
-
VISION 7 GMBH and CHEMOTHERAPEUTISCHES FORSCHUNGSINSTITUT GEORG SPEYER HAUS
- Abstract
The invention relates to the treatment of an HIV infection by the expression of in vivo secretable peptides that prevent entry of the virus into the cell. The invention makes available nucleic acids and vectors containing the same that code for a fusion protein that contains two HIV entry inhibitory peptides connected by a cleavable or non-cleavable, flexible linker.
9. FIBRONECTIN FOR USE IN THE TREATMENT OF LEUKEMIA
- Author
-
CHEMOTHERAPEUTISCHES FORSCHUNGSINSTITUT GEORG SPEYER HAUS
- Abstract
The present invention pertains to fibronectin in the treatment of imatinib resistant leukemia. The invention relates to the use of recombinant or isolated fibronectin as adjuvant therapy during leukemia treatment, either as single ingredient medicament or in a combination therapy, preferably with Abl1 inhibitors such as imatinib or nilotinib.
10. Transgenic overexpression of a dominant negative mutant of FADD that, although counterselected during tumor progression, cooperates in L-myc-induced tumorigenesis
- Author
-
Anne-Odile, Hueber, Susanne, Bösser, Martin, Zörnig, Institut de signalisation, biologie du développement et cancer (ISBDC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, and Georg-Speyer-Haus
- Subjects
MESH: Mutation ,Fas-Associated Death Domain Protein ,Genes, myc ,bcl-X Protein ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Lymphoma, T-Cell ,urologic and male genital diseases ,MESH: Mice, Knockout ,MESH: bcl-X Protein ,Animals, Genetically Modified ,MESH: Animals, Genetically Modified ,Mice ,MESH: Mice, Inbred C57BL ,Animals ,MESH: Animals ,fas Receptor ,MESH: Tumor Suppressor Protein p53 ,MESH: Mice ,MESH: Genes, myc ,Adaptor Proteins, Signal Transducing ,Genes, Dominant ,MESH: Adaptor Proteins, Signal Transducing ,Mice, Knockout ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,MESH: Fas-Associated Death Domain Protein ,MESH: Antigens, CD95 ,Mice, Inbred C57BL ,Proto-Oncogene Proteins c-bcl-2 ,MESH: Proto-Oncogene Proteins c-bcl-2 ,Mutation ,Disease Progression ,MESH: Disease Progression ,MESH: Lymphoma, T-Cell ,Tumor Suppressor Protein p53 ,biological phenomena, cell phenomena, and immunity ,MESH: Genes, Dominant - Abstract
International audience; Activation of the so-called death receptors, e.g., CD95/Fas/Apo-1, is a potent stimulus to trigger apoptosis. Overexpression of the C-terminal FADD deletion mutant FADD-DN blocks death receptor-induced apoptosis, but despite this antiapoptotic activity, lck FADD-DN transgenic mice do not develop lymphomas. To analyze whether functional inactivation of FADD cooperates with Myc overexpression in tumorigenesis, lck FADD-DN transgenic mice were crossed with Emicro L-myc transoncogenic animals. While no tumors were detected in single transgenic FADD-DN or L-myc mice within 15 months, 5 of 17 (29%) FADD-DN/L-myc double transgenic animals developed lymphomas with an average latency period of 47 weeks. Protein analysis of FADD-DN/L-myc tumors showed, however, undetectable levels of FADD-DN protein. FADD-DN protein expression was again lost in 16 of 17 FADD-DN/p53 k.o. T-cell lymphomas, though no significant acceleration of tumorigenesis in P53-deficient lck FADD-DN mice compared to p53 k.o. animals was observed. These data suggest a strong counterselection against the FADD-DN protein during tumor progression, which could be explained by the cell cycle inhibitory activity of FADD-DN. Such counterselection would have to be compensated for by other antiapoptotic mutations, and indeed, strong upregulation of the antiapoptotic Bcl-2 family member Bcl-xL was found in one of the tumors. This in vivo mouse model demonstrates that an antiapoptotic protein involved in the onset of tumorigenesis is selected against and consequently lost during tumor progression because of its additional antiproliferative activity.
- Published
- 2004
- Full Text
- View/download PDF
11. The EHA Research Roadmap
- Author
-
Jaffredo, Thierry, Balduini, Alessandra, Bigas, Anna, Bernardi, Rosa, Bonnet, Dominique, Canque, Bruno, Charbord, Pierre, Cumano, Anna, Delwel, Ruud, Durand, Charles, Fibbe, Willem, Forrester, Lesley, De Franceschi, Lucia, Ghevaert, Cedric, Gjertsen, Bjørn, Gottgens, Berthold, Graf, Thomas, Heidenreich, Olaf, Hermine, Olivier, Higgs, Douglas, Kleanthous, Marina, Klump, Hannes, Kouskoff, Valerie, Krause, Daniela, Lacaud, George, Celso, Cristina Lo, Martens, Joost H.A., Méndez-Ferrer, Simón, Menendez, Pablo, Oostendorp, Robert, Philipsen, Sjaak, Porse, Bo, Raaijmakers, Marc, Robin, Catherine, Stunnenberg, Henk, Theilgaard-Mönch, Kim, Touw, Ivo, Vainchenker, William, Corrons, Joan-Lluis Vives, Yvernogeau, Laurent, Schuringa, Jan Jacob, Celso, Cristina, Martens, Joost, Schuringa, Jan, Laboratoire de Biologie du Développement [IBPS] (LBD), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Università degli Studi di Pavia = University of Pavia (UNIPV), IMIM-Hospital del Mar, Generalitat de Catalunya, Josep Carreras Leukaemia Research Institute (IJC), Instituto de Salud Carlos III [Madrid] (ISC), IRCCS San Raffaele Scientific Institute [Milan, Italie], The Francis Crick Institute [London], Immunologie humaine, physiopathologie & immunothérapie (HIPI (UMR_S_976 / U976)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Institut de Recherche Saint-Louis - Hématologie Immunologie Oncologie (Département de recherche de l’UFR de médecine, ex- Institut Universitaire Hématologie-IUH) (IRSL), Université Paris Cité (UPCité), Lymphopoïèse (Lymphopoïèse (UMR_1223 / U1223 / U-Pasteur_4)), Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Erasmus University Medical Center [Rotterdam] (Erasmus MC), Leiden University Medical Center (LUMC), Universiteit Leiden, University of Edinburgh, Università degli studi di Verona = University of Verona (UNIVR), University of Cambridge [UK] (CAM), Haukeland University Hospital, University of Bergen (UiB), Barcelona Institute of Science and Technology (BIST), Universitat Pompeu Fabra [Barcelona] (UPF), Princess Máxima Center for Pediatric Oncology [Utrecht, Netherlands], Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), John Radcliffe Hospital [Oxford University Hospital], Cyprus Institute of Neurology and Genetics, Universitätsklinikum Essen [Universität Duisburg-Essen] (Uniklinik Essen), University of Manchester [Manchester], Goethe-Universität Frankfurt am Main, Georg-Speyer-Haus, Imperial College London, Radboud University [Nijmegen], NHSBT, Universitat de Barcelona (UB), Institució Catalana de Recerca i Estudis Avançats (ICREA), Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM), University of Copenhagen = Københavns Universitet (UCPH), Hubrecht Institute [Utrecht, Netherlands], University Medical Center [Utrecht]-Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center [Utrecht], Institut Gustave Roussy (IGR), Dynamique moléculaire de la transformation hématopoïétique (Dynamo), Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay, University Medical Center Groningen [Groningen] (UMCG), Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Università degli Studi di Pavia, Laboratoire d'Innovation Thérapeutique (LIT), Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Institut de Chimie du CNRS (INC), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Université de Paris (UP), Laboratoire de Biologie du Développement [Paris] (LBD), Institut Pasteur [Paris], Hubrecht Institute for Developmental Biology and Stem Cell Research, Ghevaert, Cedric [0000-0002-9251-0934], Gottgens, Berthold [0000-0001-6302-5705], Apollo - University of Cambridge Repository, and Stem Cell Aging Leukemia and Lymphoma (SALL)
- Subjects
Medizin ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Bioinformatics ,03 medical and health sciences ,0302 clinical medicine ,SINGLE-CELL ,REVEALS ,Medicine ,Diseases of the blood and blood-forming organs ,Molecular Biology ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,DAMAGE ,0303 health sciences ,VASCULAR NICHE ,business.industry ,ORIGIN ,Normal hematopoiesis ,Hematology ,3. Good health ,ddc ,MODEL ,030220 oncology & carcinogenesis ,Perspective ,RC633-647.5 ,business ,COMMITMENT ,STEM-CELLS - Abstract
International audience; In 2016, the European Hematology Association (EHA) published the EHA Roadmap for European Hematology Research1 aiming to highlight achievements in the diagnostics and treatment of blood disorders, and to better inform European policy makers and other stakeholders about the urgent clinical and scientific needs and priorities in the field of hematology. Each section was coordinated by 1–2 section editors who were leading international experts in the field. In the 5 years that have followed, advances in the field of hematology have been plentiful. As such, EHA is pleased to present an updated Research Roadmap, now including 11 sections, each of which will be published separately. The updated EHA Research Roadmap identifies the most urgent priorities in hematology research and clinical science, therefore supporting a more informed, focused, and ideally a more funded future for European hematology research. The 11 EHA Research Roadmap sections include Normal Hematopoiesis; Malignant Lymphoid Diseases; Malignant Myeloid Diseases; Anemias and Related Diseases; Platelet Disorders; Blood Coagulation and Hemostatic Disorders; Transfusion Medicine; Infections in Hematology; Hematopoietic Stem Cell Transplantation; CAR-T and Other Cell-based Immune Therapies; and Gene Therapy.
- Published
- 2021
- Full Text
- View/download PDF
12. Optimization of the EC26-2A4 Epitope in the gp41 Membrane Proximal External Region Targeted by Neutralizing Antibodies from an Elite Controller
- Author
-
Timo Wolf, Ursula Dietrich, Karsten Müller, Oliver Ringel, Boris Brill, Vincent Vieillard, Patrice Debré, Christoph Stephan, Joachim Koch, Georg-Speyer-Haus, Johannes Gutenberg - Universität Mainz (JGU), Universitätsklinikum Frankfurt, Centre d'Immunologie et de Maladies Infectieuses (CIMI), and Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)
- Subjects
0301 basic medicine ,Env ,medicine.drug_class ,Anti-HIV Agents ,viruses ,Immunology ,Human immunodeficiency virus (HIV) ,HIV Infections ,Biology ,HIV Antibodies ,Gp41 ,Monoclonal antibody ,medicine.disease_cause ,Epitope ,HIV Long-Term Survivors ,03 medical and health sciences ,Epitopes ,Mice ,0302 clinical medicine ,Blood serum ,Viral envelope ,Control theory ,Virology ,medicine ,Animals ,Humans ,neutralizing antibodies ,030212 general & internal medicine ,ComputingMilieux_MISCELLANEOUS ,AIDS Vaccines ,Vaccines ,epitope ,virus diseases ,Antibodies, Monoclonal ,gp41 ,Antibodies, Neutralizing ,HIV Envelope Protein gp41 ,3. Good health ,MPER ,CD4 Lymphocyte Count ,030104 developmental biology ,Infectious Diseases ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,biology.protein ,HIV-1 ,Antibody ,Peptides ,Broadly Neutralizing Antibodies - Abstract
The analysis of patient derived HIV neutralizing antibodies (nAbs) and their target epitopes in the viral envelope (Env) protein provides important basic information for vaccine design. In this study we optimized an epitope, EC26-2A4, that is targeted by neutralizing antibodies from an elite controller (EC26) and localizes in the membrane-proximal external region from the gp41 transmembrane protein. Due to its overlap with the epitope of the first generation broadly neutralizing monoclonal Ab (mAb) 2F5 associated with autoreactivity, we first defined the minimal core epitope reacting with antibodies from EC26 plasma, but not with mAb 2F5. The optimized minimal epitope, EC26-2A4ΔM, was able to induce neutralizing antibodies in vaccinated mice. We further analyzed the frequency of antibodies against the EC26-2A4ΔM peptide in HIV-positive patient sera from a treated cohort and an untreated long-term nonprogressor (LTNP) cohort. Interestingly, 27% of the LTNP sera reacted with the peptide, whereas only 9% showed reactivity in the treated cohort. Although there was no association between the presence of antibodies against the EC26-2A4ΔM epitope and viral load or CD4 count in these patients, the CD4 nadir in the treated cohort was higher in patients positive for EC26-2A4ΔM antibodies, in particular in patients having such antibodies at an early and a late timepoint after infection.
