112 results on '"Martina, M"'
Search Results
2. Effects of overfeeding on energy balance and brown fat thermogenesis in obese (ob/ob) mice
- Author
-
Pauline M. Jones, Paul Trayhurn, Anne E. Goodbody, and Martina M. McGuckin
- Subjects
medicine.medical_specialty ,Food intake ,Energy balance ,Mice, Obese ,Adipose tissue ,Cafeteria ,Mice ,Adipose Tissue, Brown ,Internal medicine ,Brown adipose tissue ,medicine ,Animals ,Obesity ,Multidisciplinary ,biology ,Body Weight ,biology.organism_classification ,medicine.disease ,Diet ,medicine.anatomical_structure ,Endocrinology ,Energy Intake ,Energy Metabolism ,Excess energy ,Thermogenesis ,Body Temperature Regulation - Abstract
The genetically obese (ob/ob) mouse has been used extensively in experimental studies on obesity and to determine the mechanisms involved in regulating energy balance1. Excess energy begins to accumulate in the ob/ob mouse from ∼12 days of age2, but food intake during the suckling period and up to 4 weeks of age is no greater than in lean siblings3–5. This indicates that the initial development of obesity in the ob/ob mutant is entirely due to a low energy expenditure, and this has been attributed to a reduction in thermoregulatory non-shivering thermogenesis6–8, particularly that mediated by brown adipose tissue9,10. Hyperphagia does, however, play an important part in the later development of obesity in ob/ob mice. Recent studies with young rats of the Sprague–Dawley strain fed a ‘cafeteria diet’ have suggested that an adaptive diet-induced thermogenesis, also mediated by brown adipose tissue, has a major role in minimizing the accumulation of energy during voluntary overfeeding11–13. We now report the effects of overfeeding young lean and obese mice with a cafeteria-type diet, and show that while the lean animals retain little of their excess energy intake, most of the extra intake of the obese mice is deposited. This suggests that the capacity for diet-induced thermogenesis is reduced in the obese compared with the lean mice. We also show that the energy balance measurements are paralleled by different adaptive changes in brown adipose tissue in the two genotypes.
- Published
- 1982
- Full Text
- View/download PDF
3. Brown adipose tissue thermogenesis is ‘suppressed’ during lactation in mice
- Author
-
Trayhurn, Paul, primary, Douglas, Jenny B., additional, and McGuckin, Martina M., additional
- Published
- 1982
- Full Text
- View/download PDF
4. Effects of overfeeding on energy balance and brown fat thermogenesis in obese (ob/ob) mice
- Author
-
Trayhurn, Paul, primary, Jones, Pauline M., additional, McGuckin, Martina M., additional, and Goodbody, Anne E., additional
- Published
- 1982
- Full Text
- View/download PDF
5. Brown adipose tissue thermogenesis is ‘suppressed’ during lactation in mice
- Author
-
Paul Trayhurn, Jenny B. Douglas, and Martina M. McGuckin
- Subjects
medicine.medical_specialty ,Multidisciplinary ,Adipose tissue ,Biology ,Guanosine Diphosphate ,Energy requirement ,Metabolic efficiency ,Mitochondria ,Electron Transport Complex IV ,Mice ,Endocrinology ,medicine.anatomical_structure ,Environmental temperature ,Adipose Tissue, Brown ,Pregnancy ,Internal medicine ,Lactation ,Brown adipose tissue ,medicine ,Animals ,Female ,Thermogenesis ,Body Temperature Regulation - Abstract
Brown adipose tissue is considered to be the main site of thermoregulatory non-shivering thermogenesis in the newborn of many mammalian species, in arousing hibernators, and in adult cold-adapted rats and mice1–4. Several recent studies have now suggested that the tissue may also be important in the regulation of energy balance. Evidence has been obtained indicating that the ability of genetically obese (ob/ob and db/db) mice to become obese on a normal energy intake is mainly due to reduced brown adipose tissue thermogenesis5–7, and a reduction in the activity of the tissue has also been observed in rats made obese by lesions of the ventromedial hypothalamus8. In contrast, increases in thermogenesis in brown adipose tissue have been implicated in the immediate thermic effect of food9 and in cancer cachexia10, and a substantial augmentation of the activity of the tissue has been demonstrated in rats and mice exhibiting regulatory diet-induced thermogenesis as a response to chronic voluntary over-feeding11–14. Whether or not brown adipose tissue thermogenesis changes in other, more physiological, states where energy flux and metabolic efficiency are altered (independently of environmental temperature) has not been established. We now report that the thermogenic activity of brown adipose tissue is suppressed in mice during the ‘physiological hyperphagia’ of lactation, and show that this probably results in a substantial reduction in the energy requirement for maintenance of the lactating animal.
- Published
- 1982
- Full Text
- View/download PDF
6. IL-33-activated ILC2s induce tertiary lymphoid structures in pancreatic cancer.
- Author
-
Amisaki M, Zebboudj A, Yano H, Zhang SL, Payne G, Chandra AK, Yu R, Guasp P, Sethna ZM, Ohmoto A, Rojas LA, Cheng C, Waters T, Solovyov A, Martis S, Doane AS, Reiche C, Bruno EM, Milighetti M, Soares K, Odgerel Z, Moral JA, Zhao JN, Gönen M, Gardner R, Tumanov AV, Khan AG, Vergnolle O, Nyakatura EK, Lorenz IC, Baca M, Patterson E, Greenbaum B, Artis D, Merghoub T, and Balachandran VP
- Abstract
Tertiary lymphoid structures (TLSs) are de novo ectopic lymphoid aggregates that regulate immunity in chronically inflamed tissues, including tumours. Although TLSs form due to inflammation-triggered activation of the lymphotoxin (LT)-LTβ receptor (LTβR) pathway
1 , the inflammatory signals and cells that induce TLSs remain incompletely identified. Here we show that interleukin-33 (IL-33), the alarmin released by inflamed tissues2 , induces TLSs. In mice, Il33 deficiency severely attenuates inflammation- and LTβR-activation-induced TLSs in models of colitis and pancreatic ductal adenocarcinoma (PDAC). In PDAC, the alarmin domain of IL-33 activates group 2 innate lymphoid cells (ILC2s) expressing LT that engage putative LTβR+ myeloid organizer cells to initiate tertiary lymphoneogenesis. Notably, lymphoneogenic ILC2s migrate to PDACs from the gut, can be mobilized to PDACs in different tissues and are modulated by gut microbiota. Furthermore, we detect putative lymphoneogenic ILC2s and IL-33-expressing cells within TLSs in human PDAC that correlate with improved prognosis. To harness this lymphoneogenic pathway for immunotherapy, we engineer a recombinant human IL-33 protein that expands intratumoural lymphoneogenic ILC2s and TLSs and demonstrates enhanced anti-tumour activity in PDAC mice. In summary, we identify the molecules and cells of a druggable pathway that induces inflammation-triggered TLSs. More broadly, we reveal a lymphoneogenic function for alarmins and ILC2s., Competing Interests: Competing interests: O.V., E.K.N., I.C.L., M.B. and A.G.K. (employees of the Tri-Institutional Therapeutics Discovery Institute) may benefit from the further development and licensure of molecules described here. L.A.R. is listed as an inventor of a patent related to oncolytic viral therapy. V.P.B. reports honoraria from Genentech, and Abbvie, research support from Bristol Myers Squibb and Genentech, has consulted for Merck and is listed as an inventor on a patent application related to the use of IL-33 and enhanced IL-33 for cancer immunotherapy. T.M. reports that he is a consultant for Daiichi Sankyo, Leap Therapeutics, Immunos Therapeutics and Pfizer, and co-founder of Imvaq Therapeutics. T.M. has equity in Imvaq therapeutics. T.M. has received research funding from Surface Oncology, Kyn Therapeutics, Infinity Pharmaceuticals, Peregrine Pharmaceuticals, Adaptive Biotechnologies, Leap Therapeutics and Aprea Therapeutics, and currently receives research funding from Bristol-Myers Squibb, Enterome SA and Realta Life Sciences. T.M. is listed as an inventor on patent applications related to work on oncolytic viral therapy, alphavirus-based vaccines, neo-antigen modelling, CD40, GITR, OX40, PD-1 and CTLA-4. D.A. has contributed to scientific advisory boards at Pfizer, Takeda, FARE and the KRF. The other authors declare no competing interests., (© 2025. The Author(s).)- Published
- 2025
- Full Text
- View/download PDF
7. Diversity and biogeography of the bacterial microbiome in glacier-fed streams.
- Author
-
Ezzat L, Peter H, Bourquin M, Busi SB, Michoud G, Fodelianakis S, Kohler TJ, Lamy T, Geers A, Pramateftaki P, Baier F, Marasco R, Daffonchio D, Deluigi N, Wilmes P, Styllas M, Schön M, Tolosano M, De Staercke V, and Battin TJ
- Subjects
- Metagenomics, DNA Barcoding, Taxonomic, Climate Change, Ice Cover microbiology, Rivers microbiology, Microbiota genetics, Biodiversity, Bacteria classification, Bacteria genetics, Bacteria isolation & purification, Phylogeny, Phylogeography
- Abstract
The rapid melting of mountain glaciers and the vanishing of their streams is emblematic of climate change
1,2 . Glacier-fed streams (GFSs) are cold, oligotrophic and unstable ecosystems in which life is dominated by microbial biofilms2,3 . However, current knowledge on the GFS microbiome is scarce4,5 , precluding an understanding of its response to glacier shrinkage. Here, by leveraging metabarcoding and metagenomics, we provide a comprehensive survey of bacteria in the benthic microbiome across 152 GFSs draining the Earth's major mountain ranges. We find that the GFS bacterial microbiome is taxonomically and functionally distinct from other cryospheric microbiomes. GFS bacteria are diverse, with more than half being specific to a given mountain range, some unique to single GFSs and a few cosmopolitan and abundant. We show how geographic isolation and environmental selection shape their biogeography, which is characterized by distinct compositional patterns between mountain ranges and hemispheres. Phylogenetic analyses furthermore uncovered microdiverse clades resulting from environmental selection, probably promoting functional resilience and contributing to GFS bacterial biodiversity and biogeography. Climate-induced glacier shrinkage puts this unique microbiome at risk. Our study provides a global reference for future climate-change microbiology studies on the vanishing GFS ecosystem., Competing Interests: Competing interests: The authors declare no competing interests., (© 2025. The Author(s).)- Published
- 2025
- Full Text
- View/download PDF
8. Two-factor authentication underpins the precision of the piRNA pathway.
- Author
-
Dias Mirandela M, Zoch A, Leismann J, Webb S, Berrens RV, Valsakumar D, Kabayama Y, Auchynnikava T, Schito M, Chowdhury T, MacLeod D, Xiang X, Zou J, Rappsilber J, Allshire RC, Voigt P, Cook AG, Barau J, and O'Carroll D
- Subjects
- Animals, Female, Male, Mice, Alleles, Histones chemistry, Histones metabolism, Protein Binding, Spermatogenesis genetics, Testis metabolism, Promoter Regions, Genetic genetics, RNA Precursors genetics, RNA Precursors metabolism, Argonaute Proteins metabolism, Argonaute Proteins genetics, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, DNA Methylation genetics, Long Interspersed Nucleotide Elements genetics, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Piwi-Interacting RNA genetics, Piwi-Interacting RNA metabolism, DNA Transposable Elements, Phosphoproteins genetics, Phosphoproteins metabolism
- Abstract
The PIWI-interacting RNA (piRNA) pathway guides the DNA methylation of young, active transposons during germline development in male mice
1 . piRNAs tether the PIWI protein MIWI2 (PIWIL4) to the nascent transposon transcript, resulting in DNA methylation through SPOCD1 (refs.2-5 ). Transposon methylation requires great precision: every copy needs to be methylated but off-target methylation must be avoided. However, the underlying mechanisms that ensure this precision remain unknown. Here, we show that SPOCD1 interacts directly with SPIN1 (SPINDLIN1), a chromatin reader that primarily binds to H3K4me3-K9me3 (ref.6 ). The prevailing assumption is that all the molecular events required for piRNA-directed DNA methylation occur after the engagement of MIWI2. We find that SPIN1 expression precedes that of both SPOCD1 and MIWI2. Furthermore, we demonstrate that young LINE1 copies, but not old ones, are marked by H3K4me3, H3K9me3 and SPIN1 before the initiation of piRNA-directed DNA methylation. We generated a Spocd1 separation-of-function allele in the mouse that encodes a SPOCD1 variant that no longer interacts with SPIN1. We found that the interaction between SPOCD1 and SPIN1 is essential for spermatogenesis and piRNA-directed DNA methylation of young LINE1 elements. We propose that piRNA-directed LINE1 DNA methylation requires a developmentally timed two-factor authentication process. The first authentication is the recruitment of SPIN1-SPOCD1 to the young LINE1 promoter, and the second is MIWI2 engagement with the nascent transcript. In summary, independent authentication events underpin the precision of piRNA-directed LINE1 DNA methylation., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
