Skip to main content

Publications search

Found 37048 matches. Displaying 1451-1460
He X, Ashbrook AW, Du Y, Wu J, Hoffmann HH, Zhang C, Xia L, Peng YC, Tumas KC, Singh BK, Qi CF, Myers TG, Long CA, Liu CY, Wang RF, Rice CM, Su XZ
Show All Authors

RTP4 inhibits IFN-I response and enhances experimental cerebral malaria and neuropathology

PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 2020 AUG 11; 117(32):19465-19474
Infection by malaria parasites triggers dynamic immune responses leading to diverse symptoms and pathologies; however, the molecular mechanisms responsible for these reactions are largely unknown. We performed Trans-species Expression Quantitative Trait Locus analysis to identify a large number of host genes that respond to malaria parasite infections. Here we functionally characterize one of the host genes called receptor transporter protein 4 (RTP4) in responses to malaria parasite and virus infections. RTP4 is induced by type I IFN (IFN-I) and binds to the TANK-binding kinase (TBK1) complex where it negatively regulates TBK1 signaling by interfering with expression and phosphorylation of both TBK1 and IFN regulatory factor 3. Rtp4(-/-) mice were generated and infected with malaria parasite Plasmodiun berghei ANKA. Significantly higher levels of IFN-I response in microglia, lower parasitemia, fewer neurologic symptoms, and better survival rateswere observed in Rtp4(-/-) than in wild-type mice. Similarly, RTP4 deficiency significantly reduced West Nile virus titers in the brain, but not in the heart and the spleen, of infected mice, suggesting a specific role for RTP4 in brain infection and pathology. This study reveals functions of RTP4 in IFN-I response and a potential target for therapy in diseases with neuropathology.
Pisa R, Kapoor TM
Show All Authors

Chemical strategies to overcome resistance against targeted anticancer therapeutics

NATURE CHEMICAL BIOLOGY 2020 AUG 1; 16(8):817-825
Emergence of resistance is a major factor limiting the efficacy of molecularly targeted anticancer drugs. Understanding the specific mutations, or other genetic or cellular changes, that confer drug resistance can help in the development of therapeutic strategies with improved efficacies. Here, we outline recent progress in understanding chemotype-specific mechanisms of resistance and present chemical strategies, such as designing drugs with distinct binding modes or using proteolysis targeting chimeras, to overcome resistance. We also discuss how targeting multiple binding sites with bifunctional inhibitors or identifying collateral sensitivity profiles can be exploited to limit the emergence of resistance. Finally, we highlight how incorporating analyses of resistance early in drug development can help with the design and evaluation of therapeutics that can have long-term benefits for patients.
Garris CS, Luke JJ
Show All Authors

Dendritic Cells, the T-cell-inflamed Tumor Microenvironment, and Immunotherapy Treatment Response

CLINICAL CANCER RESEARCH 2020 AUG 1; 26(15):3901-3907
The development of the most successful cancer immunotherapies in solid tumors, immune-checkpoint blockade, has focused on factors regulating T-cell activation. Until recently, the field has maintained a predominately T-cell centric view of immunotherapy, leaving aside the impact of innate immunity and especially myeloid cells. Dendritic cells (DC) are dominant partners of T cells, necessary for initiation of adaptive immune responses. Emerging evidence supports a broader role for DCs in tumors including the maintenance and support of effector functions during T-cell responses. This relationship is evidenced by the association of activated DCs with immune-checkpoint blockade responses and transcriptional analysis of responding tumors demonstrating the presence of type I IFN transcripts and DC relevant chemokines. T-cell-inflamed tumors preferentially respond to immunotherapies compared with non-T-cell-inflamed tumors and this model suggests a potentially modifiable spectrum of tumor microenvironmental immunity. Although host and commensal factors may limit the T-cell-inflamed phenotype, tumor cell intrinsic factors are gaining prominence as therapeutic targets. For example, tumor WNT/beta-catenin signaling inhibits production of chemokine gradients and blocking DC recruitment to tumors. Conversely, mechanisms of innate immune nucleic acid sensing, normally operative during pathogen response, may enhance DC accumulation and make tumors more susceptible to cancer immunotherapy. Elucidating mechanisms whereby DCs infiltrate and become activated within tumors may provide new opportunities for therapeutic intervention. Conceptually, this would facilitate conversion of non-T-cellinflamed to T-cell-inflamed states or overcome secondary resistance mechanisms in T-cell-inflamed tumors, expanding the proportion of patients who benefit from cancer immunotherapy.
Sato S, Tanaka N, Arimura Y, Kujirai T, Kurumizaka H
Show All Authors