- Published
- 2018
- Full Text
- View/download PDF
13. Human APOBEC3G incorporation into murine leukemia virus particles
- Author
-
Schnierle, Barbara [Georg-Speyer-Haus, Institute for Biomedical Research, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt (Germany) and Paul-Ehrlich Institute, Abt. 2/01, Paul-Ehrlich Strasse 51-59, D-63225 Langen (Germany)]
- Published
- 2005
- Full Text
- View/download PDF
14. Global and regional molecular epidemiology of HIV-1, 1990–2015: a systematic review, global survey, and trend analysis
- Author
-
Hemelaar, Joris, Elangovan, Ramyiadarsini, Yun, Jason, Dickson-Tetteh, Leslie, Fleminger, Isabella, Kirtley, Shona, Williams, Brian, Gouws-Williams, Eleanor, Ghys, Peter D, Abimiku, Alash'le G, Agwale, Simon, Archibald, Chris, Avidor, Boaz, Barbás, María Gabriela, Barre-Sinoussi, Francoise, Barugahare, Banson, Belabbes, El Hadj, Bertagnolio, Silvia, Birx, Deborah, Bobkov, Aleksei F, Brandful, James, Bredell, Helba, Brennan, Catherine A, Brooks, James, Bruckova, Marie, Buonaguro, Luigi, Buonaguro, Franco, Buttò, Stefano, Buve, Anne, Campbell, Mary, Carr, Jean, Carrera, Alex, Carrillo, Manuel Gómez, Celum, Connie, Chaplin, Beth, Charles, Macarthur, Chatzidimitriou, Dimitrios, Chen, Zhiwei, Chijiwa, Katsumi, Cooper, David, Cunningham, Philip, Dagnra, Anoumou, de Gascun, Cillian F, Del Amo, Julia, Delgado, Elena, Dietrich, Ursula, Dwyer, Dominic, Ellenberger, Dennis, Ensoli, Barbara, Essex, Max, Gao, Feng, Fleury, Herve, Fonjungo, Peter N, Foulongne, Vincent, Gadkari, Deepak A, García, Federico, Garsia, Roger, Gershy-Damet, Guy Michel, Glynn, Judith R, Goodall, Ruth, Grossman, Zehava, Lindenmeyer-Guimarães, Monick, Hahn, Beatrice, Hamers, Raph L, Hamouda, Osamah, Handema, Ray, He, Xiang, Herbeck, Joshua, Ho, David D, Holguin, Africa, Hosseinipour, Mina, Hunt, Gillian, Ito, Masahiko, Bel Hadj Kacem, Mohamed Ali, Kahle, Erin, Kaleebu, Pontiano Kaleebu, Kalish, Marcia, Kamarulzaman, Adeeba, Kang, Chun, Kanki, Phyllis, Karamov, Edward, Karasi, Jean-Claude, Kayitenkore, Kayitesi, Kelleher, Tony, Kitayaporn, Dwip, Kostrikis, Leondios G, Kucherer, Claudia, Lara, Claudia, Leitner, Thomas, Liitsola, Kirsi, Lingappa, Jai, Linka, Marek, Lorenzana de Rivera, Ivette, Lukashov, Vladimir, Maayan, Shlomo, Mayr, Luzia, McCutchan, Francine, Meda, Nicolas, Menu, Elisabeth, Mhalu, Fred, Mloka, Doreen, Mokili, John L, Montes, Brigitte, Mor, Orna, Morgado, Mariza, Mosha, Fausta, Moussi, Awatef, Mullins, James, Najera, Rafael, Nasr, Mejda, Ndembi, Nicaise, Neilson, Joel R, Nerurkar, Vivek R, Neuhann, Florian, Nolte, Claudine, Novitsky, Vlad, Nyambi, Philippe, Ofner, Marianna, Paladin, Fem J, Papa, Anna, Pape, Jean, Parkin, Neil, Parry, Chris, Peeters, Martine, Pelletier, Alexandra, Pérez-Álvarez, Lucía, Pillay, Deenan, Pinto, Angie, Quang, Trinh Duy, Rademeyer, Cecilia, Raikanikoda, Filimone, Rayfield, Mark A, Reynes, Jean-Marc, Rinke de Wit, Tobias, Robbins, Kenneth E, Rolland, Morgane, Rousseau, Christine, Salazar-Gonzales, Jesus, Salem, Hanan, Salminen, Mika, Salomon, Horacio, Sandstrom, Paul, Santiago, Mario L, Sarr, Abdoulaye D, Schroeder, Bryan, Segondy, Michel, Selhorst, Philippe, Sempala, Sylvester, Servais, Jean, Shaik, Ansari, Shao, Yiming, Slim, Amine, Soares, Marcelo A, Songok, Elijah, Stewart, Debbie, Stokes, Julie, Subbarao, Shambavi, Sutthent, Ruengpung, Takehisa, Jun, Tanuri, Amilcar, Tee, Kok Keng, Thapa, Kiran, Thomson, Michael, Tran, Tyna, Urassa, Willy, Ushijima, Hiroshi, van de Perre, Philippe, van der Groen, Guido, van Laethem, Kristel, van Oosterhout, Joep, van Sighem, Ard, van Wijngaerden, Eric, Vandamme, Anne-Mieke, Vercauteren, Jurgen, Vidal, Nicole, Wallace, Lesley, Williamson, Carolyn, Wolday, Dawit, Xu, Jianqing, Yang, Chunfu, Zhang, Linqi, Zhang, Rong, John Radcliffe Hospital [Oxford University Hospital], Centre for Statistics in Medicine, University of Oxford [Oxford], Stellenbosch University, UNAIDS [Genève, Suisse] (ONUSIDA), Institut Pasteur [Paris], Noguchi Memorial Institute for Medical Research [Accra, Ghana] (NMIMR), University of Ghana, Institute of Tropical Medicine [Antwerp] (ITM), State Key Laboratory of Silkworm Genome Biology, Southwest University, Commissariat à l'énergie atomique et aux énergies alternatives - Laboratoire d'Electronique et de Technologie de l'Information (CEA-LETI), Direction de Recherche Technologique (CEA) (DRT (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Georg-Speyer-Haus, The University of Sydney, National AIDS Centre, Istituto Superiore di Sanita [Rome], Harvard School of Public Health, Digital Enterprise Research Institute (DERI-NUIG), National University of Ireland [Galway] (NUI Galway), Microbiologie cellulaire et moléculaire et pathogénicité (MCMP), Université Bordeaux Segalen - Bordeaux 2-Centre National de la Recherche Scientifique (CNRS), Laboratoire de Virologie, Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Pathogénèse et contrôle des infections chroniques (PCCI), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)-Centre Hospitalier Universitaire de Montpellier (CHU Montpellier ), Departments of Medicine and Microbiology, University of Alabama at Birmingham [ Birmingham] (UAB), Robert Koch Institute [Berlin] (RKI), Beihang University (BUAA), Statens Serum Institut [Copenhagen], Immuno-Rhumatologie Moléculaire, Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM), SANTE/SIDA [Bobo-Dioulasso, Burkina Faso], Institut de Recherche en Sciences de la Santé (IRSS) / Centre Muraz, Department of Microbiology, University of Washington School of Medicine, Department of Microbiology, Medical School, University of Thessaly [Volos] (UTH), Recherches Translationnelles sur le VIH et les maladies infectieuses endémiques er émergentes (TransVIHMI), Université Cheikh Anta Diop [Dakar, Sénégal] (UCAD)-Institut de Recherche pour le Développement (IRD)-Université de Yaoundé I-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)-Université Montpellier 1 (UM1), Africa Centre for Health and Population Studies, University of KwaZulu-Natal (UKZN)-Medical Research Council of South Africa, Lab-STICC_UBO_CID_IHSEV, Laboratoire des sciences et techniques de l'information, de la communication et de la connaissance (Lab-STICC), École Nationale d'Ingénieurs de Brest (ENIB)-Université de Bretagne Sud (UBS)-Université de Brest (UBO)-Télécom Bretagne-Institut Brestois du Numérique et des Mathématiques (IBNM), Université de Brest (UBO)-Université européenne de Bretagne - European University of Brittany (UEB)-École Nationale Supérieure de Techniques Avancées Bretagne (ENSTA Bretagne)-Institut Mines-Télécom [Paris] (IMT)-Centre National de la Recherche Scientifique (CNRS)-École Nationale d'Ingénieurs de Brest (ENIB)-Université de Bretagne Sud (UBS)-Université de Brest (UBO)-Télécom Bretagne-Institut Brestois du Numérique et des Mathématiques (IBNM), Université de Brest (UBO)-Université européenne de Bretagne - European University of Brittany (UEB)-École Nationale Supérieure de Techniques Avancées Bretagne (ENSTA Bretagne)-Institut Mines-Télécom [Paris] (IMT)-Centre National de la Recherche Scientifique (CNRS), State Key Laboratory of Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Laboratory of Virology, CHU Rouen, Normandie Université (NU)-Normandie Université (NU), Universidade Federal do Rio de Janeiro (UFRJ), Departements of Medicine and Microbiology, University of Alabama [Tuscaloosa] (UA), Stichting HIV Monitoring [Amsterdam], Universiteit van Amsterdam (UvA), Département Génétique Internal Médecine, Hôpital Universitaire Leuven, Department of Chemical and Biomolecular Engineering [Baltimore], Johns Hopkins University (JHU), Centers for Disease Control and Prevention [Atlanta] (CDC), Centers for Disease Control and Prevention, China Academy of Chinese Medical Sciences, Chatzidimitriou, Dimitrios [0000-0001-9656-5898], University of Oxford, Institut Pasteur [Paris] (IP), Istituto Superiore di Sanità (ISS), Microbiologie Fondamentale et Pathogénicité (MFP), Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Recherches Translationnelles sur le VIH et les maladies infectieuses endémiques et émergentes (TransVIHMI), Institut de Recherche pour le Développement (IRD)-Université de Yaoundé I-Université Cheikh Anta Diop [Dakar, Sénégal] (UCAD)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), University of KwaZulu-Natal [Durban, Afrique du Sud] (UKZN)-Medical Research Council of South Africa, Université européenne de Bretagne - European University of Brittany (UEB)-École Nationale d'Ingénieurs de Brest (ENIB)-Université de Bretagne Sud (UBS)-Université de Brest (UBO)-Télécom Bretagne-Institut Brestois du Numérique et des Mathématiques (IBNM), Université de Brest (UBO)-École Nationale Supérieure de Techniques Avancées Bretagne (ENSTA Bretagne)-Institut Mines-Télécom [Paris] (IMT)-Centre National de la Recherche Scientifique (CNRS)-Université européenne de Bretagne - European University of Brittany (UEB)-École Nationale d'Ingénieurs de Brest (ENIB)-Université de Bretagne Sud (UBS)-Université de Brest (UBO)-Télécom Bretagne-Institut Brestois du Numérique et des Mathématiques (IBNM), Université de Brest (UBO)-École Nationale Supérieure de Techniques Avancées Bretagne (ENSTA Bretagne)-Institut Mines-Télécom [Paris] (IMT)-Centre National de la Recherche Scientifique (CNRS), UNAIDS, Noguchi Memorial Institute for Medical Research, University of Ghana, Laboratoire d'Electronique et des Technologies de l'Information (CEA-LETI), Université Grenoble Alpes (UGA)-Direction de Recherche Technologique (CEA) (DRT (CEA)), Beihang University, Recherches Translationnelles sur le VIH et les maladies infectieuses (TransVIHMI), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Recherche pour le Développement (IRD)-Université Montpellier 1 (UM1)-Université Cheikh Anta Diop [Dakar, Sénégal] (UCAD)-Universtié Yaoundé 1 [Cameroun]-Université de Montpellier (UM), Universidade Federal do Rio de Janeiro [Rio de Janeiro] (UFRJ), Nuffield Department of Women's and Reproductive Health (NDWRH), University of Oxford- John Radcliffe Hospital [Oxford University Hospital], South African Centre for Epidemiological Modelling and Analysis, JH is supported by the Oxford University Clinical Academic Graduate School (Oxford, UK) and Linacre College, Oxford University (Oxford, UK)., WHO–UNAIDS Network for HIV Isolation Characterisation : Alash'le G Abimiku, Simon Agwale, Chris Archibald, Boaz Avidor, María Gabriela Barbás, Francoise Barre-Sinoussi, Banson Barugahare, El Hadj Belabbes, Silvia Bertagnolio, Deborah Birx, Aleksei F Bobkov, James Brandful, Helba Bredell, Catherine A Brennan, James Brooks, Marie Bruckova, Luigi Buonaguro, Franco Buonaguro, Stefano Buttò, Anne Buve, Mary Campbell, Jean Carr, Alex Carrera, Manuel Gómez Carrillo, Connie Celum, Beth Chaplin, Macarthur Charles, Dimitrios Chatzidimitriou, Zhiwei Chen, Katsumi Chijiwa, David Cooper, Philip Cunningham, Anoumou Dagnra, Cillian F de Gascun, Julia Del Amo, Elena Delgado, Ursula Dietrich, Dominic Dwyer, Dennis Ellenberger, Barbara Ensoli, Max Essex, Feng Gao, Herve Fleury, Peter N Fonjungo, Vincent Foulongne, Deepak A Gadkari, Federico García, Roger Garsia, Guy Michel Gershy-Damet, Judith R Glynn, Ruth Goodall, Zehava Grossman, Monick Lindenmeyer-Guimarães, Beatrice Hahn, Raph L Hamers, Osamah Hamouda, Ray Handema, Xiang He, Joshua Herbeck, David D Ho, Africa Holguin, Mina Hosseinipour, Gillian Hunt, Masahiko Ito, Mohamed Ali Bel Hadj Kacem, Erin Kahle, Pontiano Kaleebu Kaleebu, Marcia Kalish, Adeeba Kamarulzaman, Chun Kang, Phyllis Kanki, Edward Karamov, Jean-Claude Karasi, Kayitesi Kayitenkore, Tony Kelleher, Dwip Kitayaporn, Leondios G Kostrikis, Claudia Kucherer, Claudia Lara, Thomas Leitner, Kirsi Liitsola, Jai Lingappa, Marek Linka, Ivette Lorenzana de Rivera, Vladimir Lukashov, Shlomo Maayan, Luzia Mayr, Francine McCutchan, Nicolas Meda, Elisabeth Menu, Fred Mhalu, Doreen Mloka, John L Mokili, Brigitte Montes, Orna Mor, Mariza Morgado, Fausta Mosha, Awatef Moussi, James Mullins, Rafael Najera, Mejda Nasr, Nicaise Ndembi, Joel R Neilson, Vivek R Nerurkar, Florian Neuhann, Claudine Nolte, Vlad Novitsky, Philippe Nyambi, Marianna Ofner, Fem J Paladin, Anna Papa, Jean Pape, Neil Parkin, Chris Parry, Martine Peeters, Alexandra Pelletier, Lucía Pérez-Álvarez, Deenan Pillay, Angie Pinto, Trinh Duy Quang, Cecilia Rademeyer, Filimone Raikanikoda, Mark A Rayfield, Jean-Marc Reynes, Tobias Rinke de Wit, Kenneth E Robbins, Morgane Rolland, Christine Rousseau, Jesus Salazar-Gonzales, Hanan Salem, Mika Salminen, Horacio Salomon, Paul Sandstrom, Mario L Santiago, Abdoulaye D Sarr, Bryan Schroeder, Michel Segondy, Philippe Selhorst, Sylvester Sempala, Jean Servais, Ansari Shaik, Yiming Shao, Amine Slim, Marcelo A Soares, Elijah Songok, Debbie Stewart, Julie Stokes, Shambavi Subbarao, Ruengpung Sutthent, Jun Takehisa, Amilcar Tanuri, Kok Keng Tee, Kiran Thapa, Michael Thomson, Tyna Tran, Willy Urassa, Hiroshi Ushijima, Philippevan de Perre, Guidovan der Groen, Kristel van Laethem, Joep van Oosterhout, Ard van Sighem, Eric van Wijngaerden, Anne-Mieke Vandamme, Jurgen Vercauteren, Nicole Vidal, Lesley Wallace, Carolyn Williamson, Dawit Wolday, Jianqing Xu, Chunfu Yang, Linqi Zhang, and Rong Zhang
