Anasuya Dighe, PhD
Associate Research ScientistCards
Appointments
Medical Oncology and Hematology
Primary
Contact Info
About
Copy Link
Titles
Associate Research Scientist
Appointments
Medical Oncology and Hematology
Associate Research ScientistPrimary
Other Departments & Organizations
- Medical Oncology and Hematology
Education & Training
- PhD
- Indian Institute of Science, Computational Biology
- MSc
- University of Pune, Bioinformatics
- BSc
- University of Pune, Biotechnology
Research
Copy Link
Overview
Medical Research Interests
Biological Evolution; Computational Biology; Exome Sequencing
ORCID
0000-0002-0444-5969
Research at a Glance
Yale Co-Authors
Frequent collaborators of Anasuya Dighe's published research.
Publications Timeline
A big-picture view of Anasuya Dighe's research output by year.
Research Interests
Research topics Anasuya Dighe is interested in exploring.
David A. Braun, MD, PhD
Soki Kashima, MD, PhD
Michael Hurwitz, MD, PhD
Yansheng Liu, PhD
Adebowale Adeniran, MD
David Schoenfeld, MD, PhD
16Publications
124Citations
Computational Biology
Biological Evolution
Publications
2025
24TGF-Beta alters CD8+ T cell phenotype and drives resistance to immune checkpoint inhibitors (ICI) in renal cell carcinoma (RCC)
Rout R, Kashima S, Hugaboom M, Ye Z, Schindler N, Dighe A, Sun M, Lee G, Xu W, Signoretti S, McGregor B, McKay R, Choueri T, Braun D. 24TGF-Beta alters CD8+ T cell phenotype and drives resistance to immune checkpoint inhibitors (ICI) in renal cell carcinoma (RCC). The Oncologist 2025, 30: oyaf276.025. PMCID: PMC12509635, DOI: 10.1093/oncolo/oyaf276.025.Peer-Reviewed Original ResearchConceptsCD8+ T cellsImmune checkpoint inhibitorsRenal cell carcinomaCD8+ T cell phenotypesRCC tumor microenvironmentICI responseAdvanced renal cell carcinomaPD-1 expressionAnti-tumor immunityTex cellsT-cell phenotypeT cellsTumor microenvironmentPD-1TGF-betaICI resistanceTissue residencyGene programT cell-mediated anti-tumor immunityHealthy donor peripheral blood mononuclear cellsResistance to immune checkpoint inhibitorsDonor peripheral blood mononuclear cellsCD8+ T cell populationsICI-based combination therapyImmune checkpoint inhibitor treatment31Lymphocyte heat shock signature predicts response to immune checkpoint blockade in renal cell carcinoma
Burns E, Kashima S, Rout R, Hugaboom M, Ye Z, Schindler N, Dighe A, Sun M, Lee G, Xu W, Atkins M, Signoretti S, McGregor B, McKay R, Choueri T, Braun D. 31Lymphocyte heat shock signature predicts response to immune checkpoint blockade in renal cell carcinoma. The Oncologist 2025, 30: oyaf276.032. PMCID: PMC12509613, DOI: 10.1093/oncolo/oyaf276.032.Peer-Reviewed Original ResearchConceptsImmune checkpoint blockade therapyImmune checkpoint blockadeRenal cell carcinomaProgression free survivalResponse to ICBCD8+ T cellsCD4+ T cellsT cellsAssociated with responseCheckpoint blockadeCell carcinomaCD4+ T cell clustersResponse to immune checkpoint blockadeAssociated with clinical benefitInnate lymphoid cellsModule scoresPotential mechanisms of responseT cell clustersPotential mechanismsFree survivalImmune therapyLymphocyte subsetsProgressive diseaseTumor microenvironmentTherapeutic response41Single-cell dissection of immunosuppressive myeloid subclusters driving resistance to immune checkpoint therapy in renal cell carcinoma (RCC)
Kashima S, Rout R, Hugaboom M, Ye Z, Schindler N, Dighe A, Sun M, Lee G, Xu W, Signoretti S, McGregor B, McKay R, Choueri T, Braun D. 41Single-cell dissection of immunosuppressive myeloid subclusters driving resistance to immune checkpoint therapy in renal cell carcinoma (RCC). The Oncologist 2025, 30: oyaf276.042. PMCID: PMC12509592, DOI: 10.1093/oncolo/oyaf276.042.Peer-Reviewed Original ResearchConceptsImmune checkpoint inhibitorsRenal cell carcinomaTumor microenvironmentTumor samplesResistance to immune checkpoint therapyImmune checkpoint inhibitor regimensResponse to ICI therapyImmune checkpoint inhibitor therapyMetastatic renal cell carcinomaAdvanced renal cell carcinomaContribution of myeloid cellsICI-based regimensImmune checkpoint therapyPotential mechanisms of resistanceT cell statesHypoxia-related signalingTranscriptional programsWilcoxon signed-rank testPotential mechanismsMechanisms of resistanceICI resistanceICI therapyImmunoregulatory populationTAM subsetsCheckpoint inhibitorsInvestigation of tumor-associated macrophages (TAMs) and therapeutic resistance to immune checkpoint inhibitors (ICI) through single-cell analysis of renal cell carcinoma (RCC).