9. Amplification of autoimmune organ damage by NKp46-activated ILC1s.
- Author
-
Biniaris-Georgallis SI, Aschman T, Stergioula K, Schreiber F, Jafari V, Taranko A, Karmalkar T, Kasapi A, Lenac Rovis T, Jelencic V, Bejarano DA, Fabry L, Papacharalampous M, Mattiola I, Molgora M, Hou J, Hublitz KW, Heinrich F, Guerra GM, Durek P, Patone G, Lindberg EL, Maatz H, Hölsken O, Krönke G, Mortha A, Voll RE, Clarke AJ, Hauser AE, Colonna M, Thurley K, Schlitzer A, Schneider C, Stamatiades EG, Mashreghi MF, Jonjic S, Hübner N, Diefenbach A, Kanda M, and Triantafyllopoulou A
- Subjects
- Animals, Female, Humans, Mice, Antigens, Ly metabolism, Autoantibodies immunology, Epithelial Cells metabolism, Epithelial Cells pathology, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Macrophages immunology, Macrophages metabolism, Macrophages pathology, Mice, Inbred C57BL, Signal Transduction, Single-Cell Analysis, Autoimmunity, Immunity, Innate, Kidney pathology, Kidney immunology, Kidney metabolism, Lupus Nephritis immunology, Lupus Nephritis pathology, Lupus Nephritis metabolism, Lymphocytes immunology, Lymphocytes metabolism, Lymphocytes pathology, Natural Cytotoxicity Triggering Receptor 1 antagonists & inhibitors, Natural Cytotoxicity Triggering Receptor 1 deficiency, Natural Cytotoxicity Triggering Receptor 1 genetics, Natural Cytotoxicity Triggering Receptor 1 metabolism, Parenchymal Tissue immunology, Parenchymal Tissue metabolism, Parenchymal Tissue pathology
- Abstract
In systemic lupus erythematosus, loss of immune tolerance, autoantibody production and immune complex deposition are required but not sufficient for organ damage
1 . How inflammatory signals are initiated and amplified in the setting of autoimmunity remains elusive. Here we set out to dissect layers and hierarchies of autoimmune kidney inflammation to identify tissue-specific cellular hubs that amplify autoinflammatory responses. Using high-resolution single-cell profiling of kidney immune and parenchymal cells, in combination with antibody blockade and genetic deficiency, we show that tissue-resident NKp46+ innate lymphoid cells (ILCs) are crucial signal amplifiers of disease-associated macrophage expansion and epithelial cell injury in lupus nephritis, downstream of autoantibody production. NKp46 signalling in a distinct subset of group 1 ILCs (ILC1s) instructed an unconventional immune-regulatory transcriptional program, which included the expression of the myeloid cell growth factor CSF2. CSF2 production by NKp46+ ILCs promoted the population expansion of monocyte-derived macrophages. Blockade of the NKp46 receptor (using the antibody clone mNCR1.15; ref.2 ) or genetic deficiency of NKp46 abrogated epithelial cell injury. The same cellular and molecular patterns were operative in human lupus nephritis. Our data provide support for the idea that NKp46+ ILC1s promote parenchymal cell injury by granting monocyte-derived macrophages access to epithelial cell niches. NKp46 activation in ILC1s therefore constitutes a previously unrecognized, crucial tissue rheostat that amplifies organ damage in autoimmune hosts, with broad implications for inflammatory pathologies and therapies., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
10. Author Correction: TDP-43 loss and ALS-risk SNPs drive mis-splicing and depletion of UNC13A.
- Author
-
Brown AL, Wilkins OG, Keuss MJ, Hill SE, Zanovello M, Lee WC, Bampton A, Lee FCY, Masino L, Qi YA, Bryce-Smith S, Gatt A, Hallegger M, Fagegaltier D, Phatnani H, Newcombe J, Gustavsson EK, Seddighi S, Reyes JF, Coon SL, Ramos D, Schiavo G, Fisher EMC, Raj T, Secrier M, Lashley T, Ule J, Buratti E, Humphrey J, Ward ME, and Fratta P
- Published
- 2024
- Full Text
- View/download PDF
11. Plasmacytoid dendritic cells control homeostasis of megakaryopoiesis.
- Author
-
Gaertner F, Ishikawa-Ankerhold H, Stutte S, Fu W, Weitz J, Dueck A, Nelakuditi B, Fumagalli V, van den Heuvel D, Belz L, Sobirova G, Zhang Z, Titova A, Navarro AM, Pekayvaz K, Lorenz M, von Baumgarten L, Kranich J, Straub T, Popper B, Zheden V, Kaufmann WA, Guo C, Piontek G, von Stillfried S, Boor P, Colonna M, Clauß S, Schulz C, Brocker T, Walzog B, Scheiermann C, Aird WC, Nerlov C, Stark K, Petzold T, Engelhardt S, Sixt M, Hauschild R, Rudelius M, Oostendorp RAJ, Iannacone M, Heinig M, and Massberg S
- Subjects
- Animals, Female, Humans, Male, Mice, Apoptosis, Blood Platelets cytology, Bone Marrow, Cell Lineage, Cell Proliferation, Feedback, Physiological, Immunity, Innate, Intravital Microscopy, Mice, Inbred C57BL, SARS-CoV-2 immunology, COVID-19 immunology, COVID-19 physiopathology, COVID-19 virology, Dendritic Cells immunology, Dendritic Cells cytology, Homeostasis, Megakaryocytes cytology, Megakaryocytes immunology, Thrombopoiesis
- Abstract
Platelet homeostasis is essential for vascular integrity and immune defence
1,2 . Although the process of platelet formation by fragmenting megakaryocytes (MKs; thrombopoiesis) has been extensively studied, the cellular and molecular mechanisms required to constantly replenish the pool of MKs by their progenitor cells (megakaryopoiesis) remains unclear3,4 . Here we use intravital imaging to track the cellular dynamics of megakaryopoiesis over days. We identify plasmacytoid dendritic cells (pDCs) as homeostatic sensors that monitor the bone marrow for apoptotic MKs and deliver IFNα to the MK niche triggering local on-demand proliferation and maturation of MK progenitors. This pDC-dependent feedback loop is crucial for MK and platelet homeostasis at steady state and under stress. pDCs are best known for their ability to function as vigilant detectors of viral infection5 . We show that virus-induced activation of pDCs interferes with their function as homeostatic sensors of megakaryopoiesis. Consequently, activation of pDCs by SARS-CoV-2 leads to excessive megakaryopoiesis. Together, we identify a pDC-dependent homeostatic circuit that involves innate immune sensing and demand-adapted release of inflammatory mediators to maintain homeostasis of the megakaryocytic lineage., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
12. A virally encoded high-resolution screen of cytomegalovirus dependencies.
- Author
-
Finkel Y, Nachshon A, Aharon E, Arazi T, Simonovsky E, Dobešová M, Saud Z, Gluck A, Fisher T, Stanton RJ, Schwartz M, and Stern-Ginossar N
- Subjects
- Humans, Cell Line, Genome, Viral genetics, Virion genetics, Virion metabolism, Virus Assembly genetics, Virus Release genetics, CRISPR-Cas Systems genetics, Cytomegalovirus genetics, Cytomegalovirus physiology, Cytomegalovirus Infections genetics, Cytomegalovirus Infections virology, Host-Pathogen Interactions genetics, RNA, Guide, CRISPR-Cas Systems genetics, RNA, Guide, CRISPR-Cas Systems metabolism, Virus Replication genetics
- Abstract
Genetic screens have transformed our ability to interrogate cellular factor requirements for viral infections
1,2 , but most current approaches are limited in their sensitivity, biased towards early stages of infection and provide only simplistic phenotypic information that is often based on survival of infected cells2-4 . Here, by engineering human cytomegalovirus to express single guide RNA libraries directly from the viral genome, we developed virus-encoded CRISPR-based direct readout screening (VECOS), a sensitive, versatile, viral-centric approach that enables profiling of different stages of viral infection in a pooled format. Using this approach, we identified hundreds of host dependency and restriction factors and quantified their direct effects on viral genome replication, viral particle secretion and infectiousness of secreted particles, providing a multi-dimensional perspective on virus-host interactions. These high-resolution measurements reveal that perturbations altering late stages in the life cycle of human cytomegalovirus (HCMV) mostly regulate viral particle quality rather than quantity, establishing correct virion assembly as a critical stage that is heavily reliant on virus-host interactions. Overall, VECOS facilitates systematic high-resolution dissection of the role of human proteins during the infection cycle, providing a roadmap for in-depth study of host-herpesvirus interactions., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
13. Author Correction: Controlling the helicity of light by electrical magnetization switching.
- Author
-
Dainone PA, Prestes NF, Renucci P, Bouché A, Morassi M, Devaux X, Lindemann M, George JM, Jaffrès H, Lemaitre A, Xu B, Stoffel M, Chen T, Lombez L, Lagarde D, Cong G, Ma T, Pigeat P, Vergnat M, Rinnert H, Marie X, Han X, Mangin S, Rojas-Sánchez JC, Wang JP, Beard MC, Gerhardt NC, Žutić I, and Lu Y
- Published
- 2024
- Full Text
- View/download PDF
14. FOXO1 is a master regulator of memory programming in CAR T cells.
- Author
-
Doan AE, Mueller KP, Chen AY, Rouin GT, Chen Y, Daniel B, Lattin J, Markovska M, Mozarsky B, Arias-Umana J, Hapke R, Jung IY, Wang A, Xu P, Klysz D, Zuern G, Bashti M, Quinn PJ, Miao Z, Sandor K, Zhang W, Chen GM, Ryu F, Logun M, Hall J, Tan K, Grupp SA, McClory SE, Lareau CA, Fraietta JA, Sotillo E, Satpathy AT, Mackall CL, and Weber EW
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Chromatin metabolism, Chromatin genetics, Gene Editing, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Forkhead Box Protein O1 metabolism, Immunologic Memory, Immunotherapy, Adoptive, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism, Receptors, Chimeric Antigen genetics, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes cytology
- Abstract
A major limitation of chimeric antigen receptor (CAR) T cell therapies is the poor persistence of these cells in vivo
1 . The expression of memory-associated genes in CAR T cells is linked to their long-term persistence in patients and clinical efficacy2-6 , suggesting that memory programs may underpin durable CAR T cell function. Here we show that the transcription factor FOXO1 is responsible for promoting memory and restraining exhaustion in human CAR T cells. Pharmacological inhibition or gene editing of endogenous FOXO1 diminished the expression of memory-associated genes, promoted an exhaustion-like phenotype and impaired the antitumour activity of CAR T cells. Overexpression of FOXO1 induced a gene-expression program consistent with T cell memory and increased chromatin accessibility at FOXO1-binding motifs. CAR T cells that overexpressed FOXO1 retained their function, memory potential and metabolic fitness in settings of chronic stimulation, and exhibited enhanced persistence and tumour control in vivo. By contrast, overexpression of TCF1 (encoded by TCF7) did not enforce canonical memory programs or enhance the potency of CAR T cells. Notably, FOXO1 activity correlated with positive clinical outcomes of patients treated with CAR T cells or tumour-infiltrating lymphocytes, underscoring the clinical relevance of FOXO1 in cancer immunotherapy. Our results show that overexpressing FOXO1 can increase the antitumour activity of human CAR T cells, and highlight memory reprogramming as a broadly applicable approach for optimizing therapeutic T cell states., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
15. Publisher Correction: FOXO1 is a master regulator of memory programming in CAR T cells.
- Author
-
Doan AE, Mueller KP, Chen AY, Rouin GT, Chen Y, Daniel B, Lattin J, Markovska M, Mozarsky B, Arias-Umana J, Hapke R, Jung IY, Wang A, Xu P, Klysz D, Zuern G, Bashti M, Quinn PJ, Miao Z, Sandor K, Zhang W, Chen GM, Ryu F, Logun M, Hall J, Tan K, Grupp SA, McClory SE, Lareau CA, Fraietta JA, Sotillo E, Satpathy AT, Mackall CL, and Weber EW
- Published
- 2024
- Full Text
- View/download PDF
16. Controlling the helicity of light by electrical magnetization switching.
- Author
-
Dainone PA, Prestes NF, Renucci P, Bouché A, Morassi M, Devaux X, Lindemann M, George JM, Jaffrès H, Lemaitre A, Xu B, Stoffel M, Chen T, Lombez L, Lagarde D, Cong G, Ma T, Pigeat P, Vergnat M, Rinnert H, Marie X, Han X, Mangin S, Rojas-Sánchez JC, Wang JP, Beard MC, Gerhardt NC, Žutić I, and Lu Y
- Abstract
Controlling the intensity of emitted light and charge current is the basis of transferring and processing information
1 . By contrast, robust information storage and magnetic random-access memories are implemented using the spin of the carrier and the associated magnetization in ferromagnets2 . The missing link between the respective disciplines of photonics, electronics and spintronics is to modulate the circular polarization of the emitted light, rather than its intensity, by electrically controlled magnetization. Here we demonstrate that this missing link is established at room temperature and zero applied magnetic field in light-emitting diodes2-7 , through the transfer of angular momentum between photons, electrons and ferromagnets. With spin-orbit torque8-11 , a charge current generates also a spin current to electrically switch the magnetization. This switching determines the spin orientation of injected carriers into semiconductors, in which the transfer of angular momentum from the electron spin to photon controls the circular polarization of the emitted light2 . The spin-photon conversion with the nonvolatile control of magnetization opens paths to seamlessly integrate information transfer, processing and storage. Our results provide substantial advances towards electrically controlled ultrafast modulation of circular polarization and spin injection with magnetization dynamics for the next-generation information and communication technology12 , including space-light data transfer. The same operating principle in scaled-down structures or using two-dimensional materials will enable transformative opportunities for quantum information processing with spin-controlled single-photon sources, as well as for implementing spin-dependent time-resolved spectroscopies., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