- Subjects
0301 basic medicine ,Serotype ,Genotype ,Genotyping Techniques ,030106 microbiology ,DIVERSITY ,MULTICENTER ,VACCINE ,HIV Infections ,Genome, Viral ,Biology ,Global Health ,Serogroup ,SUBTYPES ,03 medical and health sciences ,[SDV.MHEP.MI]Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,Surveys and Questionnaires ,INFECTION ,Genetic variation ,Global health ,Humans ,HIV vaccine ,Serotyping ,AIDS Vaccines ,Science & Technology ,Molecular epidemiology ,Genetic Variation ,Subtyping ,3. Good health ,Trend analysis ,030104 developmental biology ,Infectious Diseases ,13. Climate action ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,HIV-1 ,[SDV.SPEE]Life Sciences [q-bio]/Santé publique et épidémiologie ,Life Sciences & Biomedicine ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,Demography - Abstract
BACKGROUND: Global genetic diversity of HIV-1 is a major challenge to the development of HIV vaccines. We aimed to estimate the regional and global distribution of HIV-1 subtypes and recombinants during 1990-2015. METHODS: We searched PubMed, EMBASE (Ovid), CINAHL (Ebscohost), and Global Health (Ovid) for HIV-1 subtyping studies published between Jan 1, 1990, and Dec 31, 2015. We collected additional unpublished HIV-1 subtyping data through a global survey. We included prevalence studies with HIV-1 subtyping data collected during 1990-2015. We grouped countries into 14 regions and analysed data for four time periods (1990-99, 2000-04, 2005-09, and 2010-15). The distribution of HIV-1 subtypes, circulating recombinant forms (CRFs), and unique recombinant forms (URFs) in individual countries was weighted according to the UNAIDS estimates of the number of people living with HIV (PLHIV) in each country to generate regional and global estimates of HIV-1 diversity in each time period. The primary outcome was the number of samples designated as HIV-1 subtypes A, B, C, D, F, G, H, J, K, CRFs, and URFs. The systematic review is registered with PROSPERO, number CRD42017067164. FINDINGS: This systematic review and global survey yielded 2203 datasets with 383 519 samples from 116 countries in 1990-2015. Globally, subtype C accounted for 46·6% (16 280 897/34 921 639 of PLHIV) of all HIV-1 infections in 2010-15. Subtype B was responsible for 12·1% (4 235 299/34 921 639) of infections, followed by subtype A (10·3%; 3 587 003/34 921 639), CRF02_AG (7·7%; 2 705 110/34 921 639), CRF01_AE (5·3%; 1 840 982/34 921 639), subtype G (4·6%; 1 591 276/34 921 639), and subtype D (2·7%; 926 255/34 921 639). Subtypes F, H, J, and K combined accounted for 0·9% (311 332/34 921 639) of infections. Other CRFs accounted for 3·7% (1 309 082/34 921 639), bringing the proportion of all CRFs to 16·7% (5 844 113/34 921 639). URFs constituted 6·1% (2 134 405/34 921 639), resulting in recombinants accounting for 22·8% (7 978 517/34 921 639) of all global HIV-1 infections. The distribution of HIV-1 subtypes and recombinants changed over time in countries, regions, and globally. At a global level during 2005-15, subtype B increased, subtypes A and D were stable, and subtypes C and G and CRF02_AG decreased. CRF01_AE, other CRFs, and URFs increased, leading to a consistent increase in the global proportion of recombinants over time. INTERPRETATION: Global and regional HIV diversity is complex and evolving, and is a major challenge to HIV vaccine development. Surveillance of the global molecular epidemiology of HIV-1 remains crucial for the design, testing, and implementation of HIV vaccines. FUNDING: None. ispartof: LANCET INFECTIOUS DISEASES vol:19 issue:2 pages:143-155 ispartof: location:United States status: published
- Published
- 2018
- Full Text
- View/download PDF
15. Interplay between transcription regulators RUNX1 and FUBP1 activates an enhancer of the oncogene c-KIT and amplifies cell proliferation
- Author
-
Lydie, Debaize, Hélène, Jakobczyk, Stéphane, Avner, Jérémie, Gaudichon, Anne-Gaëlle, Rio, Aurélien A, Sérandour, Lena, Dorsheimer, Frédéric, Chalmel, Jason S, Carroll, Martin, Zörnig, Michael A, Rieger, Olivier, Delalande, Gilles, Salbert, Marie-Dominique, Galibert, Virginie, Gandemer, Marie-Bérengère, Troadec, Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes (UR)-Centre National de la Recherche Scientifique (CNRS)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Integrative Oncogenomics of Multiple Myeloma Pathogenesis and Progression (CRCINA-ÉQUIPE 11), Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes), École Centrale de Nantes (ECN), Department of Medicine, Hematology/Oncology [Frankfurt, Germany], Goethe-University Frankfurt am Main, Institut de recherche en santé, environnement et travail (Irset), Université d'Angers (UA)-Université de Rennes (UR)-École des Hautes Études en Santé Publique [EHESP] (EHESP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Cancer Research UK [Cambridge, UK] (Cambridge Institute), University of Cambridge [UK] (CAM), Georg-Speyer-Haus [Frankfurt, Germany], Institute for Tumor Biology and Experimental Therapy [Frankfurt, Germany], Génétique Somatique des Cancers [CHU Rennes], CHU Pontchaillou [Rennes], Département d'oncohématologie pédiatrique [CHU Rennes], Ligue Régionale contre le cancer [comité 22,35,56,79,41 to M.B.T., L.D., V.G.], SFR Biosit UMS CNRS 3480-INSERM 018 (to M.B.T.), Région Bretagne (to L.D., M.B.T.), The Société Française d’Hématologie (to L.D.), Rennes Métropole (to M.B.T.), French Research Ministry (to H.J.), FHUCAMIN (to J.G., V.G.), the société française de lutte contre les cancers et les leucémies de l’enfant et de l’adolescent and the Fédération Enfants et Santé (to M.B.T.), a private donator Mrs M-Dominique Blanc-Bert (to M.B.T.), Cancéropole Grand Ouest (to L.D.), The Société Française de Biochimie et Biologie Moléculaire (to H.J.), CNRS, Université de Rennes 1 and the People Programme (Marie Curie Actions) of the European Union’s Seventh Framework Programme [FP7/2007-2013] under REA grant agreement [291851 to M.B.T.]. Funding for open access charge: People Programme (Marie Curie Actions) of the European Union’s Seventh Framework Programme., European Project: 291851,EC:FP7:PEOPLE,FP7-PEOPLE-2011-CIG,ALLRUN(2012), Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique )-Centre National de la Recherche Scientifique (CNRS)-Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA), Université d'Angers (UA)-Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-École des Hautes Études en Santé Publique [EHESP] (EHESP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Carroll, Jason [0000-0003-3643-0080], Apollo - University of Cambridge Repository, Bernardo, Elizabeth, and Modeling TEL/AML1 childhood lymphoblastic leukemia in zebrafish - ALLRUN - - EC:FP7:PEOPLE2012-02-01 - 2016-01-31 - 291851 - VALID
- Subjects
Transcription, Genetic ,Primary Cell Culture ,Antineoplastic Agents ,Bone Marrow Cells ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Mice ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Mice, Inbred NOD ,Cell Line, Tumor ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,hemic and lymphatic diseases ,Animals ,Humans ,ddc:610 ,Cell Proliferation ,Binding Sites ,Base Sequence ,Gene Expression Regulation, Leukemic ,Precursor Cells, B-Lymphoid ,Gene regulation, Chromatin and Epigenetics ,RNA-Binding Proteins ,Xenograft Model Antitumor Assays ,Chromatin ,DNA-Binding Proteins ,Proto-Oncogene Proteins c-kit ,Enhancer Elements, Genetic ,HEK293 Cells ,Core Binding Factor Alpha 2 Subunit ,embryonic structures ,Imatinib Mesylate ,Protein Binding ,Signal Transduction - Abstract
International audience; Runt-related transcription factor 1 (RUNX1) is a well-known master regulator of hematopoietic lineages but its mechanisms of action are still not fully understood. Here, we found that RUNX1 localizes on active chromatin together with Far Upstream Binding Protein 1 (FUBP1) in human B-cell precursor lymphoblasts, and that both factors interact in the same transcriptional regulatory complex. RUNX1 and FUBP1 chromatin localization identified c-KIT as a common target gene. We characterized two regulatory regions, at +700 bp and +30 kb within the first intron of c-KIT, bound by both RUNX1 and FUBP1, and that present active histone marks. Based on these regions, we proposed a novel FUBP1 FUSE-like DNA-binding sequence on the +30 kb enhancer. We demonstrated that FUBP1 and RUNX1 cooperate for the regulation of the expression of the onco-gene c-KIT. Notably, upregulation of c-KIT expression by FUBP1 and RUNX1 promotes cell proliferation and renders cells more resistant to the c-KIT inhibitor imatinib mesylate, a common therapeutic drug. These results reveal a new mechanism of action of RUNX1 that implicates FUBP1, as a facilitator, to trigger transcriptional regulation of c-KIT and to regulate cell proliferation. Deregulation of this regulatory mechanism may explain some oncogenic function of RUNX1 and FUBP1.
- Published
- 2018
- Full Text
- View/download PDF
16. A point mutation in the Ncr1 signal peptide impairs the development of innate lymphoid cell subsets
- Author
-
Ariane Giannattasio, Sophie Ugolini, Marco J Herold, Thierry Walzer, Andrew J. Kueh, Wei Shi, Alexander Steinle, Tobias Zöller, Francisca F. Almeida, Miriam E. Friede, Kylie Luong, Bushra Rais, Nicholas D. Huntington, Justine Galluso, Joachim Koch, Emilie Narni-Mancinelli, Matthew A. Firth, David T. Scadden, Sara Tognarelli, Simon N. Willis, Evelyn Ullrich, Fabrice Faure, Yang Liao, Ulrike Schleicher, Antoine Marçais, Eric Vivier, Sandra Weil, Francois Mercier, Gabrielle T. Belz, Andreas G. Chiocchetti, Franziska Kalensee, Francesco Spallotta, The Walter and Eliza Hall Institute of Medical Research (WEHI), Goethe-Universität Frankfurt am Main, Réponse immunitaire innée dans les maladies infectieuses et auto-immunes – Innate immunity in infectious and autoimmune diseases, Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Department of Medecine [Montréal], McGill University = Université McGill [Montréal, Canada], Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Harvard Stem Cell Institute [Cambridge, USA] (HSCI), Harvard University, Georg-Speyer-Haus, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Innate Pharma, Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU), and Harvard University [Cambridge]
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Signal peptide ,intracellular trafficking ,Immunology ,Cell ,Congenic ,innate lymphoid cells ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immunology and Allergy ,ddc:610 ,congenic mice ,Receptor ,Original Research ,Mutation ,Endoplasmic reticulum ,Innate lymphoid cell ,HEK 293 cells ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,activation receptors ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,lcsh:RC581-607 ,030215 immunology - Abstract
International audience; NKp46 (CD335) is a surface receptor shared by both human and mouse natural killer (NK) cells and innate lymphoid cells (ILCs) that transduces activating signals necessary to eliminate virus-infected cells and tumors. Here, we describe a spontaneous point mutation of cysteine to arginine (C14R) in the signal peptide of the NKp46 protein in congenic Ly5.1 mice and the newly generated NCR(B6C14R) strain. Ly5.1(C14R) NK cells expressed similar levels of Ncr1 mRNA as C57BL/6, but showed impaired surface NKp46 and reduced ability to control melanoma tumors in vivo. Expression of the mutant NKp46(C14R) in 293T cells showed that NKp46 protein trafficking to the cell surface was compromised. Although Ly5.1(C14R) mice had normal number of NK cells, they showed an increased number of early maturation stage NK cells. CD49a(+)ILC1s were also increased but these cells lacked the expression of TRAIL. ILC3s that expressed NKp46 were not detectable and were not apparent when examined by T-bet expression. Thus, the C14R mutation reveals that NKp46 is important for NK cell and ILC differentiation, maturation and function. Significance Innate lymphoid cells (ILCs) play important roles in immune protection. Various subsets of ILCs express the activating receptor NKp46 which is capable of recognizing pathogen derived and tumor ligands and is necessary for immune protection. Here, we describe a spontaneous point mutation in the signal peptide of the NKp46 protein in congenic Ly5.1 mice which are widely used for tracking cells in vivo. This Ncr1 C14R mutation impairs NKp46 surface expression resulting in destabilization of Ncr1 and accumulation of NKp46 in the endoplasmic reticulum. Loss of stable NKp46 expression impaired the maturation of NKp46(+) ILCs and altered the expression of TRAIL and T-bet in ILC1 and ILC3, respectively.
