Searched for: person:parkc07
in-biosketch:true
DOGMA-seq and multimodal, single-cell analysis in acute myeloid leukemia
Kim, JangKeun; Schanzer, Nathan; Singh, Ruth Subhash; Zaman, Mohammed I; Garcia-Medina, J Sebastian; Proszynski, Jacqueline; Ganesan, Saravanan; Dan Landau,; Park, Christopher Y; Melnick, Ari M; Mason, Christopher E
Acute myeloid leukemia (AML) is a complex cancer, yet advances in recent years from integrated genomics methods have helped improve diagnosis, treatment, and means of patient stratification. A recent example of a powerful, multimodal method is DOGMA-seq, which can measure chromatin accessibility, gene expression, and cell-surface protein levels from the same individual cell simultaneously. Previous bimodal single-cell techniques, such as CITE-seq (Cellular indexing of transcriptomes and epitopes), have only permitted the transcriptome and cell-surface protein expression measurement. DOGMA-seq, however, builds on this foundation and has implications for examining epigenomic, transcriptomic, and proteomic interactions between various cell types. This technique has the potential to be particularly useful in the study of cancers such as AML. This is because the cellular mechanisms that drive AML are rather heterogeneous and require a more complete understanding of the interplay between the genetic mutations, disruptions in RNA transcription and translation, and surface protein expression that cause these cancers to develop and evolve. This technique will hopefully contribute to a more clear and complete understanding of the growth and progression of complex cancers.
PMID: 39864897
ISSN: 1937-6448
CID: 5780482
Co-occurrence of thymoma and acute T-lymphoblastic leukemia/lymphoma: a case report and literature review [Case Report]
Frazzette, Nicholas; Ordner, Jeffrey; Narula, Navneet; Moreira, Andre L; Park, Christopher Y; Ward, Nicholas D
BACKGROUND/UNASSIGNED:A thymoma is a tumor originating from thymic epithelial cells variably associated with non-neoplastic lymphocytes. T-lymphoblastic leukemia/lymphoma (T-LBL) is thought to arise from precursor T-cells from bone marrow-derived hematopoietic stem cells that migrate to the thymus. While the association of secondary hematopoietic malignancies in thymoma is well established, only rarely in the literature have T-LBL and thymoma been seen in association and the relationship is poorly understood. Occasionally, distinction between the two can be difficult as immature lymphocytes in thymoma resemble T-LBL both morphologically and immunophenotypically. An accurate diagnosis is essential as treatments vary between these two entities. CASE DESCRIPTION/UNASSIGNED:We present the interesting case of a 64-year-old male, former smoker, originally from Uzbekistan, with a mediastinal mass diagnosed as small cell carcinoma in his home country and treated with chemotherapy. After immigrating to the United States, a positron emission tomography (PET) scan demonstrated a large, metabolically active mediastinal mass. He presented to our institution where a biopsy with histomorphologic and immunohistochemical analysis was diagnostic of type B1 thymoma. He was lost to follow-up, but represented months later with B symptoms. Flow cytometry, cytogenetics, and bone marrow biopsy were diagnostic of T-LBL. Although he was started on chemotherapy, his disease progressed and he expired 6 months after initial presentation. Post-mortem analysis of the mediastinal mass revealed the co-occurrence of benign thymocytes and neoplastic T-LBL lymphoblasts, further confirmed as two distinct entities by T-cell receptor (TCR) sequencing. CONCLUSIONS/UNASSIGNED:Co-occurrence of thymoma and T-LBL is a well-documented, though poorly understood, phenomenon. Literature review for this phenomenon reveals that type B thymoma is most commonly associated with T-LBL in these co-occurrences. Most cases are diagnosed synchronously, though in metachronous cases, the diagnosis of thymoma has always preceded the diagnosis of T-LBL. Of note, recently developed LMO2 immunohistochemical stain is positive in malignant lymphoblasts but negative in benign thymocytes, allowing for post-mortem evaluation of this case to be determined as a synchronous presentation. These entities are difficult to distinguish and require a multimodal diagnostic approach including histology, immunohistochemistry, flow cytometry, cytogenetics, and TCR sequencing.