Kashima S, Rout R, Hugaboom M, Ye Z, Schindler N, Malik R, Dighe A, Sun M, Lee G, Xu W, Signoretti S, Schoenfeld D, Hurwitz M, Adeniran A, Humphrey P, Kenney P, McGregor B, McKay R, Choueiri T, Braun D. Investigation of tumor-associated macrophages (TAMs) and therapeutic resistance to immune checkpoint inhibitors (ICI) through single-cell analysis of renal cell carcinoma (RCC). Journal Of Clinical Oncology 2025, 43: 4527-4527. DOI: 10.1200/jco.2025.43.16_suppl.4527.Peer-Reviewed Original ResearchConceptsImmune checkpoint inhibitorsTumor-associated macrophagesRenal cell carcinomaICI-based therapyRCC tumor microenvironmentTumor microenvironmentNon-respondersGene programResistance to immune checkpoint inhibitorsImmune checkpoint inhibitor resistanceTumor-associated macrophage populationSuppress antitumor immunityWilcoxon Signed Rank TestInterferon-stimulated genesMechanisms of resistanceCheckpoint inhibitorsAntitumor immunityStable diseaseSystemic therapyCombination therapyCell carcinomaPotential therapeutic targetTreatment resistanceAntigen presentationTumor samplesCo-expression network-based analysis of gene programs contributing to immune checkpoint inhibitor (ICI) resistance in renal cell carcinoma (RCC).
Malik R, Rout R, Kashima S, Saad E, Kane H, Shah V, Hugaboom M, Ye Z, Schindler N, Dighe A, Sun M, Lee G, Xu W, Signoretti S, McGregor B, McKay R, Atkins M, Van Allen E, Choueiri T, Braun D. Co-expression network-based analysis of gene programs contributing to immune checkpoint inhibitor (ICI) resistance in renal cell carcinoma (RCC). Journal Of Clinical Oncology 2025, 43: 4530-4530. DOI: 10.1200/jco.2025.43.16_suppl.4530.Peer-Reviewed Original ResearchConceptsImmune checkpoint inhibitorsRenal cell carcinomaImmune cellsImmune infiltrationSignature scoreRCC tumor microenvironmentResistance to immune checkpoint inhibitorsAssociated with worse PFSICI-based therapyScRNA-seq dataGene programCD8+ TPhase II trialICI resistanceImmune-lowCheckpoint inhibitorsRibosomal proteinsCD4+II trialPrognostic roleRenal cell carcinoma tumor sampleRNA-seqCell carcinomaTumor microenvironmentScRNA-seqProgressive natural killer cell dysfunction in advanced-stage clear-cell renal cell carcinoma and association with clinical outcomes
Xu W, Birch G, Meliki A, Moritz V, Bharadwaj M, Schindler N, Labaki C, Saliby R, Dinh K, Horst J, Sun M, Kashima S, Hugaboom M, Dighe A, Machaalani M, Lee G, Hurwitz M, McGregor B, Hirsch M, Shukla S, McDermott D, Signoretti S, Romee R, Choueiri T, Braun D. Progressive natural killer cell dysfunction in advanced-stage clear-cell renal cell carcinoma and association with clinical outcomes. ESMO Open 2025, 10: 104105. PMID: 39813824, PMCID: PMC11783098, DOI: 10.1016/j.esmoop.2024.104105.Peer-Reviewed Original ResearchCitationsAltmetricMeSH Keywords and ConceptsConceptsClear-cell renal cell carcinomaAdvanced clear-cell renal cell carcinomaNK cell phenotypeNK cell functionNK cellsRenal cell carcinomaClinical outcomesCell carcinomaCell phenotypeRestoration of NK cell functionNormal kidneyNatural killer (NK) cellsMarkers of tissue residencyNatural killer cell dysfunctionAssociated with worse overall survivalClear-cell renal cell carcinoma patientsLocalized clear-cell renal cell carcinomaTumor-infiltrating NK cellsAnalyzed single-cell RNA sequencing dataAssociated with worse survivalExpression of cytotoxic genesPaired normal kidneyLocal tumor extensionNK cell subsetsPrimary tumor specimensSpatial Genomic Approaches to Investigate HOX Genes in Mouse Brain Tissues