- Published
- 2018
- Full Text
- View/download PDF
17. The Hard Way towards an Antibody-Based HIV-1 Env Vaccine: Lessons from Other Viruses
- Author
-
Hildegard Büning, Jutta Eichler, Patrice Debré, Vincent Vieillard, Ursula Dietrich, Oliver Ringel, HAL-UPMC, Gestionnaire, Georg-Speyer-Haus, Centre d'Immunologie et de Maladies Infectieuses (CIMI), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Hannover Medical School [Hannover] (MHH), German Center for Infection Research - partner site Hannover-Braunschweig (DZIF), Institute for Virology, University of Cologne, and Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)
- Subjects
Env ,0301 basic medicine ,lcsh:QR1-502 ,epitope vaccine ,vectored vaccine ,Drug Evaluation, Preclinical ,HIV Infections ,Viremia ,Review ,HIV Antibodies ,Biology ,lcsh:Microbiology ,Epitope ,Virus ,03 medical and health sciences ,[SDV.IMM.VAC] Life Sciences [q-bio]/Immunology/Vaccinology ,vaccine ,Virology ,Drug Discovery ,medicine ,Animals ,Humans ,ddc:610 ,ComputingMilieux_MISCELLANEOUS ,[SDV.MP.VIR] Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,AIDS Vaccines ,broadly neutralizing antibodies ,adeno-associated viruses (AAV) ,Reverse vaccinology ,env Gene Products, Human Immunodeficiency Virus ,Naturwissenschaftliche Fakultät ,medicine.disease ,Antibodies, Neutralizing ,3. Good health ,Vaccination ,Chronic infection ,030104 developmental biology ,Infectious Diseases ,Immunization ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,HIV-1 ,structure-based reverse vaccinology ,[SDV.IMM.VAC]Life Sciences [q-bio]/Immunology/Vaccinology ,Vaccine failure - Abstract
International audience; Although effective antibody-based vaccines have been developed against multiple viruses, such approaches have so far failed for the human immunodeficiency virus type 1 (HIV-1). Despite the success of anti-retroviral therapy (ART) that has turned HIV-1 infection into a chronic disease and has reduced the number of new infections worldwide, a vaccine against HIV-1 is still urgently needed. We discuss here the major reasons for the failure of " classical " vaccine approaches, which are mostly due to the biological properties of the virus itself. HIV-1 has developed multiple mechanisms of immune escape, which also account for vaccine failure. So far, no vaccine candidate has been able to induce broadly neutralizing antibodies (bnAbs) against primary patient viruses from different clades. However, such antibodies were identified in a subset of patients during chronic infection and were shown to protect from infection in animal models and to reduce viremia in first clinical trials. Their detailed characterization has guided structure-based reverse vaccinology approaches to design better HIV-1 envelope (Env) immunogens. Furthermore, conserved Env epitopes have been identified, which are promising candidates in view of clinical applications. Together with new vector-based technologies, considerable progress has been achieved in recent years towards the development of an effective antibody-based HIV-1 vaccine.
- Published
- 2018
- Full Text
- View/download PDF
18. The Fas ligand intracellular domain is released by ADAM10 and SPPL2a cleavage in T-cells
- Author
-
B. Martoglio, Sébastien Huault, E. Friedmann, Martin Zörnig, F. Guardiola-Serrano, W. S. Wels, Katharina Lückerath, N. Novac, Nathalie Cahuzac, Anne-Odile Hueber, Vladimir Kirkin, Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Institut de signalisation, biologie du développement et cancer (ISBDC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), Institute of Biochemistry, Eidgenössische Technische Hochschule - Swiss Federal Institute of Technology [Zürich] (ETH Zürich), MerckKGaA, Merck & Co. Inc, and Novartis Institutes for BioMedical Research (NIBR)
- Subjects
MESH: ADAM Proteins ,Signal peptide ,Proteases ,MESH: Cell Line, Tumor ,Fas Ligand Protein ,T-Lymphocytes ,Medizin ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,chemical and pharmacologic phenomena ,Fas ligand ,MESH: Protein Structure, Tertiary ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,MESH: RNA, Small Interfering ,Disintegrin ,MESH: Microscopy, Confocal ,Aspartic Acid Endopeptidases ,Humans ,RNA, Small Interfering ,Molecular Biology ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,MESH: Humans ,Microscopy, Confocal ,biology ,MESH: Aspartic Endopeptidases ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,hemic and immune systems ,Cell Biology ,Fas receptor ,Molecular biology ,MESH: Fas Ligand Protein ,Transmembrane protein ,Protein Structure, Tertiary ,Cell biology ,ADAM Proteins ,MESH: T-Lymphocytes ,Apoptosis ,030220 oncology & carcinogenesis ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Intracellular ,MESH: Cells, Cultured - Abstract
Fas ligand (FasL) is a type II transmembrane protein belonging to the tumor necrosis factor family. Its binding to the cognate Fas receptor triggers the apoptosis that plays a pivotal role in the maintenance of immune system homeostasis. The cell death-inducing property of FasL has been associated with its extracellular domain, which can be cleaved off by metalloprotease activity to produce soluble FasL. The fate of the remaining membrane-anchored N-terminal part of the FasL molecule has not been determined. Here we show that post-translational processing of overexpressed and endogenous FasL in T-cells by the disintegrin and metalloprotease ADAM10 generates a 17-kDa N-terminal fragment, which lacks the receptor-binding extracellular domain. This FasL remnant is membrane anchored and further processed by SPPL2a, a member of the signal peptide peptidase-like family of intramembrane-cleaving proteases. SPPL2a cleavage liberates a smaller and highly unstable fragment mainly containing the intracellular FasL domain (FasL ICD). We show that this fragment translocates to the nucleus and is capable of inhibiting gene transcription. With ADAM10 and SPPL2a we have identified two proteases implicated in FasL processing and release of the FasL ICD, which has been shown to be important for retrograde FasL signaling.
- Published
- 2007
- Full Text
- View/download PDF
19. Exclusive transduction of human CD4+ T Cells upon systemic delivery of CD4-targeted lentiviral vectors
- Author
-
Alexandra Trkola, Dorothee von Laer, Qi Zhou, Robert C. Münch, Cheick Coulibaly, Janine Kimpel, Winfried S. Wels, Anett Pfeiffer, Anke Muth, Camille Lévy, Katharina M. Uhlig, Els Verhoeyen, Janna Seifried, Udo F. Hartwig, Christian J. Buchholz, University of Zurich, Buchholz, C J, Paul-Ehrlich Institut, The Medical University of Innsbruck, Virus enveloppés, vecteurs et immunothérapie – Enveloped viruses, Vectors and Immuno-therapy (EVIR), Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Universität Zürich [Zürich] = University of Zurich (UZH), Paul-Ehrlich-Institute, Georg-Speyer-Haus [Frankfurt, Germany], Institute for Tumor Biology and Experimental Therapy [Frankfurt, Germany], University Medical Center of the Johannes Gutenberg-University, Centre méditerranéen de médecine moléculaire (C3M), Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM), German Cancer Consortium [Heidelberg] (DKTK), Innsbruck Medical University = Medizinische Universität Innsbruck (IMU), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Johannes Gutenberg - Universität Mainz = Johannes Gutenberg University (JGU), Université Nice Sophia Antipolis (1965 - 2019) (UNS), and COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Côte d'Azur (UCA)
- Subjects
CD4-Positive T-Lymphocytes ,10028 Institute of Medical Virology ,Cell Transplantation ,Genetic enhancement ,Adoptive ,Mice, SCID ,Immunotherapy, Adoptive ,Interleukin 21 ,Mice ,Mice, Inbred NOD ,Transduction, Genetic ,Bone Marrow ,Leukocytes ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Luciferases ,Cells, Cultured ,Mice, Knockout ,Heterologous ,Tumor ,Cultured ,Forkhead Transcription Factors ,Acquired immune system ,Flow Cytometry ,3. Good health ,Cell biology ,medicine.anatomical_structure ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,2723 Immunology and Allergy ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Immunotherapy ,Regulatory T cell ,Cells ,Knockout ,Transplantation, Heterologous ,Immunology ,Mononuclear ,Genetic Vectors ,Green Fluorescent Proteins ,610 Medicine & health ,Streptamer ,Thymus Gland ,Biology ,SCID ,Cell Line ,Transduction ,Genetic ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Interleukin 3 ,Transplantation ,2403 Immunology ,Lentivirus ,Genetic Therapy ,Molecular biology ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,HEK293 Cells ,Leukocytes, Mononuclear ,Inbred NOD ,570 Life sciences ,biology ,Spleen - Abstract
Playing a central role in both innate and adaptive immunity, CD4+ T cells are a key target for genetic modifications in basic research and immunotherapy. In this article, we describe novel lentiviral vectors (CD4-LV) that have been rendered selective for human or simian CD4+ cells by surface engineering. When applied to PBMCs, CD4-LV transduced CD4+ but not CD4− cells. Notably, also unstimulated T cells were stably genetically modified. Upon systemic or intrasplenic administration into mice reconstituted with human PBMCs or hematopoietic stem cells, reporter gene expression was predominantly detected in lymphoid organs. Evaluation of GFP expression in organ-derived cells and blood by flow cytometry demonstrated exclusive gene transfer into CD4+ human lymphocytes. In bone marrow and spleen, memory T cells were preferentially hit. Toward therapeutic applications, we also show that CD4-LV can be used for HIV gene therapy, as well as for tumor therapy, by delivering chimeric Ag receptors. The potential for in vivo delivery of the FOXP3 gene was also demonstrated, making CD4-LV a powerful tool for inducible regulatory T cell generation. In summary, our work demonstrates the exclusive gene transfer into a T cell subset upon systemic vector administration opening an avenue toward novel strategies in immunotherapy.
- Published
- 2015
- Full Text
- View/download PDF
20. Immune modulation by Fas ligand reverse signaling: lymphocyte proliferation is attenuated by the intracellular Fas ligand domain
- Author
-
Markus Ollert, Martin Hrabé de Angelis, Vladimir Kirkin, Anne-Odile Hueber, Ursula Zimber-Strobl, Wiebke Milani, Geert Michel, Helmut Fuchs, Thure Adler, Antonio Aguilar-Pimentel, Krishnaraj Rajalingam, Lothar J. Strobl, Dirk H. Busch, Johannes Beckers, Marion Horsch, Inga Maria Melzer, Frank J. T. Staal, Valerie Gailus-Durner, Martin Zörnig, Frederic B. Thalheimer, Katharina Lückerath, Dagmar Siele, Institute of Immunology, University Hospital Schleswig-Holstein, GSF research center, Institute of Experimental Genetis, Institute for Experimental Genetics, GSF - National Research Center for Environment and Health, Institute of Developmental Biology and Cancer (IBDC), Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), and Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus
- Subjects
Fas Ligand Protein ,PROTEIN-TYROSINE-PHOSPHATASE ,T-CELLS ,B-CELLS ,SECRETORY LYSOSOMES ,CYTOPLASMIC TAIL ,SCHWANN-CELLS ,EXPRESSION ,ACTIVATION ,DEATH ,WNT ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Medizin ,chemical and pharmacologic phenomena ,Lymphocyte proliferation ,Biology ,Lymphocyte Activation ,Biochemistry ,Fas ligand ,Immunomodulation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Gene Knock-In Techniques ,Protein kinase A ,Transcription factor ,[SDV.BDD]Life Sciences [q-bio]/Development Biology ,030304 developmental biology ,Cell Proliferation ,Mice, Knockout ,0303 health sciences ,B-Lymphocytes ,Reverse Transcriptase Polymerase Chain Reaction ,Germinal center ,hemic and immune systems ,Cell Biology ,Hematology ,Molecular biology ,protein-tyrosine-phosphatase t-cells b-cells secretory lysosomes cytoplasmic tail schwann-cells expression activation death wnt ,Cell biology ,Cytokine ,Gene Expression Regulation ,Apoptosis ,Signal transduction ,030215 immunology ,Signal Transduction - Abstract
Fas ligand (FasL) not only induces apoptosis in Fas receptor-bearing target cells, it is also able to transmit signals into the FasL-expressing cell via its intracellular domain (ICD). Recently, we described a Notch-like proteolytic processing of FasL that leads to the release of the FasL ICD into the cytoplasm and subsequent translocation into the nucleus where it may influence gene transcription. To study the molecular mechanism underlying such reverse FasL signaling in detail and to analyze its physiological importance in vivo, we established a knockout/knockin mouse model, in which wild-type FasL was replaced with a deletion mutant lacking the ICD. Our results demonstrate that FasL ICD signaling impairs activation-induced proliferation in B and T cells by diminishing phosphorylation of phospholipase C γ, protein kinase C, and extracellular signal-regulated kinase 1/2. We also demonstrate that the FasL ICD interacts with the transcription factor lymphoid-enhancer binding factor-1 and inhibits lymphoid-enhancer binding factor-1–dependent transcription. In vivo, plasma cell numbers, generation of germinal center B cells, and, consequently, production of antigen-specific immunoglobulin M antibodies in response to immunization with T cell–dependent or T cell–independent antigen are negatively affected in presence of the FasL ICD, suggesting that FasL reverse signaling participates in negative fine-tuning of certain immune responses.
- Published
- 2011
- Full Text
- View/download PDF
21. Funktionelle Charakterisierung der SANT-Domänen des Korepressors N-CoR
- Author
-
Tiefenbach, Jens and Georg Speyer Haus, Chemotherapeutisches Forschungsinstitut
- Subjects
N-CoR ,Korepressor ,ddc:570 ,Hefe-Zwei-Hybrid-Screen ,Life sciences - Abstract
Der Korepressor N-CoR vermittelt die Repression von nukleären Hormonrezptoren in Abwesenheit ihrer Liganden und ist darber hinaus für die embryonale Entwicklung von Säugern entscheidend. N-CoR ist in der Zelle mit Histondeacetylasen (HDACs) komplexiert. Diese Enzyme bewirken im Zusammenspiel mit ihren Gegenspielern, den Histonacetyltransferasen, durch Deacetylierung und Acetylierung von Histonen eine dynamische Modifikation des Chromatins und beeinflussen so die Transkription von Genen. Die Interaktion zwischen regulatorischen Proteinen, Histonen und histonmodifizierenden Proteinen ist ein fundamentaler und konservierter Mechanismus der Genregulation in höheren Eukaryonten. Koregulatoren (Koaktivatoren und Korepressoren) und Transkriptionsfaktoren enthalten eine Fülle noch uncharakterisierter Domänen, die in ähnlicher Weise wirken könnten. Der Korepressor N-CoR enthält beispielsweise zwei uncharakterisierte SANT-Domänen. Die SANT-Domäne ist zwischen Hefe und Sugern konserviert und spielt vermutlich in der Chromatinmodifizierung oder Transkription eine Rolle. In der vorliegenden Arbeit wurden verschiedene Screeningmethoden zur Identifikation von Interaktionspartnern der N-CoR SANT-Domänen eingesetzt. In einem Hefe-Zwei-Hybrid Screen war es möglich, Interaktionspartner der N-CoR SANT-Domäne zu isolieren. Insgesamt 14 verschiedene Proteine wurden in dem Hefescreen identifiziert. Die Proteine PIAS1, Ubc9, TDG, Hoxa-4, TAFII250 und cDNA I interagierten in vitro mit der N-CoR SANT1-Domäne. Drei der im Hefescreen gefundenen Proteine (PIAS1, Ubc9 und Pc2) deuteten darauf hin, dass N-CoR durch die Konjugation von SUMO-Proteinen posttranslational modifiziert sein könnte. In Ko-Immunopräziptitationen konnte die Interaktion zwischen N-CoR und der SUMO-E3-Ligase PIAS1 in vivo bestätigt werden. Die N-CoR SUMO-Modifikation wurde indirekt in vivo und in einem in vitro Sumoylierungstest nachgewiesen. Endogenes N-CoR und PIAS1 kolokalisieren im Zellkern, wobei die heterologe Expression der SUMO-E3 Ligase die für N-CoR typische Aggregatbildung im Kern verhindert und eine diffusere Verteilung und eine Häufung in der Nähe der Kernmembran induziert. In Kolokalisierungsstudien konnte ferner gezeigt werden, dass eine N-CoR-DSUMO Konsensussequenzmutante eine veränderte zytoplasmatisch-nukleäre Lokalisation aufweist. Dies traf ebenfalls auf DSANT-Mutanten von N-CoR zu, die im Vergleich zum N-CoR Wildtypprotein eine unterschiedliche Lokalisation bei heterologer Expression von PIAS1 zeigten. Die Koexpression von PIAS1 reduzierte die Repression eines Luziferasereporters durch N-CoR. Andererseits war die Repression des Proteins ebenfalls durch Alanin-Substitution in potentiellen SUMO-Stellen vermindert.