17. The UFM1 E3 ligase recognizes and releases 60S ribosomes from ER translocons.
- Author
-
Makhlouf L, Peter JJ, Magnussen HM, Thakur R, Millrine D, Minshull TC, Harrison G, Varghese J, Lamoliatte F, Foglizzo M, Macartney T, Calabrese AN, Zeqiraj E, and Kulathu Y
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Binding Sites, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Cycle Proteins ultrastructure, Cryoelectron Microscopy, Homeostasis, Intracellular Membranes metabolism, Peptidyl Transferases chemistry, Peptidyl Transferases metabolism, Peptidyl Transferases ultrastructure, Ribosomal Proteins chemistry, Ribosomal Proteins metabolism, Ribosomal Proteins ultrastructure, RNA, Transfer metabolism, SEC Translocation Channels chemistry, SEC Translocation Channels metabolism, SEC Translocation Channels ultrastructure, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins metabolism, Tumor Suppressor Proteins ultrastructure, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum ultrastructure, Protein Processing, Post-Translational, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases ultrastructure, Ribosome Subunits, Large, Eukaryotic chemistry, Ribosome Subunits, Large, Eukaryotic metabolism, Ribosome Subunits, Large, Eukaryotic ultrastructure
- Abstract
Stalled ribosomes at the endoplasmic reticulum (ER) are covalently modified with the ubiquitin-like protein UFM1 on the 60S ribosomal subunit protein RPL26 (also known as uL24)
1,2 . This modification, which is known as UFMylation, is orchestrated by the UFM1 ribosome E3 ligase (UREL) complex, comprising UFL1, UFBP1 and CDK5RAP3 (ref.3 ). However, the catalytic mechanism of UREL and the functional consequences of UFMylation are unclear. Here we present cryo-electron microscopy structures of UREL bound to 60S ribosomes, revealing the basis of its substrate specificity. UREL wraps around the 60S subunit to form a C-shaped clamp architecture that blocks the tRNA-binding sites at one end, and the peptide exit tunnel at the other. A UFL1 loop inserts into and remodels the peptidyl transferase centre. These features of UREL suggest a crucial function for UFMylation in the release and recycling of stalled or terminated ribosomes from the ER membrane. In the absence of functional UREL, 60S-SEC61 translocon complexes accumulate at the ER membrane, demonstrating that UFMylation is necessary for releasing SEC61 from 60S subunits. Notably, this release is facilitated by a functional switch of UREL from a 'writer' to a 'reader' module that recognizes its product-UFMylated 60S ribosomes. Collectively, we identify a fundamental role for UREL in dissociating 60S subunits from the SEC61 translocon and the basis for UFMylation in regulating protein homeostasis at the ER., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
18. A model of human neural networks reveals NPTX2 pathology in ALS and FTLD.
- Author
-
Hruska-Plochan M, Wiersma VI, Betz KM, Mallona I, Ronchi S, Maniecka Z, Hock EM, Tantardini E, Laferriere F, Sahadevan S, Hoop V, Delvendahl I, Pérez-Berlanga M, Gatta B, Panatta M, van der Bourg A, Bohaciakova D, Sharma P, De Vos L, Frontzek K, Aguzzi A, Lashley T, Robinson MD, Karayannis T, Mueller M, Hierlemann A, and Polymenidou M
- Subjects
- Humans, Neural Stem Cells cytology, Neuroglia cytology, Reproducibility of Results, Amyotrophic Lateral Sclerosis metabolism, Amyotrophic Lateral Sclerosis pathology, C-Reactive Protein metabolism, DNA-Binding Proteins deficiency, DNA-Binding Proteins metabolism, Frontotemporal Lobar Degeneration metabolism, Frontotemporal Lobar Degeneration pathology, Nerve Net metabolism, Nerve Net pathology, Nerve Tissue Proteins metabolism, Neurons cytology, Neurons metabolism
- Abstract
Human cellular models of neurodegeneration require reproducibility and longevity, which is necessary for simulating age-dependent diseases. Such systems are particularly needed for TDP-43 proteinopathies
1 , which involve human-specific mechanisms2-5 that cannot be directly studied in animal models. Here, to explore the emergence and consequences of TDP-43 pathologies, we generated induced pluripotent stem cell-derived, colony morphology neural stem cells (iCoMoNSCs) via manual selection of neural precursors6 . Single-cell transcriptomics and comparison to independent neural stem cells7 showed that iCoMoNSCs are uniquely homogenous and self-renewing. Differentiated iCoMoNSCs formed a self-organized multicellular system consisting of synaptically connected and electrophysiologically active neurons, which matured into long-lived functional networks (which we designate iNets). Neuronal and glial maturation in iNets was similar to that of cortical organoids8 . Overexpression of wild-type TDP-43 in a minority of neurons within iNets led to progressive fragmentation and aggregation of the protein, resulting in a partial loss of function and neurotoxicity. Single-cell transcriptomics revealed a novel set of misregulated RNA targets in TDP-43-overexpressing neurons and in patients with TDP-43 proteinopathies exhibiting a loss of nuclear TDP-43. The strongest misregulated target encoded the synaptic protein NPTX2, the levels of which are controlled by TDP-43 binding on its 3' untranslated region. When NPTX2 was overexpressed in iNets, it exhibited neurotoxicity, whereas correcting NPTX2 misregulation partially rescued neurons from TDP-43-induced neurodegeneration. Notably, NPTX2 was consistently misaccumulated in neurons from patients with amyotrophic lateral sclerosis and frontotemporal lobar degeneration with TDP-43 pathology. Our work directly links TDP-43 misregulation and NPTX2 accumulation, thereby revealing a TDP-43-dependent pathway of neurotoxicity., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
19. In vitro production of cat-restricted Toxoplasma pre-sexual stages.
- Author
-
Antunes AV, Shahinas M, Swale C, Farhat DC, Ramakrishnan C, Bruley C, Cannella D, Robert MG, Corrao C, Couté Y, Hehl AB, Bougdour A, Coppens I, and Hakimi MA
- Subjects
- Animals, Humans, Chromatin genetics, Chromatin metabolism, Disease Models, Animal, Epigenesis, Genetic, Merozoites genetics, Nuclear Proteins metabolism, Promoter Regions, Genetic genetics, Protozoan Proteins genetics, Protozoan Proteins metabolism, Toxoplasmosis genetics, Toxoplasmosis parasitology, Toxoplasmosis transmission, Transcription, Genetic, Cats parasitology, In Vitro Techniques methods, Life Cycle Stages genetics, Toxoplasma genetics, Toxoplasma growth & development, Toxoplasma physiology
- Abstract
Sexual reproduction of Toxoplasma gondii, confined to the felid gut, remains largely uncharted owing to ethical concerns regarding the use of cats as model organisms. Chromatin modifiers dictate the developmental fate of the parasite during its multistage life cycle, but their targeting to stage-specific cistromes is poorly described
1,2 . Here we found that the transcription factors AP2XII-1 and AP2XI-2 operate during the tachyzoite stage, a hallmark of acute toxoplasmosis, to silence genes necessary for merozoites, a developmental stage critical for subsequent sexual commitment and transmission to the next host, including humans. Their conditional and simultaneous depletion leads to a marked change in the transcriptional program, promoting a full transition from tachyzoites to merozoites. These in vitro-cultured pre-gametes have unique protein markers and undergo typical asexual endopolygenic division cycles. In tachyzoites, AP2XII-1 and AP2XI-2 bind DNA as heterodimers at merozoite promoters and recruit MORC and HDAC3 (ref.1 ), thereby limiting chromatin accessibility and transcription. Consequently, the commitment to merogony stems from a profound epigenetic rewiring orchestrated by AP2XII-1 and AP2XI-2. Successful production of merozoites in vitro paves the way for future studies on Toxoplasma sexual development without the need for cat infections and holds promise for the development of therapies to prevent parasite transmission., (© 2023. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
20. MYC protein helps cancer to take its vitamins.
- Author
-
Wallace M
- Subjects
- Humans, Vitamin A, Vitamin K, Proto-Oncogene Proteins c-myc metabolism, Vitamins, Neoplasms metabolism
- Published
- 2023
- Full Text
- View/download PDF
21. Neutralization, effector function and immune imprinting of Omicron variants.
- Author
-
Addetia A, Piccoli L, Case JB, Park YJ, Beltramello M, Guarino B, Dang H, de Melo GD, Pinto D, Sprouse K, Scheaffer SM, Bassi J, Silacci-Fregni C, Muoio F, Dini M, Vincenzetti L, Acosta R, Johnson D, Subramanian S, Saliba C, Giurdanella M, Lombardo G, Leoni G, Culap K, McAlister C, Rajesh A, Dellota E Jr, Zhou J, Farhat N, Bohan D, Noack J, Chen A, Lempp FA, Quispe J, Kergoat L, Larrous F, Cameroni E, Whitener B, Giannini O, Cippà P, Ceschi A, Ferrari P, Franzetti-Pellanda A, Biggiogero M, Garzoni C, Zappi S, Bernasconi L, Kim MJ, Rosen LE, Schnell G, Czudnochowski N, Benigni F, Franko N, Logue JK, Yoshiyama C, Stewart C, Chu H, Bourhy H, Schmid MA, Purcell LA, Snell G, Lanzavecchia A, Diamond MS, Corti D, and Veesler D
- Subjects
- Animals, Cricetinae, Humans, Mice, Angiotensin-Converting Enzyme 2 immunology, Angiotensin-Converting Enzyme 2 metabolism, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal immunology, Cross Reactions, Immune Evasion, Membrane Fusion, Neutralization Tests, Mutation, Memory B Cells immunology, COVID-19 Vaccines immunology, Antibodies, Neutralizing chemistry, Antibodies, Neutralizing immunology, COVID-19 immunology, COVID-19 prevention & control, COVID-19 virology, SARS-CoV-2 classification, SARS-CoV-2 genetics, SARS-CoV-2 immunology
- Abstract
Currently circulating SARS-CoV-2 variants have acquired convergent mutations at hot spots in the receptor-binding domain
1 (RBD) of the spike protein. The effects of these mutations on viral infection and transmission and the efficacy of vaccines and therapies remains poorly understood. Here we demonstrate that recently emerged BQ.1.1 and XBB.1.5 variants bind host ACE2 with high affinity and promote membrane fusion more efficiently than earlier Omicron variants. Structures of the BQ.1.1, XBB.1 and BN.1 RBDs bound to the fragment antigen-binding region of the S309 antibody (the parent antibody for sotrovimab) and human ACE2 explain the preservation of antibody binding through conformational selection, altered ACE2 recognition and immune evasion. We show that sotrovimab binds avidly to all Omicron variants, promotes Fc-dependent effector functions and protects mice challenged with BQ.1.1 and hamsters challenged with XBB.1.5. Vaccine-elicited human plasma antibodies cross-react with and trigger effector functions against current Omicron variants, despite a reduced neutralizing activity, suggesting a mechanism of protection against disease, exemplified by S309. Cross-reactive RBD-directed human memory B cells remained dominant even after two exposures to Omicron spikes, underscoring the role of persistent immune imprinting., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
22. PD-1 maintains CD8 T cell tolerance towards cutaneous neoantigens.
- Author
-
Damo M, Hornick NI, Venkat A, William I, Clulo K, Venkatesan S, He J, Fagerberg E, Loza JL, Kwok D, Tal A, Buck J, Cui C, Singh J, Damsky WE, Leventhal JS, Krishnaswamy S, and Joshi NS
- Subjects
- Animals, Humans, Mice, Biopsy, Epidermis immunology, Epidermis metabolism, Gene Expression Profiling, Lichen Planus immunology, Lichen Planus pathology, Antigens immunology, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes pathology, Immune Tolerance, Programmed Cell Death 1 Receptor immunology, Programmed Cell Death 1 Receptor metabolism, Skin cytology, Skin immunology, Skin metabolism, Skin pathology
- Abstract
The peripheral T cell repertoire of healthy individuals contains self-reactive T cells
1,2 . Checkpoint receptors such as PD-1 are thought to enable the induction of peripheral tolerance by deletion or anergy of self-reactive CD8 T cells3-10 . However, this model is challenged by the high frequency of immune-related adverse events in patients with cancer who have been treated with checkpoint inhibitors11 . Here we developed a mouse model in which skin-specific expression of T cell antigens in the epidermis caused local infiltration of antigen-specific CD8 T cells with an effector gene-expression profile. In this setting, PD-1 enabled the maintenance of skin tolerance by preventing tissue-infiltrating antigen-specific effector CD8 T cells from (1) acquiring a fully functional, pathogenic differentiation state, (2) secreting significant amounts of effector molecules, and (3) gaining access to epidermal antigen-expressing cells. In the absence of PD-1, epidermal antigen-expressing cells were eliminated by antigen-specific CD8 T cells, resulting in local pathology. Transcriptomic analysis of skin biopsies from two patients with cutaneous lichenoid immune-related adverse events showed the presence of clonally expanded effector CD8 T cells in both lesional and non-lesional skin. Thus, our data support a model of peripheral T cell tolerance in which PD-1 allows antigen-specific effector CD8 T cells to co-exist with antigen-expressing cells in tissues without immunopathology., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