PMCID:11982990
PMID: 40224340
ISSN: 2522-6711
CID: 5827212
Emetine induces oxidative stress, cell differentiation and NF-κB inhibition, suppressing AML stem/progenitor cells
Silva, Suellen L.R.; Dias, Ingrid R.S.B.; Rodrigues, Ana Carolina B.da C.; Costa, Rafaela G.A.; Oliveira, Maiara de S.; Barbosa, Gabriela A.da C.; Soares, Milena B.P.; Dias, Rosane B.; Valverde, Ludmila F.; Rocha, Clarissa A.G.; Roy, Nainita; Park, Christopher Y.; Bezerra, Daniel P.
Acute myeloid leukemia (AML) is a fatal malignancy of the blood and bone marrow. Leukemic stem cells (LSCs) are a rare subset of leukemic cells that promote the development and progression of AML, and eradication of LSCs is critical for effective control of this disease. Emetine is an FDA-approved antiparasitic drug with antitumor properties; however, little is known about its potential against LSCs. Herein, we explored the antileukemic potential of emetine, focusing on its effects on AML stem/progenitor cells. Emetine exhibited potent cytotoxic activity both in hematologic and solid cancer cells and induced AML cell differentiation. Emetine also inhibited AML stem/progenitor cells, as evidenced by decreased expression of CD34, CD97, CD99, and CD123 in KG-1a cells, indicating anti-AML stem/progenitor cell activities. The administration of emetine at a dosage of 10 mg/kg for two weeks showed no significant toxicity and significantly reduced xenograft leukemic growth in vivo. NF-κB activation was reduced in emetine-treated KG-1a cells, as shown by reduced phospho-NF-κB p65 (S529) and nuclear NF-κB p65. DNA fragmentation, YO-PRO-1 staining, mitochondrial depolarization and increased levels of active caspase-3 and cleaved PARP (Asp214) were detected in emetine-treated KG-1a cells. Moreover, treatment with the pancaspase inhibitor Z-VAD(OMe)-FMK partially prevented the apoptotic cell death induced by emetine. Emetine treatment also increased cellular and mitochondrial reactive oxygen species, and emetine-induced apoptosis in KG-1a cells was partially prevented by the antioxidant N-acetylcysteine, indicating that emetine induces apoptosis, at least in part, by inducing oxidative stress. Overall, these studies indicate that emetine is a novel potential anti-AML agent with promising activity against stem/progenitor cells, encouraging the development of further studies aimed at its clinical application.
SCOPUS:85191831799
ISSN: 2058-7716
CID: 5658942
BRAF V600E mutation and high expression of PD-L1 in Rosai-Dorfman disease: case report and review of the literature [Case Report]
Moen, Farnoush M; Youssef, Mariam M; Shukla, Mihir; Nierodzik, Mary Lynn; Mayerhoefer, Marius E; Park, Christopher
BRAF V600E mutations are frequently found in histiocytic/dendritic cell neoplasms such as Erdheim-Chester disease (ECD) and Langerhans cell histiocytosis (LCH), but few reports have also described BRAF mutations in Rosai-Dorfman disease (RDD), and even these cases may predominantly represent mixed histiocytosis. BRAF mutations have been studied in histiocytic/dendritic cell neoplasms and described to be associated with increased risk of relapse and long-term consequences, but few studies have examined BRAF V600E mutation in RDD, which is recognized as a neoplasm given the high frequency of MAPK pathway alterations. Here, we report a case of BRAF V600E-mutated RDD in a patient who presented with generalized lymphadenopathy. During our evaluation of this patient, we also found expression of PD-L1 in neoplastic histiocytes. During our review period, only few cases of RDD reported to harbor BRAF mutation or were evaluated for the expression of PDL1 by neoplastic cells. Given the potential challenges in distinguishing RDD from other histiocytic/dendritic cell neoplasms, including mixed histiocytosis with similar clinicopathological manifestations, we will discuss the current state of knowledge regarding the frequency and clinical impact of BRAF V600E in RDD, as well as the role of BRAF mutations in RDD pathogenesis. Distinction of BRAF V600E mutated histiocytic/dendritic cell neoplasms requires consideration of distinctive histopathological and immunophenotypic findings in appropriate clinical and radiologic setting. Given the increasing use of BRAF inhibitors as well as checkpoint blockade inhibitors to treat a number of cancers, we will discuss the clinical implications of the presence of BRAF V600E mutation and PD-L1 expression in RDD.