- Published
- 2003
22. Author Correction: Octyl itaconate enhances VSVΔ51 oncolytic virotherapy by multitarget inhibition of antiviral and inflammatory pathways.
- Author
-
Kurmasheva N, Said A, Wong B, Kinderman P, Han X, Rahimic AHF, Kress A, Carter-Timofte ME, Holm E, van der Horst D, Kollmann CF, Liu Z, Wang C, Hoang HD, Kovalenko E, Chrysopoulou M, Twayana KS, Ottosen RN, Svenningsen EB, Begnini F, Kiib AE, Kromm FEH, Weiss HJ, Di Carlo D, Muscolini M, Higgins M, van der Heijden M, Arulanandam R, Bardoul A, Tong T, Ozsvar A, Hou WH, Schack VR, Holm CK, Zheng Y, Ruzek M, Kalucka J, de la Vega L, Elgaher WAM, Korshoej AR, Lin R, Hiscott J, Poulsen TB, O'Neill LA, Roy DG, Rinschen MM, van Montfoort N, Diallo JS, Farin HF, Alain T, and Olagnier D
- Published
- 2024
- Full Text
- View/download PDF
23. Adult skull bone marrow is an expanding and resilient haematopoietic reservoir.
- Author
-
Koh BI, Mohanakrishnan V, Jeong HW, Park H, Kruse K, Choi YJ, Nieminen-Kelhä M, Kumar R, Pereira RS, Adams S, Lee HJ, Bixel MG, Vajkoczy P, Krause DS, and Adams RH
- Subjects
- Animals, Female, Male, Mice, Pregnancy, Adipogenesis, Cytokines metabolism, Femur cytology, Mice, Inbred C57BL, Stem Cell Niche genetics, Stroke pathology, Humans, Young Adult, Adult, Middle Aged, Aged, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Inflammation Mediators metabolism, Aging genetics, Aging physiology, Bone Marrow metabolism, Hematopoiesis genetics, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Skull anatomy & histology, Skull blood supply, Skull cytology
- Abstract
The bone marrow microenvironment is a critical regulator of haematopoietic stem cell self-renewal and fate
1 . Although it is appreciated that ageing, chronic inflammation and other insults compromise bone marrow function and thereby negatively affect haematopoiesis2 , it is not known whether different bone compartments exhibit distinct microenvironmental properties and functional resilience. Here we use imaging, pharmacological approaches and mouse genetics to uncover specialized properties of bone marrow in adult and ageing skull. Specifically, we show that the skull bone marrow undergoes lifelong expansion involving vascular growth, which results in an increasing contribution to total haematopoietic output. Furthermore, skull is largely protected against major hallmarks of ageing, including upregulation of pro-inflammatory cytokines, adipogenesis and loss of vascular integrity. Conspicuous rapid and dynamic changes to the skull vasculature and bone marrow are induced by physiological alterations, namely pregnancy, but also pathological challenges, such as stroke and experimental chronic myeloid leukaemia. These responses are highly distinct from femur, the most extensively studied bone marrow compartment. We propose that skull harbours a protected and dynamically expanding bone marrow microenvironment, which is relevant for experimental studies and, potentially, for clinical treatments in humans., Competing Interests: Competing interests: The authors declare no competing interests., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
24. Toward target 2035: EUbOPEN - a public-private partnership to enable & unlock biology in the open.
- Author
-
Tredup C, Ackloo S, Beck H, Brown PJ, Bullock AN, Ciulli A, Dikic I, Edfeldt K, Edwards AM, Elkins JM, Farin HF, Fon EA, Gstaiger M, Günther J, Gustavsson AL, Häberle S, Isigkeit L, Huber KVM, Kotschy A, Krämer O, Leach AR, Marsden BD, Matsui H, Merk D, Montel F, Mulder MPC, Müller S, Owen DR, Proschak E, Röhm S, Stolz A, Sundström M, von Delft F, Willson TM, Arrowsmith CH, and Knapp S
- Abstract
Target 2035 is a global initiative that seeks to identify a pharmacological modulator of most human proteins by the year 2035. As part of an ongoing series of annual updates of this initiative, we summarise here the efforts of the EUbOPEN project whose objectives and results are making a strong contribution to the goals of Target 2035. EUbOPEN is a public-private partnership with four pillars of activity: (1) chemogenomic library collections, (2) chemical probe discovery and technology development for hit-to-lead chemistry, (3) profiling of bioactive compounds in patient-derived disease assays, and (4) collection, storage and dissemination of project-wide data and reagents. The substantial outputs of this programme include a chemogenomic compound library covering one third of the druggable proteome, as well as 100 chemical probes, both profiled in patient derived assays, as well as hundreds of data sets deposited in existing public data repositories and a project-specific data resource for exploring EUbOPEN outputs., Competing Interests: There are no conflicts to declare., (This journal is © The Royal Society of Chemistry.)
- Published
- 2024
- Full Text
- View/download PDF
25. Establishment of Patient-Derived Organoids from Colorectal Cancer Resection Samples.
- Author
-
Seven D, Baron S, and Farin HF
- Abstract
Colorectal cancer (CRC) organoids can serve as powerful preclinical cell models that accurately reflect individual tumor characteristics. Establishing a patient-derived CRC biobank facilitates a wide range of applications, including basic oncology research, new drug discovery, drug testing, and personalized medicine. This chapter details the process of generation of organoids from surgical samples of primary and metastatic CRC as well as from tumor adjacent normal colon tissues. Furthermore, best practices for cultivation and cryostorage of CRC organoids are described., (© 2024. Springer Science+Business Media, LLC.)
- Published
- 2024
- Full Text
- View/download PDF
26. Disturbance in cerebral blood microcirculation and hypoxic-ischemic microenvironment are associated with the development of brain metastasis.
- Author
-
Roesler J, Spitzer D, Jia X, Aasen SN, Sommer K, Roller B, Olshausen N, Hebach NR, Albinger N, Ullrich E, Zhu L, Wang F, Macas J, Forster MT, Steinbach JP, Sevenich L, Devraj K, Thorsen F, Karreman MA, Plate KH, Reiss Y, and Harter PN
- Subjects
- Animals, Mice, Humans, Cerebrovascular Circulation physiology, Male, Female, Blood-Brain Barrier pathology, Blood-Brain Barrier metabolism, Mice, Inbred C57BL, Brain Neoplasms secondary, Brain Neoplasms pathology, Tumor Microenvironment, Microcirculation, Angiopoietin-2 metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Background: Brain metastases (BM) constitute an increasing challenge in oncology due to their impact on neurological function, limited treatment options, and poor prognosis. BM occurs through extravasation of circulating tumor cells across the blood-brain barrier. However, the extravasation processes are still poorly understood. We here propose a brain colonization process which mimics infarction-like microenvironmental reactions, that are dependent on Angiopoietin-2 (Ang-2) and vascular endothelial growth factor (VEGF)., Methods: In this study, intracardiac BM models were used, and cerebral blood microcirculation was monitored by 2-photon microscopy through a cranial window. BM formation was observed using cranial magnetic resonance, bioluminescent imaging, and postmortem autopsy. Ang-2/VEGF targeting strategies and Ang-2 gain-of-function (GOF) mice were employed to interfere with BM formation. In addition, vascular and stromal factors as well as clinical outcomes were analyzed in BM patients., Results: Blood vessel occlusions by cancer cells were detected, accompanied by significant disturbances of cerebral blood microcirculation, and focal stroke-like histological signs. Cerebral endothelial cells showed an elevated Ang-2 expression both in mouse and human BM. Ang-2 GOF resulted in an increased BM burden. Combined anti-Ang-2/anti-VEGF therapy led to a decrease in brain metastasis size and number. Ang-2 expression in tumor vessels of established human BM negatively correlated with survival., Conclusions: Our observations revealed a relationship between disturbance of cerebral blood microcirculation and brain metastasis formation. This suggests that vessel occlusion by tumor cells facilitates brain metastatic extravasation and seeding, while combined inhibition of microenvironmental effects of Ang-2 and VEGF prevents the outgrowth of macrometastases., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2024
- Full Text
- View/download PDF
27. Gut microbial carcinogen metabolism: another avenue to cancer.
- Author
-
Greten FR and Arkan MC
- Published
- 2024
- Full Text
- View/download PDF
28. Differential inflammatory conditioning of the bone marrow by acute myeloid leukemia and its impact on progression.
- Author
-
Minciacchi VR, Karantanou C, Bravo J, Pereira RS, Zanetti C, Krack T, Kumar R, Bankov K, Hartmann S, Huntly BJP, Meduri E, Ruf W, and Krause DS
- Subjects
- Humans, Animals, Mice, Tumor Microenvironment, Tumor Necrosis Factor-alpha metabolism, Disease Models, Animal, Signal Transduction, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Bone Marrow pathology, Bone Marrow metabolism, Inflammation metabolism, Inflammation pathology, Dinoprostone metabolism, Disease Progression
- Abstract
Abstract: Inflammation promotes solid tumor progression, but how regulatory mechanisms of inflammation may affect leukemia is less well studied. Using annexin A5 (ANXA5), a calcium-binding protein known for apoptosis, which we discovered to be differentially expressed in the bone marrow microenvironment (BMM) of mice with acute myeloid (AML) vs chronic myeloid leukemia, as a model system, we unravel here a circuit in which AML-derived tumor necrosis factor α (TNF-α) dose-dependently reduces ANXA5 in the BMM. This creates an inflammatory BMM via elevated levels of prostaglandin E2 (PGE2). Via binding to its EP4 receptor, PGE2 increases β-catenin and hypoxia-inducible factor 1α signaling in AML cells, thereby accelerating PGE2-sensitive AML. Human trephine biopsies may show lower ANXA5 expression and higher PGE2 expression in AML than other hematologic malignancies. Furthermore, syngeneic and xenogeneic transplantation models suggest a survival benefit after treatment with the inhibitor of prostaglandin-endoperoxide synthase 2 (cyclooxygenase 2 [COX2]), celecoxib, plus cytarabine in those AML types highly sensitive to PGE2 compared with cytarabine alone. Taken together, TNF-α/ANXA5/NF-κB/COX2/PGE2-mediated inflammation influences AML course in a highly differential and circular manner, and patients with AML with "inflammatory AML" may benefit from antiphlogistic agents as adjunct therapy., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
29. T-cell specific in vivo gene delivery with DART-AAVs targeted to CD8.
- Author
-
Demircan MB, Zinser LJ, Michels A, Guaza-Lasheras M, John F, Gorol JM, Theuerkauf SA, Günther DM, Grimm D, Greten FR, Chlanda P, Thalheimer FB, and Buchholz CJ
- Subjects
- Animals, Mice, Humans, Transduction, Genetic, Capsid Proteins genetics, Dependovirus genetics, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Genetic Vectors administration & dosage, Genetic Vectors genetics, Gene Transfer Techniques, Genetic Therapy methods
- Abstract
One of the biggest challenges for in vivo gene therapy are vectors mediating highly selective gene transfer into a defined population of therapy-relevant cells. Here we present DARPin-targeted AAVs (DART-AAVs) displaying DARPins specific for human and murine CD8. Insertion of DARPins into the GH2/GH3 loop of the capsid protein 1 (VP1) of AAV2 and AAV6 resulted in high selectivity for CD8-positive T cells with unimpaired gene delivery activity. Remarkably, the capsid core structure was unaltered with protruding DARPins detectable. In complex primary cell mixtures, including donor blood or systemic injections into mice, the CD8-targeted AAVs were by far superior to unmodified AAV2 and AAV6 in terms of selectivity, target cell viability, and gene transfer rates. In vivo, up to 80% of activated CD8+ T cells were hit upon a single vector injection into conditioned humanized or immunocompetent mice. While gene transfer rates decreased significantly under non-activated conditions, genomic modification selectively in CD8+ T cells was still detectable upon Cre delivery into indicator mice. In both mouse models, selectivity for CD8+ T cells was close to absolute with exceptional detargeting from liver. The CD8-AAVs described here expand strategies for immunological research and in vivo gene therapy options., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
30. A guideline on the molecular ecosystem regulating ferroptosis.
- Author
-
Dai E, Chen X, Linkermann A, Jiang X, Kang R, Kagan VE, Bayir H, Yang WS, Garcia-Saez AJ, Ioannou MS, Janowitz T, Ran Q, Gu W, Gan B, Krysko DV, Zhu X, Wang J, Krautwald S, Toyokuni S, Xie Y, Greten FR, Yi Q, Schick J, Liu J, Gabrilovich DI, Liu J, Zeh HJ, Zhang DD, Yang M, Iovanna J, Kopf M, Adolph TE, Chi JT, Li C, Ichijo H, Karin M, Sankaran VG, Zou W, Galluzzi L, Bush AI, Li B, Melino G, Baehrecke EH, Lotze MT, Klionsky DJ, Stockwell BR, Kroemer G, and Tang D
- Subjects
- Humans, Animals, Signal Transduction, Reactive Oxygen Species metabolism, Iron metabolism, Ferroptosis genetics, Lipid Peroxidation
- Abstract
Ferroptosis, an intricately regulated form of cell death characterized by uncontrolled lipid peroxidation, has garnered substantial interest since this term was first coined in 2012. Recent years have witnessed remarkable progress in elucidating the detailed molecular mechanisms that govern ferroptosis induction and defence, with particular emphasis on the roles of heterogeneity and plasticity. In this Review, we discuss the molecular ecosystem of ferroptosis, with implications that may inform and enable safe and effective therapeutic strategies across a broad spectrum of diseases., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
31. Bispecific killer cell engagers employing species cross-reactive NKG2D binders redirect human and murine lymphocytes to ErbB2/HER2-positive malignancies.