23. Cooperation between bHLH transcription factors and histones for DNA access.
- Author
-
Michael AK, Stoos L, Crosby P, Eggers N, Nie XY, Makasheva K, Minnich M, Healy KL, Weiss J, Kempf G, Cavadini S, Kater L, Seebacher J, Vecchia L, Chakraborty D, Isbel L, Grand RS, Andersch F, Fribourgh JL, Schübeler D, Zuber J, Liu AC, Becker PB, Fierz B, Partch CL, Menet JS, and Thomä NH
- Subjects
- ARNTL Transcription Factors genetics, Helix-Loop-Helix Motifs genetics, Nucleosomes chemistry, Nucleosomes genetics, Nucleosomes metabolism, Protein Binding, CLOCK Proteins chemistry, CLOCK Proteins metabolism, Proto-Oncogene Proteins c-myc chemistry, Proto-Oncogene Proteins c-myc metabolism, Allosteric Regulation, Leucine Zippers, Octamer Transcription Factor-3 metabolism, Protein Multimerization, Basic Helix-Loop-Helix Transcription Factors metabolism, DNA genetics, DNA metabolism, Histones chemistry, Histones metabolism
- Abstract
The basic helix-loop-helix (bHLH) family of transcription factors recognizes DNA motifs known as E-boxes (CANNTG) and includes 108 members
1 . Here we investigate how chromatinized E-boxes are engaged by two structurally diverse bHLH proteins: the proto-oncogene MYC-MAX and the circadian transcription factor CLOCK-BMAL1 (refs.2,3 ). Both transcription factors bind to E-boxes preferentially near the nucleosomal entry-exit sites. Structural studies with engineered or native nucleosome sequences show that MYC-MAX or CLOCK-BMAL1 triggers the release of DNA from histones to gain access. Atop the H2A-H2B acidic patch4 , the CLOCK-BMAL1 Per-Arnt-Sim (PAS) dimerization domains engage the histone octamer disc. Binding of tandem E-boxes5-7 at endogenous DNA sequences occurs through direct interactions between two CLOCK-BMAL1 protomers and histones and is important for circadian cycling. At internal E-boxes, the MYC-MAX leucine zipper can also interact with histones H2B and H3, and its binding is indirectly enhanced by OCT4 elsewhere on the nucleosome. The nucleosomal E-box position and the type of bHLH dimerization domain jointly determine the histone contact, the affinity and the degree of competition and cooperativity with other nucleosome-bound factors., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
24. A small-molecule PI3Kα activator for cardioprotection and neuroregeneration.
- Author
-
Gong GQ, Bilanges B, Allsop B, Masson GR, Roberton V, Askwith T, Oxenford S, Madsen RR, Conduit SE, Bellini D, Fitzek M, Collier M, Najam O, He Z, Wahab B, McLaughlin SH, Chan AWE, Feierberg I, Madin A, Morelli D, Bhamra A, Vinciauskaite V, Anderson KE, Surinova S, Pinotsis N, Lopez-Guadamillas E, Wilcox M, Hooper A, Patel C, Whitehead MA, Bunney TD, Stephens LR, Hawkins PT, Katan M, Yellon DM, Davidson SM, Smith DM, Phillips JB, Angell R, Williams RL, and Vanhaesebroeck B
- Subjects
- Humans, Neoplasms drug therapy, Protein Isoforms agonists, Signal Transduction drug effects, Class I Phosphatidylinositol 3-Kinases chemistry, Class I Phosphatidylinositol 3-Kinases drug effects, Cardiotonic Agents pharmacology, Animals, Biocatalysis drug effects, Protein Conformation drug effects, Neurites drug effects, Reperfusion Injury prevention & control, Nerve Crush, Cell Proliferation drug effects, Nerve Regeneration drug effects
- Abstract
Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development
1-5 . This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
25. Author Correction: Palaeogenomics of Upper Palaeolithic to Neolithic European hunter-gatherers.
- Author
-
Posth C, Yu H, Ghalichi A, Rougier H, Crevecoeur I, Huang Y, Ringbauer H, Rohrlach AB, Nägele K, Villalba-Mouco V, Radzeviciute R, Ferraz T, Stoessel A, Tukhbatova R, Drucker DG, Lari M, Modi A, Vai S, Saupe T, Scheib CL, Catalano G, Pagani L, Talamo S, Fewlass H, Klaric L, Morala A, Rué M, Madelaine S, Crépin L, Caverne JB, Bocaege E, Ricci S, Boschin F, Bayle P, Maureille B, Le Brun-Ricalens F, Bordes JG, Oxilia G, Bortolini E, Bignon-Lau O, Debout G, Orliac M, Zazzo A, Sparacello V, Starnini E, Sineo L, van der Plicht J, Pecqueur L, Merceron G, Garcia G, Leuvrey JM, Garcia CB, Gómez-Olivencia A, Połtowicz-Bobak M, Bobak D, Le Luyer M, Storm P, Hoffmann C, Kabaciński J, Filimonova T, Shnaider S, Berezina N, González-Rabanal B, González Morales MR, Marín-Arroyo AB, López B, Alonso-Llamazares C, Ronchitelli A, Polet C, Jadin I, Cauwe N, Soler J, Coromina N, Rufí I, Cottiaux R, Clark G, Straus LG, Julien MA, Renhart S, Talaa D, Benazzi S, Romandini M, Amkreutz L, Bocherens H, Wißing C, Villotte S, de Pablo JF, Gómez-Puche M, Esquembre-Bebia MA, Bodu P, Smits L, Souffi B, Jankauskas R, Kozakaitė J, Cupillard C, Benthien H, Wehrberger K, Schmitz RW, Feine SC, Schüler T, Thevenet C, Grigorescu D, Lüth F, Kotula A, Piezonka H, Schopper F, Svoboda J, Sázelová S, Chizhevsky A, Khokhlov A, Conard NJ, Valentin F, Harvati K, Semal P, Jungklaus B, Suvorov A, Schulting R, Moiseyev V, Mannermaa K, Buzhilova A, Terberger T, Caramelli D, Altena E, Haak W, and Krause J
- Published
- 2023
- Full Text
- View/download PDF
26. Diode effect in Josephson junctions with a single magnetic atom.
- Author
-
Trahms M, Melischek L, Steiner JF, Mahendru B, Tamir I, Bogdanoff N, Peters O, Reecht G, Winkelmann CB, von Oppen F, and Franke KJ
- Abstract
Current flow in electronic devices can be asymmetric with bias direction, a phenomenon underlying the utility of diodes
1 and known as non-reciprocal charge transport2 . The promise of dissipationless electronics has recently stimulated the quest for superconducting diodes, and non-reciprocal superconducting devices have been realized in various non-centrosymmetric systems3-10 . Here we investigate the ultimate limits of miniaturization by creating atomic-scale Pb-Pb Josephson junctions in a scanning tunnelling microscope. Pristine junctions stabilized by a single Pb atom exhibit hysteretic behaviour, confirming the high quality of the junctions, but no asymmetry between the bias directions. Non-reciprocal supercurrents emerge when inserting a single magnetic atom into the junction, with the preferred direction depending on the atomic species. Aided by theoretical modelling, we trace the non-reciprocity to quasiparticle currents flowing by means of electron-hole asymmetric Yu-Shiba-Rusinov states inside the superconducting energy gap and identify a new mechanism for diode behaviour in Josephson junctions. Our results open new avenues for creating atomic-scale Josephson diodes and tuning their properties through single-atom manipulation., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
27. Palaeogenomics of Upper Palaeolithic to Neolithic European hunter-gatherers.
- Author
-
Posth C, Yu H, Ghalichi A, Rougier H, Crevecoeur I, Huang Y, Ringbauer H, Rohrlach AB, Nägele K, Villalba-Mouco V, Radzeviciute R, Ferraz T, Stoessel A, Tukhbatova R, Drucker DG, Lari M, Modi A, Vai S, Saupe T, Scheib CL, Catalano G, Pagani L, Talamo S, Fewlass H, Klaric L, Morala A, Rué M, Madelaine S, Crépin L, Caverne JB, Bocaege E, Ricci S, Boschin F, Bayle P, Maureille B, Le Brun-Ricalens F, Bordes JG, Oxilia G, Bortolini E, Bignon-Lau O, Debout G, Orliac M, Zazzo A, Sparacello V, Starnini E, Sineo L, van der Plicht J, Pecqueur L, Merceron G, Garcia G, Leuvrey JM, Garcia CB, Gómez-Olivencia A, Połtowicz-Bobak M, Bobak D, Le Luyer M, Storm P, Hoffmann C, Kabaciński J, Filimonova T, Shnaider S, Berezina N, González-Rabanal B, González Morales MR, Marín-Arroyo AB, López B, Alonso-Llamazares C, Ronchitelli A, Polet C, Jadin I, Cauwe N, Soler J, Coromina N, Rufí I, Cottiaux R, Clark G, Straus LG, Julien MA, Renhart S, Talaa D, Benazzi S, Romandini M, Amkreutz L, Bocherens H, Wißing C, Villotte S, de Pablo JF, Gómez-Puche M, Esquembre-Bebia MA, Bodu P, Smits L, Souffi B, Jankauskas R, Kozakaitė J, Cupillard C, Benthien H, Wehrberger K, Schmitz RW, Feine SC, Schüler T, Thevenet C, Grigorescu D, Lüth F, Kotula A, Piezonka H, Schopper F, Svoboda J, Sázelová S, Chizhevsky A, Khokhlov A, Conard NJ, Valentin F, Harvati K, Semal P, Jungklaus B, Suvorov A, Schulting R, Moiseyev V, Mannermaa K, Buzhilova A, Terberger T, Caramelli D, Altena E, Haak W, and Krause J
- Subjects
- Humans, Europe ethnology, Gene Pool, History, Ancient, Archaeology, Genomics, Hunting, Paleontology, Human Genetics, Genome, Human genetics
- Abstract
Modern humans have populated Europe for more than 45,000 years
1,2 . Our knowledge of the genetic relatedness and structure of ancient hunter-gatherers is however limited, owing to the scarceness and poor molecular preservation of human remains from that period3 . Here we analyse 356 ancient hunter-gatherer genomes, including new genomic data for 116 individuals from 14 countries in western and central Eurasia, spanning between 35,000 and 5,000 years ago. We identify a genetic ancestry profile in individuals associated with Upper Palaeolithic Gravettian assemblages from western Europe that is distinct from contemporaneous groups related to this archaeological culture in central and southern Europe4 , but resembles that of preceding individuals associated with the Aurignacian culture. This ancestry profile survived during the Last Glacial Maximum (25,000 to 19,000 years ago) in human populations from southwestern Europe associated with the Solutrean culture, and with the following Magdalenian culture that re-expanded northeastward after the Last Glacial Maximum. Conversely, we reveal a genetic turnover in southern Europe suggesting a local replacement of human groups around the time of the Last Glacial Maximum, accompanied by a north-to-south dispersal of populations associated with the Epigravettian culture. From at least 14,000 years ago, an ancestry related to this culture spread from the south across the rest of Europe, largely replacing the Magdalenian-associated gene pool. After a period of limited admixture that spanned the beginning of the Mesolithic, we find genetic interactions between western and eastern European hunter-gatherers, who were also characterized by marked differences in phenotypically relevant variants., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
28. Insulin-regulated serine and lipid metabolism drive peripheral neuropathy.
- Author
-
Handzlik MK, Gengatharan JM, Frizzi KE, McGregor GH, Martino C, Rahman G, Gonzalez A, Moreno AM, Green CR, Guernsey LS, Lin T, Tseng P, Ideguchi Y, Fallon RJ, Chaix A, Panda S, Mali P, Wallace M, Knight R, Gantner ML, Calcutt NA, and Metallo CM
- Subjects
- Animals, Mice, Glycine metabolism, Diet, High-Fat, Adiposity, Sphingolipids metabolism, Small Fiber Neuropathy, Dyslipidemias, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Experimental metabolism, Insulin metabolism, Lipid Metabolism, Peripheral Nervous System Diseases metabolism, Serine metabolism
- Abstract
Diabetes represents a spectrum of disease in which metabolic dysfunction damages multiple organ systems including liver, kidneys and peripheral nerves
1,2 . Although the onset and progression of these co-morbidities are linked with insulin resistance, hyperglycaemia and dyslipidaemia3-7 , aberrant non-essential amino acid (NEAA) metabolism also contributes to the pathogenesis of diabetes8-10 . Serine and glycine are closely related NEAAs whose levels are consistently reduced in patients with metabolic syndrome10-14 , but the mechanistic drivers and downstream consequences of this metabotype remain unclear. Low systemic serine and glycine are also emerging as a hallmark of macular and peripheral nerve disorders, correlating with impaired visual acuity and peripheral neuropathy15,16 . Here we demonstrate that aberrant serine homeostasis drives serine and glycine deficiencies in diabetic mice, which can be diagnosed with a serine tolerance test that quantifies serine uptake and disposal. Mimicking these metabolic alterations in young mice by dietary serine or glycine restriction together with high fat intake markedly accelerates the onset of small fibre neuropathy while reducing adiposity. Normalization of serine by dietary supplementation and mitigation of dyslipidaemia with myriocin both alleviate neuropathy in diabetic mice, linking serine-associated peripheral neuropathy to sphingolipid metabolism. These findings identify systemic serine deficiency and dyslipidaemia as novel risk factors for peripheral neuropathy that may be exploited therapeutically., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