The N-terminal and C-terminal halves of histone H2A.Z independently function in nucleosome positioning and stability

GENES TO CELLS 2020 AUG; 25(8):538-546
Nucleosome positioning and stability affect gene regulation in eukaryotic chromatin. Histone H2A.Z is an evolutionally conserved histone variant that forms mobile and unstable nucleosomes in vivo and in vitro. In the present study, we reconstituted nucleosomes containing human H2A.Z.1 mutants, in which the N-terminal or C-terminal half of H2A.Z.1 was replaced by the corresponding canonical H2A region. We found that the N-terminal portion of H2A.Z.1 is involved in flexible nucleosome positioning, whereas the C-terminal portion leads to weak H2A.Z.1-H2B association in the nucleosome. These results indicate that the N-terminal and C-terminal portions are independently responsible for the H2A.Z.1 nucleosome characteristics.
Conti F, Carsetti R, Casanova JL, Fischer A, Cancrini C
Show All Authors

A 23-Year Follow-Up of a Patient with Gain-of-Function IkB-Alpha Mutation and Stable Full Chimerism After Hematopoietic Stem Cell Transplantation

JOURNAL OF CLINICAL IMMUNOLOGY 2020 AUG; 40(6):927-933
Frew JW
Show All Authors

Anti-Saccharomyces cervisiae antibodies in hidradenitis suppurativa: More than a gut feeling

JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY 2020 AUG; 146(2):458-458
Noureddin M, Chan JL, Barradas K, Dimick-Santos L, Schabel E, Omokaro SO, Anania FA, Myers RP, Miller V, Sanyal AJ, Chalasani N, Anania FA, Bajaj J, Barradas K, Berzigotti A, Birman P, Bosch J, Brower A, Calboli D, Chalasani N, Chan JL, Charlton W, Dickinson K, Dimick-Santos L, Filozof C, Forsgren MF, Fuchs M, Garcia-Tsao G, Gonzalez-Abraldes J, Gruss HJ, Hansen M, Hosman S, Imperial J, Jones D, Lalazar G, Leinhard OD, Lyons E, McColgan B, Mehta R, Mesenbrink P, Miller V, Myers R, Noureddin M, Omokaro SO, Pei V, Ratziu V, Regev A, Riccio R, Sanyal A, Schabel E, Seo S, Smith A, Szitanyi P, Traber P
Show All Authors

Attribution of Nonalcoholic Steatohepatitis as an Etiology of Cirrhosis for Clinical Trials Eligibility: Recommendations From the Multi-stakeholder Liver Forum

GASTROENTEROLOGY 2020 AUG; 159(2):422-427.e1
Ostendorf BN, Bilanovic J, Adaku N, Tafreshian KN, Tavora B, Vaughan RD, Tavazoie SF
Show All Authors

Common germline variants of the human APOE gene modulate melanoma progression and survival

NATURE MEDICINE 2020 JUL; 26(7):1048-1053
Heritable APOE variants in patients with melanoma influence anti-tumor immunity and modulate metastatic progression and response to immunotherapy. Common germline variants of the APOE gene are major risk modifiers of neurodegenerative and atherosclerotic diseases(1-3), but their effect on cancer outcome is poorly defined. Here we report that, in a reversal of their effect on Alzheimer's disease, the APOE4 and APOE2 variants confer favorable and poor outcomes in melanoma, respectively. Mice expressing the human APOE4 allele exhibited reduced melanoma progression and metastasis relative to APOE2 mice. APOE4 mice exhibited enhanced anti-tumor immune activation relative to APOE2 mice, and T cell depletion experiments showed that the effect of APOE genotype on melanoma progression was mediated by altered anti-tumor immunity. Consistently, patients with melanoma carrying the APOE4 variant experienced improved survival in comparison to carriers of APOE2. Notably, APOE4 mice also showed improved outcomes under PD1 immune checkpoint blockade relative to APOE2 mice, and patients carrying APOE4 experienced improved anti-PD1 immunotherapy survival after progression on frontline regimens. Finally, enhancing APOE expression via pharmacologic activation of liver X receptors, previously shown to boost anti-tumor immunity(4), exhibited therapeutic efficacy in APOE4 mice but not in APOE2 mice. These findings demonstrate that pre-existing hereditary genetics can impact progression and survival outcomes of a future malignancy and warrant prospective investigation of APOE genotype as a biomarker for melanoma outcome and therapeutic response.