PMCID:11635026
PMID: 39592527
ISSN: 1865-5785
CID: 5762152
Acute inhibition of iron-sulfur cluster biosynthesis disrupts metabolic flexibility in mice
Molenaars, Marte; Mir, Hannan; Alvarez, Samantha W; Arivazhagan, Lakshmi; Rosselot, Carolina; Zhan, Di; Park, Christopher Y; Garcia-Ocana, Adolfo; Schmidt, Ann Marie; Possemato, Richard
UNLABELLED:Iron-sulfur clusters (ISCs) are cell-essential cofactors present in ∼60 proteins including subunits of OXPHOS complexes I-III, DNA polymerases, and iron-sensing proteins. Dysfunctions in ISC biosynthesis are associated with anemias, neurodegenerative disorders, and metabolic diseases. To assess consequences of acute ISC inhibition in a whole body setting, we developed a mouse model in which key ISC biosynthetic enzyme NFS1 can be acutely and reversibly suppressed. Contrary to in vitro ISC inhibition and pharmacological OXPHOS suppression, global NFS1 inhibition rapidly enhances lipid utilization and decreases adiposity without affecting caloric intake and physical activity. ISC proteins decrease, including key proteins involved in OXPHOS (SDHB), lipoic acid synthesis (LIAS), and insulin mRNA processing (CDKAL1), causing acute metabolic inflexibility. Age-related metabolic changes decelerate loss of adiposity substantially prolonged survival of mice with NFS1 inhibition. Thus, the observation that ISC metabolism impacts organismal fuel choice will aid in understanding the mechanisms underlying ISC diseases with increased risk for diabetes. HIGHLIGHTS/UNASSIGNED:Acute ISC inhibition leads to rapid loss of adiposity in miceMulti-metabolic pathway disruption upon ISC deficiency blocks energy storageNfs1 inhibition induces glucose dyshomeostasis due to ISC deficiency in β-cellsEnergy distress caused by inhibition of ISC synthesis is attenuated in aged mice.
PMCID:11370322
PMID: 39229169
ISSN: 2692-8205
CID: 5757792
Mapping the landscape of histomorphological cancer phenotypes using self-supervised learning on unannotated pathology slides
Claudio Quiros, Adalberto; Coudray, Nicolas; Yeaton, Anna; Yang, Xinyu; Liu, Bojing; Le, Hortense; Chiriboga, Luis; Karimkhan, Afreen; Narula, Navneet; Moore, David A; Park, Christopher Y; Pass, Harvey; Moreira, Andre L; Le Quesne, John; Tsirigos, Aristotelis; Yuan, Ke
Cancer diagnosis and management depend upon the extraction of complex information from microscopy images by pathologists, which requires time-consuming expert interpretation prone to human bias. Supervised deep learning approaches have proven powerful, but are inherently limited by the cost and quality of annotations used for training. Therefore, we present Histomorphological Phenotype Learning, a self-supervised methodology requiring no labels and operating via the automatic discovery of discriminatory features in image tiles. Tiles are grouped into morphologically similar clusters which constitute an atlas of histomorphological phenotypes (HP-Atlas), revealing trajectories from benign to malignant tissue via inflammatory and reactive phenotypes. These clusters have distinct features which can be identified using orthogonal methods, linking histologic, molecular and clinical phenotypes. Applied to lung cancer, we show that they align closely with patient survival, with histopathologically recognised tumor types and growth patterns, and with transcriptomic measures of immunophenotype. These properties are maintained in a multi-cancer study.