29. Antibody feedback regulates immune memory after SARS-CoV-2 mRNA vaccination.
- Author
-
Schaefer-Babajew D, Wang Z, Muecksch F, Cho A, Loewe M, Cipolla M, Raspe R, Johnson B, Canis M, DaSilva J, Ramos V, Turroja M, Millard KG, Schmidt F, Witte L, Dizon J, Shimeliovich I, Yao KH, Oliveira TY, Gazumyan A, Gaebler C, Bieniasz PD, Hatziioannou T, Caskey M, and Nussenzweig MC
- Subjects
- Animals, Mice, Antibodies, Monoclonal immunology, Antibodies, Monoclonal therapeutic use, Antibodies, Neutralizing immunology, SARS-CoV-2 immunology, Epitopes, B-Lymphocyte chemistry, Epitopes, B-Lymphocyte immunology, Immunoglobulin M immunology, Germinal Center cytology, Germinal Center immunology, Immunization, Secondary, Somatic Hypermutation, Immunoglobulin, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 prevention & control, COVID-19 therapy, COVID-19 virology, Immunologic Memory, Vaccination, mRNA Vaccines immunology, COVID-19 Vaccines immunology, B-Lymphocytes immunology, B-Lymphocytes metabolism, Feedback, Physiological
- Abstract
Feedback inhibition of humoral immunity by antibodies was first documented in 1909
1 . Subsequent studies showed that, depending on the context, antibodies can enhance or inhibit immune responses2,3 . However, little is known about how pre-existing antibodies influence the development of memory B cells. Here we examined the memory B cell response in individuals who received two high-affinity anti-SARS-CoV-2 monoclonal antibodies and subsequently two doses of an mRNA vaccine4-8 . We found that the recipients of the monoclonal antibodies produced antigen-binding and neutralizing titres that were only fractionally lower compared than in control individuals. However, the memory B cells of the individuals who received the monoclonal antibodies differed from those of control individuals in that they predominantly expressed low-affinity IgM antibodies that carried small numbers of somatic mutations and showed altered receptor binding domain (RBD) target specificity, consistent with epitope masking. Moreover, only 1 out of 77 anti-RBD memory antibodies tested neutralized the virus. The mechanism underlying these findings was examined in experiments in mice that showed that germinal centres formed in the presence of the same antibodies were dominated by low-affinity B cells. Our results indicate that pre-existing high-affinity antibodies bias germinal centre and memory B cell selection through two distinct mechanisms: (1) by lowering the activation threshold for B cells, thereby permitting abundant lower-affinity clones to participate in the immune response; and (2) through direct masking of their cognate epitopes. This may in part explain the shifting target profile of memory antibodies elicited by booster vaccinations9 ., (© 2022. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
30. Structures and mechanism of the plant PIN-FORMED auxin transporter.
- Author
-
Ung KL, Winkler M, Schulz L, Kolb M, Janacek DP, Dedic E, Stokes DL, Hammes UZ, and Pedersen BP
- Subjects
- Antiporters metabolism, Bicarbonates metabolism, Bile Acids and Salts metabolism, Binding Sites, Biological Transport, Herbicides metabolism, Phthalimides metabolism, Plant Growth Regulators chemistry, Plant Growth Regulators metabolism, Proline metabolism, Protein Domains, Protein Multimerization, Protons, Sodium metabolism, Symporters metabolism, Arabidopsis chemistry, Arabidopsis metabolism, Arabidopsis Proteins chemistry, Arabidopsis Proteins metabolism, Indoleacetic Acids chemistry, Indoleacetic Acids metabolism, Membrane Transport Proteins chemistry, Membrane Transport Proteins metabolism
- Abstract
Auxins are hormones that have central roles and control nearly all aspects of growth and development in plants
1-3 . The proteins in the PIN-FORMED (PIN) family (also known as the auxin efflux carrier family) are key participants in this process and control auxin export from the cytosol to the extracellular space4-9 . Owing to a lack of structural and biochemical data, the molecular mechanism of PIN-mediated auxin transport is not understood. Here we present biophysical analysis together with three structures of Arabidopsis thaliana PIN8: two outward-facing conformations with and without auxin, and one inward-facing conformation bound to the herbicide naphthylphthalamic acid. The structure forms a homodimer, with each monomer divided into a transport and scaffold domain with a clearly defined auxin binding site. Next to the binding site, a proline-proline crossover is a pivot point for structural changes associated with transport, which we show to be independent of proton and ion gradients and probably driven by the negative charge of the auxin. The structures and biochemical data reveal an elevator-type transport mechanism reminiscent of bile acid/sodium symporters, bicarbonate/sodium symporters and sodium/proton antiporters. Our results provide a comprehensive molecular model for auxin recognition and transport by PINs, link and expand on a well-known conceptual framework for transport, and explain a central mechanism of polar auxin transport, a core feature of plant physiology, growth and development., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
31. Kardar-Parisi-Zhang universality in a one-dimensional polariton condensate.
- Author
-
Fontaine Q, Squizzato D, Baboux F, Amelio I, Lemaître A, Morassi M, Sagnes I, Le Gratiet L, Harouri A, Wouters M, Carusotto I, Amo A, Richard M, Minguzzi A, Canet L, Ravets S, and Bloch J
- Abstract
Revealing universal behaviours is a hallmark of statistical physics. Phenomena such as the stochastic growth of crystalline surfaces
1 and of interfaces in bacterial colonies2 , and spin transport in quantum magnets3-6 all belong to the same universality class, despite the great plurality of physical mechanisms they involve at the microscopic level. More specifically, in all these systems, space-time correlations show power-law scalings characterized by universal critical exponents. This universality stems from a common underlying effective dynamics governed by the nonlinear stochastic Kardar-Parisi-Zhang (KPZ) equation7 . Recent theoretical works have suggested that this dynamics also emerges in the phase of out-of-equilibrium systems showing macroscopic spontaneous coherence8-17 . Here we experimentally demonstrate that the evolution of the phase in a driven-dissipative one-dimensional polariton condensate falls in the KPZ universality class. Our demonstration relies on a direct measurement of KPZ space-time scaling laws18,19 , combined with a theoretical analysis that reveals other key signatures of this universality class. Our results highlight fundamental physical differences between out-of-equilibrium condensates and their equilibrium counterparts, and open a paradigm for exploring universal behaviours in driven open quantum systems., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
32. Increased memory B cell potency and breadth after a SARS-CoV-2 mRNA boost.
- Author
-
Muecksch F, Wang Z, Cho A, Gaebler C, Ben Tanfous T, DaSilva J, Bednarski E, Ramos V, Zong S, Johnson B, Raspe R, Schaefer-Babajew D, Shimeliovich I, Daga M, Yao KH, Schmidt F, Millard KG, Turroja M, Jankovic M, Oliveira TY, Gazumyan A, Caskey M, Hatziioannou T, Bieniasz PD, and Nussenzweig MC
- Subjects
- Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Humans, RNA, Messenger genetics, COVID-19 immunology, COVID-19 prevention & control, COVID-19 virology, COVID-19 Vaccines administration & dosage, COVID-19 Vaccines immunology, Immunization, Secondary, Memory B Cells immunology, SARS-CoV-2 genetics, SARS-CoV-2 immunology, mRNA Vaccines administration & dosage, mRNA Vaccines immunology
- Abstract
The Omicron variant of SARS-CoV-2 infected many vaccinated and convalescent individuals
1-3 . Despite the reduced protection from infection, individuals who received three doses of an mRNA vaccine were highly protected from more serious consequences of infection4 . Here we examine the memory B cell repertoire in a longitudinal cohort of individuals receiving three mRNA vaccine doses5,6 . We find that the third dose is accompanied by an increase in, and evolution of, receptor-binding domain (RBD)-specific memory B cells. The increase is due to expansion of memory B cell clones that were present after the second dose as well as the emergence of new clones. The antibodies encoded by these cells showed significantly increased potency and breadth when compared with antibodies obtained after the second dose. Notably, the increase in potency was especially evident among newly developing clones of memory cells, which differed from persisting clones in targeting more conserved regions of the RBD. Overall, more than 50% of the analysed neutralizing antibodies in the memory compartment after the third mRNA vaccine dose neutralized the Omicron variant. Thus, individuals receiving three doses of an mRNA vaccine have a diverse memory B cell repertoire that can respond rapidly and produce antibodies capable of clearing even diversified variants such as Omicron. These data help to explain why a third dose of a vaccine that was not specifically designed to protect against variants is effective against variant-induced serious disease., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
33. Grey wolf genomic history reveals a dual ancestry of dogs.
- Author
-
Bergström A, Stanton DWG, Taron UH, Frantz L, Sinding MS, Ersmark E, Pfrengle S, Cassatt-Johnstone M, Lebrasseur O, Girdland-Flink L, Fernandes DM, Ollivier M, Speidel L, Gopalakrishnan S, Westbury MV, Ramos-Madrigal J, Feuerborn TR, Reiter E, Gretzinger J, Münzel SC, Swali P, Conard NJ, Carøe C, Haile J, Linderholm A, Androsov S, Barnes I, Baumann C, Benecke N, Bocherens H, Brace S, Carden RF, Drucker DG, Fedorov S, Gasparik M, Germonpré M, Grigoriev S, Groves P, Hertwig ST, Ivanova VV, Janssens L, Jennings RP, Kasparov AK, Kirillova IV, Kurmaniyazov I, Kuzmin YV, Kosintsev PA, Lázničková-Galetová M, Leduc C, Nikolskiy P, Nussbaumer M, O'Drisceoil C, Orlando L, Outram A, Pavlova EY, Perri AR, Pilot M, Pitulko VV, Plotnikov VV, Protopopov AV, Rehazek A, Sablin M, Seguin-Orlando A, Storå J, Verjux C, Zaibert VF, Zazula G, Crombé P, Hansen AJ, Willerslev E, Leonard JA, Götherström A, Pinhasi R, Schuenemann VJ, Hofreiter M, Gilbert MTP, Shapiro B, Larson G, Krause J, Dalén L, and Skoglund P
- Subjects
- Africa, Animals, DNA, Ancient analysis, Domestication, Europe, History, Ancient, Middle East, Mutation, North America, Selection, Genetic, Siberia, Tumor Suppressor Proteins genetics, Dogs genetics, Genome genetics, Genomics, Phylogeny, Wolves classification, Wolves genetics
- Abstract
The grey wolf (Canis lupus) was the first species to give rise to a domestic population, and they remained widespread throughout the last Ice Age when many other large mammal species went extinct. Little is known, however, about the history and possible extinction of past wolf populations or when and where the wolf progenitors of the present-day dog lineage (Canis familiaris) lived
1-8 . Here we analysed 72 ancient wolf genomes spanning the last 100,000 years from Europe, Siberia and North America. We found that wolf populations were highly connected throughout the Late Pleistocene, with levels of differentiation an order of magnitude lower than they are today. This population connectivity allowed us to detect natural selection across the time series, including rapid fixation of mutations in the gene IFT88 40,000-30,000 years ago. We show that dogs are overall more closely related to ancient wolves from eastern Eurasia than to those from western Eurasia, suggesting a domestication process in the east. However, we also found that dogs in the Near East and Africa derive up to half of their ancestry from a distinct population related to modern southwest Eurasian wolves, reflecting either an independent domestication process or admixture from local wolves. None of the analysed ancient wolf genomes is a direct match for either of these dog ancestries, meaning that the exact progenitor populations remain to be located., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
34. Prolonged viral suppression with anti-HIV-1 antibody therapy.
- Author
-
Gaebler C, Nogueira L, Stoffel E, Oliveira TY, Breton G, Millard KG, Turroja M, Butler A, Ramos V, Seaman MS, Reeves JD, Petroupoulos CJ, Shimeliovich I, Gazumyan A, Jiang CS, Jilg N, Scheid JF, Gandhi R, Walker BD, Sneller MC, Fauci A, Chun TW, Caskey M, and Nussenzweig MC
- Subjects
- CD4-Positive T-Lymphocytes virology, Humans, Proviruses drug effects, Viremia drug therapy, Virus Latency drug effects, Anti-Retroviral Agents therapeutic use, HIV Antibodies therapeutic use, HIV Infections drug therapy, HIV-1 drug effects, HIV-1 growth & development, Viral Load drug effects
- Abstract
HIV-1 infection remains a public health problem with no cure. Anti-retroviral therapy (ART) is effective but requires lifelong drug administration owing to a stable reservoir of latent proviruses integrated into the genome of CD4
+ T cells1 . Immunotherapy with anti-HIV-1 antibodies has the potential to suppress infection and increase the rate of clearance of infected cells2,3 . Here we report on a clinical study in which people living with HIV received seven doses of a combination of two broadly neutralizing antibodies over 20 weeks in the presence or absence of ART. Without pre-screening for antibody sensitivity, 76% (13 out of 17) of the volunteers maintained virologic suppression for at least 20 weeks off ART. Post hoc sensitivity analyses were not predictive of the time to viral rebound. Individuals in whom virus remained suppressed for more than 20 weeks showed rebound viraemia after one of the antibodies reached serum concentrations below 10 µg ml-1 . Two of the individuals who received all seven antibody doses maintained suppression after one year. Reservoir analysis performed after six months of antibody therapy revealed changes in the size and composition of the intact proviral reservoir. By contrast, there was no measurable decrease in the defective reservoir in the same individuals. These data suggest that antibody administration affects the HIV-1 reservoir, but additional larger and longer studies will be required to define the precise effect of antibody immunotherapy on the reservoir., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