- Author
-
Pfeifer Serrahima J, Schoenfeld K, Kühnel I, Harwardt J, Macarrón Palacios A, Prüfer M, Kolaric M, Oberoi P, Kolmar H, and Wels WS
- Subjects
- Animals, Humans, Mice, Single-Chain Antibodies immunology, Single-Chain Antibodies genetics, Cell Line, Tumor, Neoplasms immunology, Neoplasms therapy, Immunotherapy methods, Receptor, ErbB-2 immunology, NK Cell Lectin-Like Receptor Subfamily K immunology, NK Cell Lectin-Like Receptor Subfamily K metabolism, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Cross Reactions immunology, Antibodies, Bispecific immunology, Antibodies, Bispecific pharmacology
- Abstract
NKG2D is an activating receptor expressed by natural killer (NK) cells and other cytotoxic lymphocytes that plays a pivotal role in the elimination of neoplastic cells through recognition of different stress-induced cell surface ligands (NKG2DL). To employ this mechanism for cancer immunotherapy, we generated NKG2D-engaging bispecific antibodies that selectively redirect immune effector cells to cancer cells expressing the tumor-associated antigen ErbB2 (HER2). NKG2D-specific single chain fragment variable (scFv) antibodies cross-reactive toward the human and murine receptors were derived by consecutive immunization of chicken with the human and murine antigens, followed by stringent screening of a yeast surface display immune library. Four distinct species cross-reactive (sc) scFv domains were selected, and reformatted into a bispecific engager format by linking them via an IgG4 Fc domain to a second scFv fragment specific for ErbB2. The resulting molecules (termed scNKAB-ErbB2) were expressed as disulfide-linked homodimers, and demonstrated efficient binding to ErbB2-positive cancer cells as well as NKG2D-expressing primary human and murine lymphocytes, and NK-92 cells engineered with chimeric antigen receptors derived from human and murine NKG2D (termed hNKAR and mNKAR). Two of the scNKAB-ErbB2 molecules were found to compete with the natural NKG2D ligand MICA, while the other two engagers interacted with an epitope outside of the ligand binding site. Nevertheless, all four tested scNKAB-ErbB2 antibodies were similarly effective in redirecting the cytotoxic activity of primary human and murine lymphocytes as well as hNKAR-NK-92 and mNKAR-NK-92 cells to ErbB2-expressing targets, suggesting that further development of these species cross-reactive engager molecules for cancer immunotherapy is warranted., Competing Interests: KS, JH, AM, PO, HK and WW are named as inventors on patents and patent applications in the field of cancer immunotherapy owned by their respective academic institutions. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Pfeifer Serrahima, Schoenfeld, Kühnel, Harwardt, Macarrón Palacios, Prüfer, Kolaric, Oberoi, Kolmar and Wels.)
- Published
- 2024
- Full Text
- View/download PDF
32. Fetal growth restriction induced by maternal gal-3 deficiency is associated with altered gut-placenta axis.
- Author
-
Xie Y, Zhao F, Wang Y, Borowski S, Freitag N, Tirado-Gonzalez I, Hofsink N, Matschl U, Plösch T, Garcia MG, and Blois SM
- Subjects
- Pregnancy, Female, Animals, Mice, Male, Gastrointestinal Microbiome, Mice, Inbred C57BL, Humans, Fetal Development, Insulin-Like Growth Factor II metabolism, Insulin-Like Growth Factor II genetics, Insulin-Like Growth Factor II deficiency, Trophoblasts metabolism, Fetal Growth Retardation metabolism, Fetal Growth Retardation genetics, Placenta metabolism, Galectin 3 metabolism, Galectin 3 deficiency, Galectin 3 genetics
- Abstract
Adverse intrauterine conditions may cause fetal growth restriction (FGR), a pregnancy complication frequently linked to perinatal morbidity and mortality. Although many studies have focused on FGR, the pathophysiological processes underlying this disorder are complex and incompletely understood. We have recently determined that galectin-3 (gal-3), a β-galactoside-binding protein, regulates pregnancy-associated processes, including uterine receptibility, maternal vascular adaptation and placentation. Because gal-3 is expressed at both sides of the maternal-fetal interface, we unraveled the contribution of maternal- and paternal-derived gal-3 on fetal-placental development in the prenatal window and its effects on the post-natal period. Deficiency of maternal gal-3 induced maternal gut microbiome dysbiosis, resulting in a sex-specific fetal growth restriction mainly observed in female fetuses and offspring. In addition, poor placental metabolic adaptions (characterized by decreased trophoblast glycogen content and insulin-like growth factor 2 (Igf2) gene hypomethylation) were only associated with a lack of maternal-derived gal-3. Paternal gal-3 deficiency caused compromised vascularization in the placental labyrinth without affecting fetal growth trajectory. Thus, maternal-derived gal-3 may play a key role in fetal-placental development through the gut-placenta axis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
33. ZEB1-mediated fibroblast polarization controls inflammation and sensitivity to immunotherapy in colorectal cancer.
- Author
-
Menche C, Schuhwerk H, Armstark I, Gupta P, Fuchs K, van Roey R, Mosa MH, Hartebrodt A, Hajjaj Y, Clavel Ezquerra A, Selvaraju MK, Geppert CI, Bärthel S, Saur D, Greten FR, Brabletz S, Blumenthal DB, Weigert A, Brabletz T, Farin HF, and Stemmler MP
- Subjects
- Animals, Mice, Humans, Gene Expression Regulation, Neoplastic, Fibroblasts metabolism, Cell Line, Tumor, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Epithelial-Mesenchymal Transition genetics, Zinc Finger E-box-Binding Homeobox 1 metabolism, Zinc Finger E-box-Binding Homeobox 1 genetics, Colorectal Neoplasms pathology, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Colorectal Neoplasms therapy, Colorectal Neoplasms immunology, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Inflammation metabolism, Inflammation genetics, Inflammation pathology, Immunotherapy methods
- Abstract
The EMT-transcription factor ZEB1 is heterogeneously expressed in tumor cells and in cancer-associated fibroblasts (CAFs) in colorectal cancer (CRC). While ZEB1 in tumor cells regulates metastasis and therapy resistance, its role in CAFs is largely unknown. Combining fibroblast-specific Zeb1 deletion with immunocompetent mouse models of CRC, we observe that inflammation-driven tumorigenesis is accelerated, whereas invasion and metastasis in sporadic cancers are reduced. Single-cell transcriptomics, histological characterization, and in vitro modeling reveal a crucial role of ZEB1 in CAF polarization, promoting myofibroblastic features by restricting inflammatory activation. Zeb1 deficiency impairs collagen deposition and CAF barrier function but increases NFκB-mediated cytokine production, jointly promoting lymphocyte recruitment and immune checkpoint activation. Strikingly, the Zeb1-deficient CAF repertoire sensitizes to immune checkpoint inhibition, offering a therapeutic opportunity of targeting ZEB1 in CAFs and its usage as a prognostic biomarker. Collectively, we demonstrate that ZEB1-dependent plasticity of CAFs suppresses anti-tumor immunity and promotes metastasis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
34. Engineering of potent CAR NK cells using non-viral Sleeping Beauty transposition from minimalistic DNA vectors.
- Author
-
Bexte T, Botezatu L, Miskey C, Gierschek F, Moter A, Wendel P, Reindl LM, Campe J, Villena-Ossa JF, Gebel V, Stein K, Cathomen T, Cremer A, Wels WS, Hudecek M, Ivics Z, and Ullrich E
- Subjects
- Humans, Animals, Mice, Xenograft Model Antitumor Assays, Transposases genetics, Transposases metabolism, Cell Line, Tumor, DNA Transposable Elements, Cytotoxicity, Immunologic, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Cell Engineering methods, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Genetic Vectors genetics, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism, Immunotherapy, Adoptive methods
- Abstract
Natural killer (NK) cells have high intrinsic cytotoxic capacity, and clinical trials have demonstrated their safety and efficacy for adoptive cancer therapy. Expression of chimeric antigen receptors (CARs) enhances NK cell target specificity, with these cells applicable as off-the-shelf products generated from allogeneic donors. Here, we present for the first time an innovative approach for CAR NK cell engineering employing a non-viral Sleeping Beauty (SB) transposon/transposase-based system and minimized DNA vectors termed minicircles. SB-modified peripheral blood-derived primary NK cells displayed high and stable CAR expression and more frequent vector integration into genomic safe harbors than lentiviral vectors. Importantly, SB-generated CAR NK cells demonstrated enhanced cytotoxicity compared with non-transfected NK cells. A strong antileukemic potential was confirmed using established acute lymphocytic leukemia cells and patient-derived primary acute B cell leukemia and lymphoma samples as targets in vitro and in vivo in a xenograft leukemia mouse model. Our data suggest that the SB-transposon system is an efficient, safe, and cost-effective approach to non-viral engineering of highly functional CAR NK cells, which may be suitable for cancer immunotherapy of leukemia as well as many other malignancies., Competing Interests: Declaration of interests E.U. is an Advisory Board member for Phialogics and has sponsored research projects with Gilead and BMS. Z.I. is an inventor on patents related to Sleeping Beauty and MC technology. M.H. is listed as inventor on patent applications and granted patents related to CAR technologies and transposon-based gene transfer that are, in part licensed to industry. M.H. is a co-founder and equity owner of T-CURX GmbH, Würzburg, Germany. M.H. and E.U. are inventors on patents related to CAR and MC technology. T.B., P.W., W.S.W., and E.U. are inventors on patents related to optimized CAR designs. T.B., P.W., and E.U. are inventors on non-viral gene-editing technologies of NK cells., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
35. Using the tumour microenvironment to improve therapy efficacy.
- Author
-
Tatarova Z
- Subjects
- Humans, Animals, Treatment Outcome, Tumor Microenvironment, Neoplasms pathology, Neoplasms therapy, Neoplasms drug therapy
- Published
- 2024
- Full Text
- View/download PDF
36. Engineering an inducible leukemia-associated fusion protein enables large-scale ex vivo production of functional human phagocytes.
- Author
-
Windisch R, Soliman S, Hoffmann A, Chen-Wichmann L, Danese A, Vosberg S, Bravo J, Lutz S, Kellner C, Fischer A, Gebhard C, Redondo Monte E, Hartmann L, Schneider S, Beier F, Strobl CD, Weigert O, Peipp M, Schündeln M, Stricker SH, Rehli M, Bernhagen J, Humpe A, Klump H, Brendel C, Krause DS, Greif PA, and Wichmann C
- Subjects
- Humans, Hematopoietic Stem Cells metabolism, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, Recombinant Fusion Proteins metabolism, Recombinant Fusion Proteins genetics, Myeloid-Lymphoid Leukemia Protein metabolism, Myeloid-Lymphoid Leukemia Protein genetics, Leukemia genetics, Leukemia pathology, Leukemia metabolism, Protein Engineering methods, Phagocytosis, Phagocytes metabolism, Cell Differentiation
- Abstract
Ex vivo expansion of human CD34+ hematopoietic stem and progenitor cells remains a challenge due to rapid differentiation after detachment from the bone marrow niche. In this study, we assessed the capacity of an inducible fusion protein to enable sustained ex vivo proliferation of hematopoietic precursors and their capacity to differentiate into functional phagocytes. We fused the coding sequences of an FK506-Binding Protein 12 (FKBP12)-derived destabilization domain (DD) to the myeloid/lymphoid lineage leukemia/eleven nineteen leukemia (MLL-ENL) fusion gene to generate the fusion protein DD-MLL-ENL and retrovirally expressed the protein switch in human CD34+ progenitors. Using Shield1, a chemical inhibitor of DD fusion protein degradation, we established large-scale and long-term expansion of late monocytic precursors. Upon Shield1 removal, the cells lost self-renewal capacity and spontaneously differentiated, even after 2.5 y of continuous ex vivo expansion. In the absence of Shield1, stimulation with IFN-γ, LPS, and GM-CSF triggered terminal differentiation. Gene expression analysis of the obtained phagocytes revealed marked similarity with naïve monocytes. In functional assays, the novel phagocytes migrated toward CCL2, attached to VCAM-1 under shear stress, produced reactive oxygen species, and engulfed bacterial particles, cellular particles, and apoptotic cells. Finally, we demonstrated Fcγ receptor recognition and phagocytosis of opsonized lymphoma cells in an antibody-dependent manner. Overall, we have established an engineered protein that, as a single factor, is useful for large-scale ex vivo production of human phagocytes. Such adjustable proteins have the potential to be applied as molecular tools to produce functional immune cells for experimental cell-based approaches., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
37. Future direction of total neoadjuvant therapy for locally advanced rectal cancer.
- Author
-
Kagawa Y, Smith JJ, Fokas E, Watanabe J, Cercek A, Greten FR, Bando H, Shi Q, Garcia-Aguilar J, Romesser PB, Horvat N, Sanoff H, Hall W, Kato T, Rödel C, Dasari A, and Yoshino T
- Subjects
- Humans, Chemoradiotherapy methods, Neoadjuvant Therapy, Rectal Neoplasms therapy, Rectal Neoplasms pathology
- Abstract
Despite therapeutic advancements, disease-free survival and overall survival of patients with locally advanced rectal cancer have not improved in most trials as a result of distant metastases. For treatment decision-making, both long-term oncologic outcomes and impact on quality-of-life indices should be considered (for example, bowel function). Total neoadjuvant therapy (TNT), comprised of chemotherapy and radiotherapy or chemoradiotherapy, is now a standard treatment approach in patients with features of high-risk disease to prevent local recurrence and distant metastases. In selected patients who have a clinical complete response, subsequent surgery might be avoided through non-operative management, but patients who do not respond to TNT have a poor prognosis. Refined molecular characterization might help to predict which patients would benefit from TNT and non-operative management. Specifically, integrated analysis of spatiotemporal multi-omics using artificial intelligence and machine learning is promising. Three prospective trials of TNT and non-operative management in Japan, the USA and Germany are collaborating to better understand drivers of response to TNT. Here, we address the future direction for TNT., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