Shelar A, Dighe A. Spatial Genomic Approaches to Investigate HOX Genes in Mouse Brain Tissues. Methods In Molecular Biology 2025, 2889: 235-244. PMID: 39745616, DOI: 10.1007/978-1-0716-4322-8_16.Peer-Reviewed Original ResearchMeSH Keywords and ConceptsConceptsTranscriptomic toolsHox genesTranscriptome gene expression analysisSpatial gene expression analysisExpression patternsMouse brain tissueGene expression patternsGene expression analysisBioinformatics protocolGenomic approachesExpression analysisGene expressionGenesAnalysis of mouse brain tissuePreserving tissue integrityMouse brainBrain tissueExpressionTissue integrityTranscriptomeBioinformaticsHoxMiceTissue
2023
Experimental and phylogenetic evidence for correlated gene expression evolution in endometrial and skin fibroblasts
Dighe A, Maziarz J, Ibrahim-Hashim A, Gatenby R, Kshitiz, Levchenko A, Wagner G. Experimental and phylogenetic evidence for correlated gene expression evolution in endometrial and skin fibroblasts. IScience 2023, 27: 108593. PMID: 38174318, PMCID: PMC10762354, DOI: 10.1016/j.isci.2023.108593.Peer-Reviewed Original ResearchCitationsAltmetricConceptsGene expression changesGene expression evolutionEndometrial stromal fibroblastsExpression evolutionExpression changesCell typesGene expressionSimilar gene expression changesSubstantial gene expression changesGene expression profilesSkin fibroblastsMultiple cell typesEvolutionary correlationPhylogenetic evidenceEvolutionary changeDermal skin fibroblastsMammalian speciesExpression profilesPlacental invasivenessComparative datasetCancer growthCultured skin fibroblastsStromal fibroblastsFibroblastsMouse strains
2022
Proteotype coevolution and quantitative diversity across 11 mammalian species
Ba Q, Hei Y, Dighe A, Li W, Maziarz J, Pak I, Wang S, Wagner GP, Liu Y. Proteotype coevolution and quantitative diversity across 11 mammalian species. Science Advances 2022, 8: eabn0756. PMID: 36083897, PMCID: PMC9462687, DOI: 10.1126/sciadv.abn0756.Peer-Reviewed Original ResearchCitationsAltmetricMeSH Keywords and ConceptsConceptsMammalian speciesRNA metabolic processesCommon mammalian speciesUbiquitin-proteasome systemEvolutionary profilingMammalian lineagesProteomic methodsProtein degradationProtein abundanceGene expressionProtein expression levelsHigh interspeciesMetabolic processesCovariation analysisFunctional roleNucleotide levelExpression levelsQuantitative diversityCoevolutionMammalsSpeciesRemarkable variationExpressionTranscriptomeBiological variabilityTracing the cis-regulatory changes underlying the endometrial control of placental invasion
Suhail Y, Maziarz JD, Novin A, Dighe A, Afzal J, Wagner G, Kshitiz. Tracing the cis-regulatory changes underlying the endometrial control of placental invasion. Proceedings Of The National Academy Of Sciences Of The United States Of America 2022, 119: e2111256119. PMID: 35110402, PMCID: PMC8832988, DOI: 10.1073/pnas.2111256119.Peer-Reviewed Original ResearchCitationsAltmetricMeSH Keywords and ConceptsConceptsCis-regulatory changesComparative genomic investigationCis-regulatory elementsTranscription factor GATA2Placental invasionCancer cell invasionEutherian mammalsTranscription factorsGenomic investigationsInvasibilityGene knockoutGenomic mechanismsDifferent speciesCell invasionEndometrial controlCancer malignancyDegree of invasionSpecies differencesCancer disseminationMaternal endometriumInterspecies differencesPlacental trophoblastsStromal cellsStromal characteristicsInvasion
Get In Touch
Copy Link