35. Differential cofactor dependencies define distinct types of human enhancers.
- Author
-
Neumayr C, Haberle V, Serebreni L, Karner K, Hendy O, Boija A, Henninger JE, Li CH, Stejskal K, Lin G, Bergauer K, Pagani M, Rath M, Mechtler K, Arnold CD, and Stark A
- Subjects
- Cell Cycle Proteins metabolism, Chromatin genetics, Humans, Nuclear Proteins metabolism, Tumor Suppressor Protein p53 metabolism, Enhancer Elements, Genetic genetics, Transcription Factors metabolism
- Abstract
All multicellular organisms rely on differential gene transcription regulated by genomic enhancers, which function through cofactors that are recruited by transcription factors
1,2 . Emerging evidence suggests that not all cofactors are required at all enhancers3-5 , yet whether these observations reflect more general principles or distinct types of enhancers remained unknown. Here we categorized human enhancers by their cofactor dependencies and show that these categories provide a framework to understand the sequence and chromatin diversity of enhancers and their roles in different gene-regulatory programmes. We quantified enhancer activities along the entire human genome using STARR-seq6 in HCT116 cells, following the rapid degradation of eight cofactors. This analysis identified different types of enhancers with distinct cofactor requirements, sequences and chromatin properties. Some enhancers were insensitive to the depletion of the core Mediator subunit MED14 or the bromodomain protein BRD4 and regulated distinct transcriptional programmes. In particular, canonical Mediator7 seemed dispensable for P53-responsive enhancers, and MED14-depleted cells induced endogenous P53 target genes. Similarly, BRD4 was not required for the transcription of genes that bear CCAAT boxes and a TATA box (including histone genes and LTR12 retrotransposons) or for the induction of heat-shock genes. This categorization of enhancers through cofactor dependencies reveals distinct enhancer types that can bypass broadly utilized cofactors, which illustrates how alternative ways to activate transcription separate gene expression programmes and provide a conceptual framework to understand enhancer function and regulatory specificity., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
36. Intermittent PI3Kδ inhibition sustains anti-tumour immunity and curbs irAEs.
- Author
-
Eschweiler S, Ramírez-Suástegui C, Li Y, King E, Chudley L, Thomas J, Wood O, von Witzleben A, Jeffrey D, McCann K, Simon H, Mondal M, Wang A, Dicker M, Lopez-Guadamillas E, Chou TF, Dobbs NA, Essame L, Acton G, Kelly F, Halbert G, Sacco JJ, Schache AG, Shaw R, McCaul JA, Paterson C, Davies JH, Brennan PA, Singh RP, Loadman PM, Wilson W, Hackshaw A, Seumois G, Okkenhaug K, Thomas GJ, Jones TM, Ay F, Friberg G, Kronenberg M, Vanhaesebroeck B, Vijayanand P, and Ottensmeier CH
- Subjects
- Adenosine therapeutic use, Animals, Disease Models, Animal, Humans, Immunotherapy, Mice, Phosphatidylinositol 3-Kinases, Quinolines therapeutic use, T-Lymphocytes, Regulatory, Antineoplastic Agents therapeutic use, Head and Neck Neoplasms drug therapy
- Abstract
Phosphoinositide 3-kinase δ (PI3Kδ) has a key role in lymphocytes, and inhibitors that target this PI3K have been approved for treatment of B cell malignancies
1-3 . Although studies in mouse models of solid tumours have demonstrated that PI3Kδ inhibitors (PI3Kδi) can induce anti-tumour immunity4,5 , its effect on solid tumours in humans remains unclear. Here we assessed the effects of the PI3Kδi AMG319 in human patients with head and neck cancer in a neoadjuvant, double-blind, placebo-controlled randomized phase II trial (EudraCT no. 2014-004388-20). PI3Kδ inhibition decreased the number of tumour-infiltrating regulatory T (Treg ) cells and enhanced the cytotoxic potential of tumour-infiltrating T cells. At the tested doses of AMG319, immune-related adverse events (irAEs) required treatment to be discontinued in 12 out of 21 of patients treated with AMG319, suggestive of systemic effects on Treg cells. Accordingly, in mouse models, PI3Kδi decreased the number of Treg cells systemically and caused colitis. Single-cell RNA-sequencing analysis revealed a PI3Kδi-driven loss of tissue-resident colonic ST2 Treg cells, accompanied by expansion of pathogenic T helper 17 (TH 17) and type 17 CD8+ T (TC 17) cells, which probably contributed to toxicity; this points towards a specific mode of action for the emergence of irAEs. A modified treatment regimen with intermittent dosing of PI3Kδi in mouse models led to a significant decrease in tumour growth without inducing pathogenic T cells in colonic tissue, indicating that alternative dosing regimens might limit toxicity., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
37. TDP-43 loss and ALS-risk SNPs drive mis-splicing and depletion of UNC13A.
- Author
-
Brown AL, Wilkins OG, Keuss MJ, Hill SE, Zanovello M, Lee WC, Bampton A, Lee FCY, Masino L, Qi YA, Bryce-Smith S, Gatt A, Hallegger M, Fagegaltier D, Phatnani H, Newcombe J, Gustavsson EK, Seddighi S, Reyes JF, Coon SL, Ramos D, Schiavo G, Fisher EMC, Raj T, Secrier M, Lashley T, Ule J, Buratti E, Humphrey J, Ward ME, and Fratta P
- Subjects
- Alternative Splicing, Codon, Nonsense, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Humans, Nerve Tissue Proteins, Polymorphism, Single Nucleotide genetics, Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis metabolism, Frontotemporal Dementia genetics, Frontotemporal Dementia metabolism, TDP-43 Proteinopathies
- Abstract
Variants of UNC13A, a critical gene for synapse function, increase the risk of amyotrophic lateral sclerosis and frontotemporal dementia
1-3 , two related neurodegenerative diseases defined by mislocalization of the RNA-binding protein TDP-434,5 . Here we show that TDP-43 depletion induces robust inclusion of a cryptic exon in UNC13A, resulting in nonsense-mediated decay and loss of UNC13A protein. Two common intronic UNC13A polymorphisms strongly associated with amyotrophic lateral sclerosis and frontotemporal dementia risk overlap with TDP-43 binding sites. These polymorphisms potentiate cryptic exon inclusion, both in cultured cells and in brains and spinal cords from patients with these conditions. Our findings, which demonstrate a genetic link between loss of nuclear TDP-43 function and disease, reveal the mechanism by which UNC13A variants exacerbate the effects of decreased TDP-43 function. They further provide a promising therapeutic target for TDP-43 proteinopathies., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
38. Autism genes converge on asynchronous development of shared neuron classes.
- Author
-
Paulsen B, Velasco S, Kedaigle AJ, Pigoni M, Quadrato G, Deo AJ, Adiconis X, Uzquiano A, Sartore R, Yang SM, Simmons SK, Symvoulidis P, Kim K, Tsafou K, Podury A, Abbate C, Tucewicz A, Smith SN, Albanese A, Barrett L, Sanjana NE, Shi X, Chung K, Lage K, Boyden ES, Regev A, Levin JZ, and Arlotta P
- Subjects
- Cerebral Cortex cytology, DNA-Binding Proteins genetics, GABAergic Neurons metabolism, GABAergic Neurons pathology, Histone-Lysine N-Methyltransferase genetics, Humans, Organoids cytology, Proteomics, RNA-Seq, Single-Cell Analysis, Transcription Factors genetics, Autism Spectrum Disorder genetics, Autism Spectrum Disorder metabolism, Autism Spectrum Disorder pathology, Genetic Predisposition to Disease, Neurons classification, Neurons metabolism, Neurons pathology
- Abstract
Genetic risk for autism spectrum disorder (ASD) is associated with hundreds of genes spanning a wide range of biological functions
1-6 . The alterations in the human brain resulting from mutations in these genes remain unclear. Furthermore, their phenotypic manifestation varies across individuals7,8 . Here we used organoid models of the human cerebral cortex to identify cell-type-specific developmental abnormalities that result from haploinsufficiency in three ASD risk genes-SUV420H1 (also known as KMT5B), ARID1B and CHD8-in multiple cell lines from different donors, using single-cell RNA-sequencing (scRNA-seq) analysis of more than 745,000 cells and proteomic analysis of individual organoids, to identify phenotypic convergence. Each of the three mutations confers asynchronous development of two main cortical neuronal lineages-γ-aminobutyric-acid-releasing (GABAergic) neurons and deep-layer excitatory projection neurons-but acts through largely distinct molecular pathways. Although these phenotypes are consistent across cell lines, their expressivity is influenced by the individual genomic context, in a manner that is dependent on both the risk gene and the developmental defect. Calcium imaging in intact organoids shows that these early-stage developmental changes are followed by abnormal circuit activity. This research uncovers cell-type-specific neurodevelopmental abnormalities that are shared across ASD risk genes and are finely modulated by human genomic context, finding convergence in the neurobiological basis of how different risk genes contribute to ASD pathology., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
39. Anti-SARS-CoV-2 receptor-binding domain antibody evolution after mRNA vaccination.
- Author
-
Cho A, Muecksch F, Schaefer-Babajew D, Wang Z, Finkin S, Gaebler C, Ramos V, Cipolla M, Mendoza P, Agudelo M, Bednarski E, DaSilva J, Shimeliovich I, Dizon J, Daga M, Millard KG, Turroja M, Schmidt F, Zhang F, Tanfous TB, Jankovic M, Oliveria TY, Gazumyan A, Caskey M, Bieniasz PD, Hatziioannou T, and Nussenzweig MC
- Subjects
- 2019-nCoV Vaccine mRNA-1273 immunology, Adult, Aged, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Antibody Affinity, BNT162 Vaccine immunology, Cohort Studies, Cross Reactions, Enzyme-Linked Immunosorbent Assay, Epitopes, B-Lymphocyte immunology, Female, Humans, Male, Memory B Cells immunology, Middle Aged, Neutralization Tests, Protein Domains immunology, Spike Glycoprotein, Coronavirus chemistry, Young Adult, COVID-19 Vaccines immunology, Evolution, Molecular, Spike Glycoprotein, Coronavirus immunology, Vaccines, Synthetic immunology, mRNA Vaccines immunology
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection produces B cell responses that continue to evolve for at least a year. During that time, memory B cells express increasingly broad and potent antibodies that are resistant to mutations found in variants of concern
1 . As a result, vaccination of coronavirus disease 2019 (COVID-19) convalescent individuals with currently available mRNA vaccines produces high levels of plasma neutralizing activity against all variants tested1,2 . Here we examine memory B cell evolution five months after vaccination with either Moderna (mRNA-1273) or Pfizer-BioNTech (BNT162b2) mRNA vaccine in a cohort of SARS-CoV-2-naive individuals. Between prime and boost, memory B cells produce antibodies that evolve increased neutralizing activity, but there is no further increase in potency or breadth thereafter. Instead, memory B cells that emerge five months after vaccination of naive individuals express antibodies that are similar to those that dominate the initial response. While individual memory antibodies selected over time by natural infection have greater potency and breadth than antibodies elicited by vaccination, the overall neutralizing potency of plasma is greater following vaccination. These results suggest that boosting vaccinated individuals with currently available mRNA vaccines will increase plasma neutralizing activity but may not produce antibodies with equivalent breadth to those obtained by vaccinating convalescent individuals., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