38. Autologous HER2-specific CAR T cells after lymphodepletion for advanced sarcoma: a phase 1 trial.
- Author
-
Hegde M, Navai S, DeRenzo C, Joseph SK, Sanber K, Wu M, Gad AZ, Janeway KA, Campbell M, Mullikin D, Nawas Z, Robertson C, Mathew PR, Zhang H, Mehta B, Bhat RR, Major A, Shree A, Gerken C, Kalra M, Chakraborty R, Thakkar SG, Dakhova O, Salsman VS, Grilley B, Lapteva N, Gee A, Dotti G, Bao R, Salem AH, Wang T, Brenner MK, Heslop HE, Wels WS, Hicks MJ, Gottschalk S, and Ahmed N
- Subjects
- Humans, Middle Aged, Female, Male, Adult, Aged, T-Lymphocytes immunology, Lymphocyte Depletion methods, Prospective Studies, Vidarabine analogs & derivatives, Vidarabine administration & dosage, Vidarabine therapeutic use, Cyclophosphamide therapeutic use, Cyclophosphamide administration & dosage, Treatment Outcome, Sarcoma therapy, Sarcoma immunology, Receptor, ErbB-2, Immunotherapy, Adoptive methods, Immunotherapy, Adoptive adverse effects, Receptors, Chimeric Antigen immunology
- Abstract
In this prospective, interventional phase 1 study for individuals with advanced sarcoma, we infused autologous HER2-specific chimeric antigen receptor T cells (HER2 CAR T cells) after lymphodepletion with fludarabine (Flu) ± cyclophosphamide (Cy): 1 × 10
8 T cells per m2 after Flu (cohort A) or Flu/Cy (cohort B) and 1 × 108 CAR+ T cells per m2 after Flu/Cy (cohort C). The primary outcome was assessment of safety of one dose of HER2 CAR T cells after lymphodepletion. Determination of antitumor responses was the secondary outcome. Thirteen individuals were treated in 14 enrollments, and seven received multiple infusions. HER2 CAR T cells expanded after 19 of 21 infusions. Nine of 12 individuals in cohorts A and B developed grade 1-2 cytokine release syndrome. Two individuals in cohort C experienced dose-limiting toxicity with grade 3-4 cytokine release syndrome. Antitumor activity was observed with clinical benefit in 50% of individuals treated. The tumor samples analyzed showed spatial heterogeneity of immune cells and clustering by sarcoma type and by treatment response. Our results affirm HER2 as a CAR T cell target and demonstrate the safety of this therapeutic approach in sarcoma. ClinicalTrials.gov registration: NCT00902044 ., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
39. Biophysical Investigation of RNA ⋅ DNA : DNA Triple Helix and RNA : DNA Heteroduplex Formation by the lncRNAs MEG3 and Fendrr.
- Author
-
Krause NM, Bains JK, Blechar J, Richter C, Bessi I, Grote P, Leisegang MS, Brandes RP, and Schwalbe H
- Subjects
- Humans, Nucleic Acid Heteroduplexes chemistry, RNA chemistry, RNA genetics, RNA metabolism, Thermodynamics, RNA, Long Noncoding genetics, RNA, Long Noncoding chemistry, RNA, Long Noncoding metabolism, DNA chemistry, DNA genetics, Nucleic Acid Conformation
- Abstract
Long non-coding RNAs (lncRNAs) are important regulators of gene expression and can associate with DNA as RNA : DNA heteroduplexes or RNA ⋅ DNA : DNA triple helix structures. Here, we review in vitro biochemical and biophysical experiments including electromobility shift assays (EMSA), circular dichroism (CD) spectroscopy, thermal melting analysis, microscale thermophoresis (MST), single-molecule Förster resonance energy transfer (smFRET) and nuclear magnetic resonance (NMR) spectroscopy to investigate RNA ⋅ DNA : DNA triple helix and RNA : DNA heteroduplex formation. We present the investigations of the antiparallel triplex-forming lncRNA MEG3 targeting the gene TGFB2 and the parallel triplex-forming lncRNA Fendrr with its target gene Emp2. The thermodynamic properties of these oligonucleotides lead to concentration-dependent heterogeneous mixtures, where a DNA duplex, an RNA : DNA heteroduplex and an RNA ⋅ DNA : DNA triplex coexist and their relative populations are modulated in a temperature-dependent manner. The in vitro data provide a reliable readout of triplex structures, as RNA ⋅ DNA : DNA triplexes show distinct features compared to DNA duplexes and RNA : DNA heteroduplexes. Our experimental results can be used to validate computationally predicted triple helix formation between novel disease-relevant lncRNAs and their DNA target genes., (© 2024 The Authors. ChemBioChem published by Wiley-VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
40. Octyl itaconate enhances VSVΔ51 oncolytic virotherapy by multitarget inhibition of antiviral and inflammatory pathways.
- Author
-
Kurmasheva N, Said A, Wong B, Kinderman P, Han X, Rahimic AHF, Kress A, Carter-Timofte ME, Holm E, van der Horst D, Kollmann CF, Liu Z, Wang C, Hoang HD, Kovalenko E, Chrysopoulou M, Twayana KS, Ottosen RN, Svenningsen EB, Begnini F, Kiib AE, Kromm FEH, Weiss HJ, Di Carlo D, Muscolini M, Higgins M, van der Heijden M, Arulanandam R, Bardoul A, Tong T, Ozsvar A, Hou WH, Schack VR, Holm CK, Zheng Y, Ruzek M, Kalucka J, de la Vega L, Elgaher WAM, Korshoej AR, Lin R, Hiscott J, Poulsen TB, O'Neill LA, Roy DG, Rinschen MM, van Montfoort N, Diallo JS, Farin HF, Alain T, and Olagnier D
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Interferon Type I metabolism, NF-E2-Related Factor 2 metabolism, Colonic Neoplasms therapy, Colonic Neoplasms immunology, Colonic Neoplasms drug therapy, Antiviral Agents pharmacology, NF-kappa B metabolism, I-kappa B Kinase metabolism, Kelch-Like ECH-Associated Protein 1 metabolism, Inflammation drug therapy, Female, Vesicular stomatitis Indiana virus physiology, Vesicular stomatitis Indiana virus drug effects, Signal Transduction drug effects, Oncolytic Virotherapy methods, Succinates pharmacology, Oncolytic Viruses
- Abstract
The presence of heterogeneity in responses to oncolytic virotherapy poses a barrier to clinical effectiveness, as resistance to this treatment can occur through the inhibition of viral spread within the tumor, potentially leading to treatment failures. Here we show that 4-octyl itaconate (4-OI), a chemical derivative of the Krebs cycle-derived metabolite itaconate, enhances oncolytic virotherapy with VSVΔ51 in various models including human and murine resistant cancer cell lines, three-dimensional (3D) patient-derived colon tumoroids and organotypic brain tumor slices. Furthermore, 4-OI in combination with VSVΔ51 improves therapeutic outcomes in a resistant murine colon tumor model. Mechanistically, we find that 4-OI suppresses antiviral immunity in cancer cells through the modification of cysteine residues in MAVS and IKKβ independently of the NRF2/KEAP1 axis. We propose that the combination of a metabolite-derived drug with an oncolytic virus agent can greatly improve anticancer therapeutic outcomes by direct interference with the type I IFN and NF-κB-mediated antiviral responses., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
41. Clonal hematopoiesis and its impact on the aging osteo-hematopoietic niche.
- Author
-
Winter S, Götze KS, Hecker JS, Metzeler KH, Guezguez B, Woods K, Medyouf H, Schäffer A, Schmitz M, Wehner R, Glauche I, Roeder I, Rauner M, Hofbauer LC, and Platzbecker U
- Subjects
- Humans, Bone Marrow metabolism, Bone Marrow pathology, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells cytology, Mutation, Hematologic Neoplasms genetics, Hematologic Neoplasms pathology, Animals, Hematopoiesis genetics, Clonal Hematopoiesis genetics, Stem Cell Niche, Aging genetics, Aging physiology
- Abstract
Clonal hematopoiesis (CH) defines a premalignant state predominantly found in older persons that increases the risk of developing hematologic malignancies and age-related inflammatory diseases. However, the risk for malignant transformation or non-malignant disorders is variable and difficult to predict, and defining the clinical relevance of specific candidate driver mutations in individual carriers has proved to be challenging. In addition to the cell-intrinsic mechanisms, mutant cells rely on and alter cell-extrinsic factors from the bone marrow (BM) niche, which complicates the prediction of a mutant cell's fate in a shifting pre-malignant microenvironment. Therefore, identifying the insidious and potentially broad impact of driver mutations on supportive niches and immune function in CH aims to understand the subtle differences that enable driver mutations to yield different clinical outcomes. Here, we review the changes in the aging BM niche and the emerging evidence supporting the concept that CH can progressively alter components of the local BM microenvironment. These alterations may have profound implications for the functionality of the osteo-hematopoietic niche and overall bone health, consequently fostering a conducive environment for the continued development and progression of CH. We also provide an overview of the latest technology developments to study the spatiotemporal dependencies in the CH BM niche, ideally in the context of longitudinal studies following CH over time. Finally, we discuss aspects of CH carrier management in clinical practice, based on work from our group and others., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
42. Bortezomib promotes the TRAIL-mediated killing of resistant rhabdomyosarcoma by ErbB2/Her2-targeted CAR-NK-92 cells via DR5 upregulation.
- Author
-
Heim C, Hartig L, Weinelt N, Moser LM, Salzmann-Manrique E, Merker M, Wels WS, Tonn T, Bader P, Klusmann JH, van Wijk SJL, and Rettinger E
- Abstract
Treatment resistance and immune escape are hallmarks of metastatic rhabdomyosarcoma (RMS), underscoring the urgent medical need for therapeutic agents against this disease entity as a key challenge in pediatric oncology. Chimeric antigen receptor (CAR)-based immunotherapies, such as the ErbB2 (Her2)-CAR-engineered natural killer (NK) cell line NK-92/5.28.z, provide antitumor cytotoxicity primarily through CAR-mediated cytotoxic granule release and thereafter-even in cases with low surface antigen expression or tumor escape-by triggering intrinsic NK cell-mediated apoptosis induction via additional ligand/receptors. In this study, we showed that bortezomib increased susceptibility toward apoptosis in clinically relevant RMS cell lines RH30 and RH41, and patient-derived RMS tumor organoid RMS335, by upregulation of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor DR5 in these metastatic, relapsed/refractory (r/r) RMS tumors. Subsequent administration of NK-92/5.28.z cells significantly enhanced antitumor activity in vitro . Applying recombinant TRAIL instead of NK-92/5.28.z cells confirmed that the synergistic antitumor effects of the combination treatment were mediated via TRAIL. Western blot analyses indicated that the combination treatment with bortezomib and NK-92/5.28.z cells increased apoptosis by interacting with the nuclear factor κB, JNK, and caspase pathways. Overall, bortezomib pretreatment can sensitize r/r RMS tumors to CAR- and, by upregulating DR5, TRAIL-mediated cytotoxicity of NK-92/5.28.z cells., Competing Interests: J.-H.K. has advisory roles for Bluebird Bio, Novartis, Roche, and Jazz Pharmaceuticals. T.T. and W.S.W. are named as inventors on patents and patent applications related to the therapeutic agent used in this study, owned by their respective academic institutions., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
43. Cyclophilin A supports translation of intrinsically disordered proteins and affects haematopoietic stem cell ageing.
- Author
-
Maneix L, Iakova P, Lee CG, Moree SE, Lu X, Datar GK, Hill CT, Spooner E, King JCK, Sykes DB, Saez B, Di Stefano B, Chen X, Krause DS, Sahin E, Tsai FTF, Goodell MA, Berk BC, Scadden DT, and Catic A
- Subjects
- Cyclophilin A genetics, Cyclophilin A metabolism, RNA-Binding Proteins, Hematopoietic Stem Cells metabolism, Intrinsically Disordered Proteins chemistry
- Abstract
Loss of protein function is a driving force of ageing. We have identified peptidyl-prolyl isomerase A (PPIA or cyclophilin A) as a dominant chaperone in haematopoietic stem and progenitor cells. Depletion of PPIA accelerates stem cell ageing. We found that proteins with intrinsically disordered regions (IDRs) are frequent PPIA substrates. IDRs facilitate interactions with other proteins or nucleic acids and can trigger liquid-liquid phase separation. Over 20% of PPIA substrates are involved in the formation of supramolecular membrane-less organelles. PPIA affects regulators of stress granules (PABPC1), P-bodies (DDX6) and nucleoli (NPM1) to promote phase separation and increase cellular stress resistance. Haematopoietic stem cell ageing is associated with a post-transcriptional decrease in PPIA expression and reduced translation of IDR-rich proteins. Here we link the chaperone PPIA to the synthesis of intrinsically disordered proteins, which indicates that impaired protein interaction networks and macromolecular condensation may be potential determinants of haematopoietic stem cell ageing., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
44. The activating receptor NKp65 is selectively expressed by human ILC3 and demarcates ILC3 from mature NK cells.
- Author
-
Kühnel I, Vogler I, Spreu J, Bonig H, Döring C, and Steinle A
- Subjects
- Humans, Immunoassay, Killer Cells, Natural, Lectins, C-Type metabolism, Biosensing Techniques, Immunity, Innate
- Abstract
Innate lymphocytes comprise cytotoxic natural killer (NK) cells and tissue-resident innate lymphoid cells (ILC) that are subgrouped according to their cytokine profiles into group 1 ILC (ILC1), ILC2, and ILC3. However, cell surface receptors unambiguously defining or specifically activating such ILC subsets are scarcely known. Here, we report on the physiologic expression of the human activating C-type lectin-like receptor (CTLR) NKp65, a high-affinity receptor for the CTLR keratinocyte-associated C-type lectin (KACL). Tracking rare NKp65 transcripts in human blood, we identify ILC3 to selectively express NKp65. NKp65 expression not only demarcates "bona fide" ILC3 from likewise RORγt-expressing ILC precursors and lymphoid tissue inducer cells but also from mature NK cells which acquire the NKp65-relative NKp80 during a Notch-dependent differentiation from NKp65
+ precursor cells. Hence, ILC3 and NK cells mutually exclusively and interdependently express the genetically coupled sibling receptors NKp65 and NKp80. Much alike NKp80, NKp65 promotes cytotoxicity by innate lymphocytes which may become relevant during pathophysiological reprogramming of ILC3. Altogether, we report the selective expression of the activating immunoreceptor NKp65 by ILC3 demarcating ILC3 from mature NK cells and endowing ILC3 with a dedicated immunosensor for the epidermal immune barrier., (© 2024 The Authors. European Journal of Immunology published by Wiley‐VCH GmbH.)- Published
- 2024
- Full Text
- View/download PDF
45. Towards optimized tissue regeneration: a new 3D printable bioink of alginate/cellulose hydrogel loaded with thrombocyte concentrate.