40. Are female science leaders judged more harshly than men? Study it.
- Author
-
Schraudner M and Pollitzer E
- Published
- 2021
- Full Text
- View/download PDF
41. Bioaccumulation of therapeutic drugs by human gut bacteria.
- Author
-
Klünemann M, Andrejev S, Blasche S, Mateus A, Phapale P, Devendran S, Vappiani J, Simon B, Scott TA, Kafkia E, Konstantinidis D, Zirngibl K, Mastrorilli E, Banzhaf M, Mackmull MT, Hövelmann F, Nesme L, Brochado AR, Maier L, Bock T, Periwal V, Kumar M, Kim Y, Tramontano M, Schultz C, Beck M, Hennig J, Zimmermann M, Sévin DC, Cabreiro F, Savitski MM, Bork P, Typas A, and Patil KR
- Subjects
- Animals, Antidepressive Agents metabolism, Antidepressive Agents pharmacokinetics, Caenorhabditis elegans metabolism, Cells metabolism, Click Chemistry, Duloxetine Hydrochloride adverse effects, Duloxetine Hydrochloride pharmacokinetics, Humans, Metabolomics, Models, Animal, Proteomics, Reproducibility of Results, Bacteria metabolism, Bioaccumulation, Duloxetine Hydrochloride metabolism, Gastrointestinal Microbiome physiology
- Abstract
Bacteria in the gut can modulate the availability and efficacy of therapeutic drugs. However, the systematic mapping of the interactions between drugs and bacteria has only started recently
1 and the main underlying mechanism proposed is the chemical transformation of drugs by microorganisms (biotransformation). Here we investigated the depletion of 15 structurally diverse drugs by 25 representative strains of gut bacteria. This revealed 70 bacteria-drug interactions, 29 of which had not to our knowledge been reported before. Over half of the new interactions can be ascribed to bioaccumulation; that is, bacteria storing the drug intracellularly without chemically modifying it, and in most cases without the growth of the bacteria being affected. As a case in point, we studied the molecular basis of bioaccumulation of the widely used antidepressant duloxetine by using click chemistry, thermal proteome profiling and metabolomics. We find that duloxetine binds to several metabolic enzymes and changes the metabolite secretion of the respective bacteria. When tested in a defined microbial community of accumulators and non-accumulators, duloxetine markedly altered the composition of the community through metabolic cross-feeding. We further validated our findings in an animal model, showing that bioaccumulating bacteria attenuate the behavioural response of Caenorhabditis elegans to duloxetine. Together, our results show that bioaccumulation by gut bacteria may be a common mechanism that alters drug availability and bacterial metabolism, with implications for microbiota composition, pharmacokinetics, side effects and drug responses, probably in an individual manner., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
42. A lymphocyte-microglia-astrocyte axis in chronic active multiple sclerosis.
- Author
-
Absinta M, Maric D, Gharagozloo M, Garton T, Smith MD, Jin J, Fitzgerald KC, Song A, Liu P, Lin JP, Wu T, Johnson KR, McGavern DB, Schafer DP, Calabresi PA, and Reich DS
- Subjects
- Animals, Brain pathology, Complement C1q antagonists & inhibitors, Complement C1q metabolism, Encephalomyelitis, Autoimmune, Experimental pathology, Female, Humans, Inflammation pathology, Magnetic Resonance Imaging, Male, Mice, Mice, Inbred C57BL, Middle Aged, Multiple Sclerosis diagnostic imaging, RNA-Seq, Transcriptome, White Matter pathology, Astrocytes pathology, Lymphocytes pathology, Microglia pathology, Multiple Sclerosis pathology
- Abstract
Multiple sclerosis (MS) lesions that do not resolve in the months after they form harbour ongoing demyelination and axon degeneration, and are identifiable in vivo by their paramagnetic rims on MRI scans
1-3 . Here, to define mechanisms underlying this disabling, progressive neurodegenerative state4-6 and foster development of new therapeutic agents, we used MRI-informed single-nucleus RNA sequencing to profile the edge of demyelinated white matter lesions at various stages of inflammation. We uncovered notable glial and immune cell diversity, especially at the chronically inflamed lesion edge. We define 'microglia inflamed in MS' (MIMS) and 'astrocytes inflamed in MS', glial phenotypes that demonstrate neurodegenerative programming. The MIMS transcriptional profile overlaps with that of microglia in other neurodegenerative diseases, suggesting that primary and secondary neurodegeneration share common mechanisms and could benefit from similar therapeutic approaches. We identify complement component 1q (C1q) as a critical mediator of MIMS activation, validated immunohistochemically in MS tissue, genetically by microglia-specific C1q ablation in mice with experimental autoimmune encephalomyelitis, and therapeutically by treating chronic experimental autoimmune encephalomyelitis with C1q blockade. C1q inhibition is a potential therapeutic avenue to address chronic white matter inflammation, which could be monitored by longitudinal assessment of its dynamic biomarker, paramagnetic rim lesions, using advanced MRI methods., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2021
- Full Text
- View/download PDF
43. Broad sarbecovirus neutralization by a human monoclonal antibody.
- Author
-
Tortorici MA, Czudnochowski N, Starr TN, Marzi R, Walls AC, Zatta F, Bowen JE, Jaconi S, Di Iulio J, Wang Z, De Marco A, Zepeda SK, Pinto D, Liu Z, Beltramello M, Bartha I, Housley MP, Lempp FA, Rosen LE, Dellota E Jr, Kaiser H, Montiel-Ruiz M, Zhou J, Addetia A, Guarino B, Culap K, Sprugasci N, Saliba C, Vetti E, Giacchetto-Sasselli I, Fregni CS, Abdelnabi R, Foo SC, Havenar-Daughton C, Schmid MA, Benigni F, Cameroni E, Neyts J, Telenti A, Virgin HW, Whelan SPJ, Snell G, Bloom JD, Corti D, Veesler D, and Pizzuto MS
- Subjects
- Animals, Antibodies, Monoclonal chemistry, Antibodies, Viral chemistry, Antibodies, Viral therapeutic use, Broadly Neutralizing Antibodies chemistry, COVID-19 immunology, COVID-19 virology, Cross Reactions immunology, Disease Models, Animal, Female, Humans, Immune Evasion genetics, Immune Evasion immunology, Mesocricetus immunology, Mesocricetus virology, Mutation, Neutralization Tests, SARS-CoV-2 chemistry, SARS-CoV-2 genetics, Viral Zoonoses immunology, Viral Zoonoses prevention & control, Viral Zoonoses virology, Antibodies, Monoclonal immunology, Antibodies, Monoclonal therapeutic use, Antibodies, Viral immunology, Broadly Neutralizing Antibodies immunology, Broadly Neutralizing Antibodies therapeutic use, COVID-19 prevention & control, SARS-CoV-2 classification, SARS-CoV-2 immunology
- Abstract
The recent emergence of SARS-CoV-2 variants of concern
1-10 and the recurrent spillovers of coronaviruses11,12 into the human population highlight the need for broadly neutralizing antibodies that are not affected by the ongoing antigenic drift and that can prevent or treat future zoonotic infections. Here we describe a human monoclonal antibody designated S2X259, which recognizes a highly conserved cryptic epitope of the receptor-binding domain and cross-reacts with spikes from all clades of sarbecovirus. S2X259 broadly neutralizes spike-mediated cell entry of SARS-CoV-2, including variants of concern (B.1.1.7, B.1.351, P.1, and B.1.427/B.1.429), as well as a wide spectrum of human and potentially zoonotic sarbecoviruses through inhibition of angiotensin-converting enzyme 2 (ACE2) binding to the receptor-binding domain. Furthermore, deep-mutational scanning and in vitro escape selection experiments demonstrate that S2X259 possesses an escape profile that is limited to a single substitution, G504D. We show that prophylactic and therapeutic administration of S2X259 protects Syrian hamsters (Mesocricetus auratus) against challenge with the prototypic SARS-CoV-2 and the B.1.351 variant of concern, which suggests that this monoclonal antibody is a promising candidate for the prevention and treatment of emergent variants and zoonotic infections. Our data reveal a key antigenic site that is targeted by broadly neutralizing antibodies and will guide the design of vaccines that are effective against all sarbecoviruses., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
44. SARS-CoV-2 RBD antibodies that maximize breadth and resistance to escape.
- Author
-
Starr TN, Czudnochowski N, Liu Z, Zatta F, Park YJ, Addetia A, Pinto D, Beltramello M, Hernandez P, Greaney AJ, Marzi R, Glass WG, Zhang I, Dingens AS, Bowen JE, Tortorici MA, Walls AC, Wojcechowskyj JA, De Marco A, Rosen LE, Zhou J, Montiel-Ruiz M, Kaiser H, Dillen JR, Tucker H, Bassi J, Silacci-Fregni C, Housley MP, di Iulio J, Lombardo G, Agostini M, Sprugasci N, Culap K, Jaconi S, Meury M, Dellota E Jr, Abdelnabi R, Foo SC, Cameroni E, Stumpf S, Croll TI, Nix JC, Havenar-Daughton C, Piccoli L, Benigni F, Neyts J, Telenti A, Lempp FA, Pizzuto MS, Chodera JD, Hebner CM, Virgin HW, Whelan SPJ, Veesler D, Corti D, Bloom JD, and Snell G
- Subjects
- Adult, Aged, Animals, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal immunology, Antibodies, Viral chemistry, Antibodies, Viral immunology, Antibody Affinity, Broadly Neutralizing Antibodies chemistry, COVID-19 immunology, COVID-19 Vaccines chemistry, COVID-19 Vaccines immunology, Cell Line, Cricetinae, Epitopes, B-Lymphocyte chemistry, Epitopes, B-Lymphocyte genetics, Epitopes, B-Lymphocyte immunology, Female, Humans, Male, Mesocricetus, Middle Aged, Models, Molecular, SARS-CoV-2 chemistry, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics, Vaccinology, COVID-19 Drug Treatment, Broadly Neutralizing Antibodies immunology, COVID-19 virology, Cross Reactions immunology, Immune Evasion genetics, Immune Evasion immunology, SARS-CoV-2 classification, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus immunology
- Abstract
An ideal therapeutic anti-SARS-CoV-2 antibody would resist viral escape
1-3 , have activity against diverse sarbecoviruses4-7 , and be highly protective through viral neutralization8-11 and effector functions12,13 . Understanding how these properties relate to each other and vary across epitopes would aid the development of therapeutic antibodies and guide vaccine design. Here we comprehensively characterize escape, breadth and potency across a panel of SARS-CoV-2 antibodies targeting the receptor-binding domain (RBD). Despite a trade-off between in vitro neutralization potency and breadth of sarbecovirus binding, we identify neutralizing antibodies with exceptional sarbecovirus breadth and a corresponding resistance to SARS-CoV-2 escape. One of these antibodies, S2H97, binds with high affinity across all sarbecovirus clades to a cryptic epitope and prophylactically protects hamsters from viral challenge. Antibodies that target the angiotensin-converting enzyme 2 (ACE2) receptor-binding motif (RBM) typically have poor breadth and are readily escaped by mutations despite high neutralization potency. Nevertheless, we also characterize a potent RBM antibody (S2E128 ) with breadth across sarbecoviruses related to SARS-CoV-2 and a high barrier to viral escape. These data highlight principles underlying variation in escape, breadth and potency among antibodies that target the RBD, and identify epitopes and features to prioritize for therapeutic development against the current and potential future pandemics., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
45. Genetic insights into biological mechanisms governing human ovarian ageing.
- Author
-
Ruth KS, Day FR, Hussain J, Martínez-Marchal A, Aiken CE, Azad A, Thompson DJ, Knoblochova L, Abe H, Tarry-Adkins JL, Gonzalez JM, Fontanillas P, Claringbould A, Bakker OB, Sulem P, Walters RG, Terao C, Turon S, Horikoshi M, Lin K, Onland-Moret NC, Sankar A, Hertz EPT, Timshel PN, Shukla V, Borup R, Olsen KW, Aguilera P, Ferrer-Roda M, Huang Y, Stankovic S, Timmers PRHJ, Ahearn TU, Alizadeh BZ, Naderi E, Andrulis IL, Arnold AM, Aronson KJ, Augustinsson A, Bandinelli S, Barbieri CM, Beaumont RN, Becher H, Beckmann MW, Benonisdottir S, Bergmann S, Bochud M, Boerwinkle E, Bojesen SE, Bolla MK, Boomsma DI, Bowker N, Brody JA, Broer L, Buring JE, Campbell A, Campbell H, Castelao JE, Catamo E, Chanock SJ, Chenevix-Trench G, Ciullo M, Corre T, Couch FJ, Cox A, Crisponi L, Cross SS, Cucca F, Czene K, Smith GD, de Geus EJCN, de Mutsert R, De Vivo I, Demerath EW, Dennis J, Dunning AM, Dwek M, Eriksson M, Esko T, Fasching PA, Faul JD, Ferrucci L, Franceschini N, Frayling TM, Gago-Dominguez M, Mezzavilla M, García-Closas M, Gieger C, Giles GG, Grallert H, Gudbjartsson DF, Gudnason V, Guénel P, Haiman CA, Håkansson N, Hall P, Hayward C, He C, He W, Heiss G, Høffding MK, Hopper JL, Hottenga JJ, Hu F, Hunter D, Ikram MA, Jackson RD, Joaquim MDR, John EM, Joshi PK, Karasik D, Kardia SLR, Kartsonaki C, Karlsson R, Kitahara CM, Kolcic I, Kooperberg C, Kraft P, Kurian AW, Kutalik Z, La Bianca M, LaChance G, Langenberg C, Launer LJ, Laven JSE, Lawlor DA, Le Marchand L, Li J, Lindblom A, Lindstrom S, Lindstrom T, Linet M, Liu Y, Liu S, Luan J, Mägi R, Magnusson PKE, Mangino M, Mannermaa A, Marco B, Marten J, Martin NG, Mbarek H, McKnight B, Medland SE, Meisinger C, Meitinger T, Menni C, Metspalu A, Milani L, Milne RL, Montgomery GW, Mook-Kanamori DO, Mulas A, Mulligan AM, Murray A, Nalls MA, Newman A, Noordam R, Nutile T, Nyholt DR, Olshan AF, Olsson H, Painter JN, Patel AV, Pedersen NL, Perjakova N, Peters A, Peters U, Pharoah PDP, Polasek O, Porcu E, Psaty BM, Rahman I, Rennert G, Rennert HS, Ridker PM, Ring SM, Robino A, Rose LM, Rosendaal FR, Rossouw J, Rudan I, Rueedi R, Ruggiero D, Sala CF, Saloustros E, Sandler DP, Sanna S, Sawyer EJ, Sarnowski C, Schlessinger D, Schmidt MK, Schoemaker MJ, Schraut KE, Scott C, Shekari S, Shrikhande A, Smith AV, Smith BH, Smith JA, Sorice R, Southey MC, Spector TD, Spinelli JJ, Stampfer M, Stöckl D, van Meurs JBJ, Strauch K, Styrkarsdottir U, Swerdlow AJ, Tanaka T, Teras LR, Teumer A, Þorsteinsdottir U, Timpson NJ, Toniolo D, Traglia M, Troester MA, Truong T, Tyrrell J, Uitterlinden AG, Ulivi S, Vachon CM, Vitart V, Völker U, Vollenweider P, Völzke H, Wang Q, Wareham NJ, Weinberg CR, Weir DR, Wilcox AN, van Dijk KW, Willemsen G, Wilson JF, Wolffenbuttel BHR, Wolk A, Wood AR, Zhao W, Zygmunt M, Chen Z, Li L, Franke L, Burgess S, Deelen P, Pers TH, Grøndahl ML, Andersen CY, Pujol A, Lopez-Contreras AJ, Daniel JA, Stefansson K, Chang-Claude J, van der Schouw YT, Lunetta KL, Chasman DI, Easton DF, Visser JA, Ozanne SE, Namekawa SH, Solc P, Murabito JM, Ong KK, Hoffmann ER, Murray A, Roig I, and Perry JRB
- Subjects
- Adult, Alleles, Animals, Bone and Bones metabolism, Checkpoint Kinase 1 genetics, Checkpoint Kinase 2 genetics, Diabetes Mellitus, Type 2, Diet, Europe ethnology, Asia, Eastern ethnology, Female, Fertility genetics, Fragile X Mental Retardation Protein genetics, Genetic Predisposition to Disease, Genome-Wide Association Study, Healthy Aging genetics, Humans, Longevity genetics, Menopause genetics, Menopause, Premature genetics, Mice, Mice, Inbred C57BL, Middle Aged, Primary Ovarian Insufficiency genetics, Uterus, Aging genetics, Ovary metabolism
- Abstract
Reproductive longevity is essential for fertility and influences healthy ageing in women
1,2 , but insights into its underlying biological mechanisms and treatments to preserve it are limited. Here we identify 290 genetic determinants of ovarian ageing, assessed using normal variation in age at natural menopause (ANM) in about 200,000 women of European ancestry. These common alleles were associated with clinical extremes of ANM; women in the top 1% of genetic susceptibility have an equivalent risk of premature ovarian insufficiency to those carrying monogenic FMR1 premutations3 . The identified loci implicate a broad range of DNA damage response (DDR) processes and include loss-of-function variants in key DDR-associated genes. Integration with experimental models demonstrates that these DDR processes act across the life-course to shape the ovarian reserve and its rate of depletion. Furthermore, we demonstrate that experimental manipulation of DDR pathways highlighted by human genetics increases fertility and extends reproductive life in mice. Causal inference analyses using the identified genetic variants indicate that extending reproductive life in women improves bone health and reduces risk of type 2 diabetes, but increases the risk of hormone-sensitive cancers. These findings provide insight into the mechanisms that govern ovarian ageing, when they act, and how they might be targeted by therapeutic approaches to extend fertility and prevent disease., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
46. Naturally enhanced neutralizing breadth against SARS-CoV-2 one year after infection.
- Author
-
Wang Z, Muecksch F, Schaefer-Babajew D, Finkin S, Viant C, Gaebler C, Hoffmann HH, Barnes CO, Cipolla M, Ramos V, Oliveira TY, Cho A, Schmidt F, Da Silva J, Bednarski E, Aguado L, Yee J, Daga M, Turroja M, Millard KG, Jankovic M, Gazumyan A, Zhao Z, Rice CM, Bieniasz PD, Caskey M, Hatziioannou T, and Nussenzweig MC
- Subjects
- Adult, Aged, Antibodies, Monoclonal immunology, B-Lymphocytes immunology, Enzyme-Linked Immunosorbent Assay, Epitopes immunology, Female, Humans, Immunologic Memory immunology, Male, Middle Aged, SARS-CoV-2 chemistry, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus immunology, Time Factors, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral immunology, COVID-19 blood, COVID-19 immunology, SARS-CoV-2 immunology
- Abstract
More than one year after its inception, the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains difficult to control despite the availability of several working vaccines. Progress in controlling the pandemic is slowed by the emergence of variants that appear to be more transmissible and more resistant to antibodies
1,2 . Here we report on a cohort of 63 individuals who have recovered from COVID-19 assessed at 1.3, 6.2 and 12 months after SARS-CoV-2 infection, 41% of whom also received mRNA vaccines3,4 . In the absence of vaccination, antibody reactivity to the receptor binding domain (RBD) of SARS-CoV-2, neutralizing activity and the number of RBD-specific memory B cells remain relatively stable between 6 and 12 months after infection. Vaccination increases all components of the humoral response and, as expected, results in serum neutralizing activities against variants of concern similar to or greater than the neutralizing activity against the original Wuhan Hu-1 strain achieved by vaccination of naive individuals2,5-8 . The mechanism underlying these broad-based responses involves ongoing antibody somatic mutation, memory B cell clonal turnover and development of monoclonal antibodies that are exceptionally resistant to SARS-CoV-2 RBD mutations, including those found in the variants of concern4,9 . In addition, B cell clones expressing broad and potent antibodies are selectively retained in the repertoire over time and expand markedly after vaccination. The data suggest that immunity in convalescent individuals will be very long lasting and that convalescent individuals who receive available mRNA vaccines will produce antibodies and memory B cells that should be protective against circulating SARS-CoV-2 variants., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
47. Anisotropic satellite galaxy quenching modulated by black hole activity.
- Author
-
Martín-Navarro I, Pillepich A, Nelson D, Rodriguez-Gomez V, Donnari M, Hernquist L, and Springel V
- Abstract
The evolution of satellite galaxies is shaped by their constant interaction with the circumgalactic medium surrounding central galaxies, which in turn may be affected by gas and energy ejected from the central supermassive black hole
1-6 . The nature of such a coupling between black holes and galaxies is, however, much debated7-9 and observational evidence remains scarce10,11 . Here we report an analysis of archival data on 124,163 satellite galaxies in the potential wells of 29,631 dark matter halos with masses between 1012 and 1014 solar masses. We find that quenched satellite galaxies are relatively less frequent along the minor axis of their central galaxies. This observation might appear counterintuitive given that black hole activity is expected to eject mass and energy preferentially in the direction of the minor axis of the host galaxy. We show, however, that the observed anisotropic signal results precisely from the ejective nature of black hole feedback in massive halos, as outflows powered by active galactic nuclei clear out the circumgalactic medium, reducing the ram pressure and thus preserving star formation in satellite galaxies. This interpretation is supported by the IllustrisTNG suite of cosmological numerical simulations, even though the model's sub-grid implementation of black hole feedback is effectively isotropic12 .- Published
- 2021
- Full Text
- View/download PDF
48. Author Correction: Auto-aggressive CXCR6 + CD8 T cells cause liver immune pathology in NASH.
- Author
-
Dudek M, Pfister D, Donakonda S, Filpe P, Schneider A, Laschinger M, Hartmann D, Hüser N, Meiser P, Bayerl F, Inverso D, Wigger J, Sebode M, Öllinger R, Rad R, Hegenbarth S, Anton M, Guillot A, Bowman A, Heide D, Müller F, Ramadori P, Leone V, Garcia-Caceres C, Gruber T, Seifert G, Kabat AM, Mallm JP, Reider S, Effenberger M, Roth S, Billeter AT, Müller-Stich B, Pearce EJ, Koch-Nolte F, Käser R, Tilg H, Thimme R, Boettler T, Tacke F, Dufour JF, Haller D, Murray PJ, Heeren R, Zehn D, Böttcher JP, Heikenwälder M, and Knolle PA
- Published
- 2021
- Full Text
- View/download PDF
49. A biosensor for the direct visualization of auxin.
- Author
-
Herud-Sikimić O, Stiel AC, Kolb M, Shanmugaratnam S, Berendzen KW, Feldhaus C, Höcker B, and Jürgens G
- Subjects
- Arabidopsis, Binding Sites, Biological Transport, Escherichia coli Proteins, Fluorescence Resonance Energy Transfer, Gravitation, Plant Roots metabolism, Plants, Genetically Modified, Protein Engineering, Protein Structure, Secondary, Repressor Proteins, Signal Transduction, Biosensing Techniques, Indoleacetic Acids analysis
- Abstract
One of the most important regulatory small molecules in plants is indole-3-acetic acid, also known as auxin. Its dynamic redistribution has an essential role in almost every aspect of plant life, ranging from cell shape and division to organogenesis and responses to light and gravity
1,2 . So far, it has not been possible to directly determine the spatial and temporal distribution of auxin at a cellular resolution. Instead it is inferred from the visualization of irreversible processes that involve the endogenous auxin-response machinery3-7 ; however, such a system cannot detect transient changes. Here we report a genetically encoded biosensor for the quantitative in vivo visualization of auxin distribution. The sensor is based on the Escherichia coli tryptophan repressor8 , the binding pocket of which is engineered to be specific to auxin. Coupling of the auxin-binding moiety with selected fluorescent proteins enables the use of a fluorescence resonance energy transfer signal as a readout. Unlike previous systems, this sensor enables direct monitoring of the rapid uptake and clearance of auxin by individual cells and within cell compartments in planta. By responding to the graded spatial distribution along the root axis and its perturbation by transport inhibitors-as well as the rapid and reversible redistribution of endogenous auxin in response to changes in gravity vectors-our sensor enables real-time monitoring of auxin concentrations at a (sub)cellular resolution and their spatial and temporal changes during the lifespan of a plant.- Published
- 2021
- Full Text
- View/download PDF
50. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants.
- Author
-
Wang Z, Schmidt F, Weisblum Y, Muecksch F, Barnes CO, Finkin S, Schaefer-Babajew D, Cipolla M, Gaebler C, Lieberman JA, Oliveira TY, Yang Z, Abernathy ME, Huey-Tubman KE, Hurley A, Turroja M, West KA, Gordon K, Millard KG, Ramos V, Da Silva J, Xu J, Colbert RA, Patel R, Dizon J, Unson-O'Brien C, Shimeliovich I, Gazumyan A, Caskey M, Bjorkman PJ, Casellas R, Hatziioannou T, Bieniasz PD, and Nussenzweig MC
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Adult, Aged, Antibodies, Monoclonal blood, Antibodies, Monoclonal immunology, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, B-Lymphocytes immunology, BNT162 Vaccine, COVID-19 Vaccines genetics, Cryoelectron Microscopy, Epitopes, B-Lymphocyte chemistry, Epitopes, B-Lymphocyte immunology, Epitopes, B-Lymphocyte ultrastructure, Female, Humans, Immunization, Secondary, Immunoglobulin G blood, Immunoglobulin G immunology, Immunoglobulin M blood, Immunoglobulin M immunology, Immunologic Memory immunology, Male, Middle Aged, Models, Molecular, Mutation, Neutralization Tests, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus immunology, Vaccines, Synthetic genetics, mRNA Vaccines, Antibodies, Viral blood, COVID-19 immunology, COVID-19 virology, COVID-19 Vaccines immunology, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus genetics, Vaccines, Synthetic immunology
- Abstract
Here we report on the antibody and memory B cell responses of a cohort of 20 volunteers who received the Moderna (mRNA-1273) or Pfizer-BioNTech (BNT162b2) vaccine against SARS-CoV-2
1-4 . Eight weeks after the second injection of vaccine, volunteers showed high levels of IgM and IgG anti-SARS-CoV-2 spike protein (S) and receptor-binding-domain (RBD) binding titre. Moreover, the plasma neutralizing activity and relative numbers of RBD-specific memory B cells of vaccinated volunteers were equivalent to those of individuals who had recovered from natural infection5,6 . However, activity against SARS-CoV-2 variants that encode E484K-, N501Y- or K417N/E484K/N501-mutant S was reduced by a small-but significant-margin. The monoclonal antibodies elicited by the vaccines potently neutralize SARS-CoV-2, and target a number of different RBD epitopes in common with monoclonal antibodies isolated from infected donors5-8 . However, neutralization by 14 of the 17 most-potent monoclonal antibodies that we tested was reduced or abolished by the K417N, E484K or N501Y mutation. Notably, these mutations were selected when we cultured recombinant vesicular stomatitis virus expressing SARS-CoV-2 S in the presence of the monoclonal antibodies elicited by the vaccines. Together, these results suggest that the monoclonal antibodies in clinical use should be tested against newly arising variants, and that mRNA vaccines may need to be updated periodically to avoid a potential loss of clinical efficacy.- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.