- Author
-
Grandjean T, Perumal N, Manicam C, Matthey B, Wu T, Thiem DGE, Stein S, Henrich D, Kämmerer PW, Al-Nawas B, Ritz U, and Blatt S
- Abstract
Introduction: Autologous platelet concentrate (APC) are pro-angiogenic and can promote wound healing and tissue repair, also in combination with other biomaterials. However, challenging defect situations remain demanding. 3D bioprinting of an APC based bioink encapsulated in a hydrogel could overcome this limitation with enhanced physio-mechanical interface, growth factor retention/secretion and defect-personalized shape to ultimately enhance regeneration., Methods: This study used extrusion-based bioprinting to create a novel bioink of alginate/cellulose hydrogel loaded with thrombocyte concentrate. Chemico-physical testing exhibited an amorphous structure characterized by high shape fidelity. Cytotoxicity assay and incubation of human osteogenic sarcoma cells (SaOs2) exposed excellent biocompatibility. enzyme-linked immunosorbent assay analysis confirmed pro-angiogenic growth factor release of the printed constructs, and co-incubation with HUVECS displayed proper cell viability and proliferation. Chorioallantoic membrane (CAM) assay explored the pro-angiogenic potential of the prints in vivo . Detailed proteome and secretome analysis revealed a substantial amount and homologous presence of pro-angiogenic proteins in the 3D construct., Results: This study demonstrated a 3D bioprinting approach to fabricate a novel bioink of alginate/cellulose hydrogel loaded with thrombocyte concentrate with high shape fidelity, biocompatibility, and substantial pro-angiogenic properties., Conclusion: This approach may be suitable for challenging physiological and anatomical defect situations when translated into clinical use., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Grandjean, Perumal, Manicam, Matthey, Wu, Thiem, Stein, Henrich, Kämmerer, Al-Nawas, Ritz and Blatt.)
- Published
- 2024
- Full Text
- View/download PDF
46. Influenza A virus replicates productively in primary human kidney cells and induces factors and mechanisms related to regulated cell death and renal pathology observed in virus-infected patients.
- Author
-
Koch B, Shehata M, Müller-Ruttloff C, Gouda SA, Wetzstein N, Patyna S, Scholz A, Schmid T, Dietrich U, Münch C, Ziebuhr J, Geiger H, Martinez-Sobrido L, Baer PC, Mostafa A, and Pleschka S
- Subjects
- Humans, Proteome metabolism, Influenza A Virus, H3N2 Subtype physiology, Virus Replication physiology, Kidney pathology, Influenza A virus, Influenza A Virus, H1N1 Subtype, Influenza, Human, Regulated Cell Death, Acute Kidney Injury, Orthomyxoviridae Infections pathology
- Abstract
Introduction: Influenza A virus (IAV) infection can cause the often-lethal acute respiratory distress syndrome (ARDS) of the lung. Concomitantly, acute kidney injury (AKI) is frequently noticed during IAV infection, correlating with an increased mortality. The aim of this study was to elucidate the interaction of IAV with human kidney cells and, thereby, to assess the mechanisms underlying IAV-mediated AKI., Methods: To investigate IAV effects on nephron cells we performed infectivity assays with human IAV, as well as with human isolates of either low or highly pathogenic avian IAV. Also, transcriptome and proteome analysis of IAV-infected primary human distal tubular kidney cells (DTC) was performed. Furthermore, the DTC transcriptome was compared to existing transcriptomic data from IAV-infected lung and trachea cells., Results: We demonstrate productive replication of all tested IAV strains on primary and immortalized nephron cells. Comparison of our transcriptome and proteome analysis of H1N1-type IAV-infected human primary distal tubular cells (DTC) with existing data from H1N1-type IAV-infected lung and primary trachea cells revealed enrichment of specific factors responsible for regulated cell death in primary DTC, which could be targeted by specific inhibitors., Discussion: IAV not only infects, but also productively replicates on different human nephron cells. Importantly, multi-omics analysis revealed regulated cell death as potential contributing factor for the clinically observed kidney pathology in influenza., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Koch, Shehata, Müller-Ruttloff, Gouda, Wetzstein, Patyna, Scholz, Schmid, Dietrich, Münch, Ziebuhr, Geiger, Martinez-Sobrido, Baer, Mostafa and Pleschka.)
- Published
- 2024
- Full Text
- View/download PDF
47. Bi-allelic variants in CELSR3 are implicated in central nervous system and urinary tract anomalies.
- Author
-
Stegmann JD, Kalanithy JC, Dworschak GC, Ishorst N, Mingardo E, Lopes FM, Ho YM, Grote P, Lindenberg TT, Yilmaz Ö, Channab K, Seltzsam S, Shril S, Hildebrandt F, Boschann F, Heinen A, Jolly A, Myers K, McBride K, Bekheirnia MR, Bekheirnia N, Scala M, Morleo M, Nigro V, Torella A, Pinelli M, Capra V, Accogli A, Maitz S, Spano A, Olson RJ, Klee EW, Lanpher BC, Jang SS, Chae JH, Steinbauer P, Rieder D, Janecke AR, Vodopiutz J, Vogel I, Blechingberg J, Cohen JL, Riley K, Klee V, Walsh LE, Begemann M, Elbracht M, Eggermann T, Stoppe A, Stuurman K, van Slegtenhorst M, Barakat TS, Mulhern MS, Sands TT, Cytrynbaum C, Weksberg R, Isidori F, Pippucci T, Severi G, Montanari F, Kruer MC, Bakhtiari S, Darvish H, Reutter H, Hagelueken G, Geyer M, Woolf AS, Posey JE, Lupski JR, Odermatt B, and Hilger AC
- Abstract
CELSR3 codes for a planar cell polarity protein. We describe twelve affected individuals from eleven independent families with bi-allelic variants in CELSR3. Affected individuals presented with an overlapping phenotypic spectrum comprising central nervous system (CNS) anomalies (7/12), combined CNS anomalies and congenital anomalies of the kidneys and urinary tract (CAKUT) (3/12) and CAKUT only (2/12). Computational simulation of the 3D protein structure suggests the position of the identified variants to be implicated in penetrance and phenotype expression. CELSR3 immunolocalization in human embryonic urinary tract and transient suppression and rescue experiments of Celsr3 in fluorescent zebrafish reporter lines further support an embryonic role of CELSR3 in CNS and urinary tract formation., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
48. CAR-mediated targeting of NK cells overcomes tumor immune escape caused by ICAM-1 downregulation.
- Author
-
Eitler J, Rackwitz W, Wotschel N, Gudipati V, Murali Shankar N, Sidorenkova A, Huppa JB, Ortiz-Montero P, Opitz C, Künzel SR, Michen S, Temme A, Loureiro LR, Feldmann A, Bachmann M, Boissel L, Klingemann H, Wels WS, and Tonn T
- Subjects
- Humans, Female, Intercellular Adhesion Molecule-1, Lymphocyte Function-Associated Antigen-1 metabolism, Down-Regulation, Tumor Escape, Cell Line, Tumor, Killer Cells, Natural, Trastuzumab pharmacology, Antibodies, Receptors, Fc metabolism, Receptors, Chimeric Antigen metabolism, Breast Neoplasms drug therapy, Breast Neoplasms metabolism
- Abstract
Background: The antitumor activity of natural killer (NK) cells can be enhanced by specific targeting with therapeutic antibodies that trigger antibody-dependent cell-mediated cytotoxicity (ADCC) or by genetic engineering to express chimeric antigen receptors (CARs). Despite antibody or CAR targeting, some tumors remain resistant towards NK cell attack. While the importance of ICAM-1/LFA-1 interaction for natural cytotoxicity of NK cells is known, its impact on ADCC induced by the ErbB2 (HER2)-specific antibody trastuzumab and ErbB2-CAR-mediated NK cell cytotoxicity against breast cancer cells has not been investigated., Methods: Here we used NK-92 cells expressing high-affinity Fc receptor FcγRIIIa in combination with trastuzumab or ErbB2-CAR engineered NK-92 cells (NK-92/5.28.z) as well as primary human NK cells combined with trastuzumab or modified with the ErbB2-CAR and tested cytotoxicity against cancer cells varying in ICAM-1 expression or alternatively blocked LFA-1 on NK cells. Furthermore, we specifically stimulated Fc receptor, CAR and/or LFA-1 to study their crosstalk at the immunological synapse and their contribution to degranulation and intracellular signaling in antibody-targeted or CAR-targeted NK cells., Results: Blockade of LFA-1 or absence of ICAM-1 significantly reduced cell killing and cytokine release during trastuzumab-mediated ADCC against ErbB2-positive breast cancer cells, but not so in CAR-targeted NK cells. Pretreatment with 5-aza-2'-deoxycytidine induced ICAM-1 upregulation and reversed NK cell resistance in ADCC. Trastuzumab alone did not sufficiently activate NK cells and required additional LFA-1 co-stimulation, while activation of the ErbB2-CAR in CAR-NK cells induced efficient degranulation independent of LFA-1. Total internal reflection fluorescence single molecule imaging revealed that CAR-NK cells formed an irregular immunological synapse with tumor cells that excluded ICAM-1, while trastuzumab formed typical peripheral supramolecular activation cluster (pSMAC) structures. Mechanistically, the absence of ICAM-1 did not affect cell-cell adhesion during ADCC, but rather resulted in decreased signaling via Pyk2 and ERK1/2, which was intrinsically provided by CAR-mediated targeting. Furthermore, while stimulation of the inhibitory NK cell checkpoint molecule NKG2A markedly reduced FcγRIIIa/LFA-1-mediated degranulation, retargeting by CAR was only marginally affected., Conclusions: Downregulation of ICAM-1 on breast cancer cells is a critical escape mechanism from trastuzumab-triggered ADCC. In contrast, CAR-NK cells are able to overcome cancer cell resistance caused by ICAM-1 reduction, highlighting the potential of CAR-NK cells in cancer immunotherapy., Competing Interests: Competing interests: TT and WSW are named as inventors on patents in the field of cancer immunotherapy owned by their respective institutions. HK and LB are employed by ImmunityBio, California, USA. Other authors declare that they have no competing interests., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
49. [Tumor-host cell interaction in the microenvironment: new target points for treatment?]
- Author
-
Sevenich L and Heiland DH
- Subjects
- Humans, Prognosis, Central Nervous System, Cell Communication, Tumor Microenvironment, Brain Neoplasms therapy, Glioblastoma pathology, Glioblastoma therapy
- Abstract
Background: Primary brain tumors and metastases in the central nervous system (CNS) are characterized by their unique microenvironment, which interacts with neuronal structures and influences structural and adaptive immunity., Objective: How significant are various tumor-host interactions from a prognostic and therapeutic perspective?, Material and Method: A literature search was carried out for relevant articles on the topic: microenvironment glioblastoma or metastasis through PubMed and Medline., Results: Modern high-throughput methods, such as spatial and single-cell resolution molecular characterization of tumors and their microenvironment enable a detailed mapping of changes and adaptation of individual cells within the microenvironment of tumors; however, treatment approaches based on altered tumor-host cell interactions, such as immune modeling, cell-based treatment methods or checkpoint inhibition have so far not shown any significant advantages for survival., Conclusion: A deeper understanding of the complex immune landscape and the microenvironment of metastases of the CNS and intracerebral tumors is essential to optimize future treatment strategies., (© 2024. The Author(s), under exclusive licence to Springer Medizin Verlag GmbH, ein Teil von Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
50. Dual Targeting of Glioblastoma Cells with Bispecific Killer Cell Engagers Directed to EGFR and ErbB2 (HER2) Facilitates Effective Elimination by NKG2D-CAR-Engineered NK Cells.
- Author
-
Kiefer A, Prüfer M, Röder J, Pfeifer Serrahima J, Bodden M, Kühnel I, Oberoi P, and Wels WS
- Subjects
- Humans, NK Cell Lectin-Like Receptor Subfamily K metabolism, Interleukin-15 metabolism, Cell Line, Tumor, Killer Cells, Natural, ErbB Receptors metabolism, Glioblastoma metabolism, Antibodies, Bispecific pharmacology
- Abstract
NKG2D is an activating receptor of natural killer cells that recognizes stress-induced ligands (NKG2DL) expressed by many tumor cells. Nevertheless, NKG2DL downregulation or shedding can still allow cancer cells to evade immune surveillance. Here, we used lentiviral gene transfer to engineer clinically usable NK-92 cells with a chimeric antigen receptor (NKAR) which contains the extracellular domain of NKG2D for target recognition, or an NKAR, together with the IL-15 superagonist RD-IL15, and combined these effector cells with recombinant NKG2D-interacting bispecific engagers that simultaneously recognize the tumor-associated antigens epidermal growth factor receptor (EGFR) or ErbB2 (HER2). Applied individually, in in vitro cell-killing assays, these NKAB-EGFR and NKAB-ErbB2 antibodies specifically redirected NKAR-NK-92 and NKAR_RD-IL15-NK-92 cells to glioblastoma and other cancer cells with elevated EGFR or ErbB2 levels. However, in mixed glioblastoma cell cultures, used as a model for heterogeneous target antigen expression, NKAR-NK cells only lysed the EGFR- or ErbB2-expressing subpopulations in the presence of one of the NKAB molecules. This was circumvented by applying NKAB-EGFR and NKAB-ErbB2 together, resulting in effective antitumor activity similar to that against glioblastoma cells expressing both target antigens. Our results demonstrate that combining NK cells carrying an activating NKAR receptor with bispecific NKAB antibodies allows for flexible targeting, which can enhance tumor-antigen-specific cytotoxicity and prevent immune escape.
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.