PMID: 38862472
ISSN: 2041-1723
CID: 5669022
The Effect of Diet Composition on the Post-operative Outcomes of Roux-en-Y Gastric Bypass in Mice
Stevenson, Matthew; Srivastava, Ankita; Nacher, Maria; Hall, Christopher; Palaia, Thomas; Lee, Jenny; Zhao, Chaohui Lisa; Lau, Raymond; Ali, Mohamed A E; Park, Christopher Y; Schlamp, Florencia; Heffron, Sean P; Fisher, Edward A; Brathwaite, Collin; Ragolia, Louis
PURPOSE/OBJECTIVE:Roux-en-Y gastric bypass (RYGB) leads to the improvement of many obesity-associated conditions. The degree to which post-operative macronutrient composition contributes to metabolic improvement after RYGB is understudied. METHODS:A mouse model of RYGB was used to examine the effects of diet on the post-operative outcomes of RYGB. Obese mice underwent either Sham or RYGB surgery and were administered either chow or HFD and then monitored for an additional 8 weeks. RESULTS:After RYGB, reductions to body weight, fat mass, and lean mass were similar regardless of diet. RYGB and HFD were independently detrimental to bone mineral density and plasma vitamin D levels. Independent of surgery, HFD accelerated hematopoietic stem and progenitor cell proliferation and differentiation and exhibited greater myeloid lineage commitment. Independent of diet, systemic iron deficiency was present after RYGB. In both Sham and RYGB groups, HFD increased energy expenditure. RYGB increased fecal energy loss, and HFD after RYGB increased fecal lipid content. RYGB lowered fasting glucose and liver glycogen levels but HFD had an opposing effect. Indices of insulin sensitivity improved independent of diet. HFD impaired improvements to dyslipidemia, NAFLD, and fibrosis. CONCLUSION/CONCLUSIONS:Post-operative diet plays a significant role in determining the degree to which RYGB reverses obesity-induced metabolic abnormalities such as hyperglycemia, dyslipidemia, and NAFLD. Diet composition may be targeted in order to assist in the treatment of post-RYGB bone mineral density loss and vitamin D deficiency as well as to reverse myeloid lineage commitment. HFD after RYGB continues to pose a significant multidimensional health risk.
PMID: 38191966
ISSN: 1708-0428
CID: 5707802
Correction: The Effect of Diet Composition on the Post-operative Outcomes of Roux-en-Y Gastric Bypass in Mice
Stevenson, Matthew; Srivastava, Ankita; Nacher, Maria; Hall, Christopher; Palaia, Thomas; Lee, Jenny; Zhao, Chaohui Lisa; Lau, Raymond; Ali, Mohamed A E; Park, Christopher Y; Schlamp, Florencia; Heffron, Sean P; Fisher, Edward A; Brathwaite, Collin; Ragolia, Louis
PMID: 38236349
ISSN: 1708-0428
CID: 5737482
(Reg)ulation of hematopoietic lineage fates [Comment]
Sreejit, Gopalkrishna; Park, Christopher Y
PMID: 38236614
ISSN: 1528-0020
CID: 5626682
Molecular profiling of pre- and post- 5-azacytidine myelodysplastic syndrome samples identifies predictors of response
González, Mónica Del Rey; Chakraborty, Sohini; Hernández-Sánchez, Jesús MarÃa; Diez Campelo, MarÃa; Park, Christopher Y; Hernández Rivas, Jesús MarÃa
Treatment with the hypomethylating agent 5-azacytidine (AZA) increases survival in high-risk (HR) myelodysplastic syndrome (MDS) patients, but predicting patient response and overall survival remains challenging. To address these issues, we analyzed mutational and transcriptional profiles in CD34+ hematopoietic stem/progenitor cells (HSPCs) before and following AZA therapy in MDS patients. AZA treatment led to a greater reduction in the mutational burden in both blast and hematological responders than non-responders. Blast and hematological responders showed transcriptional evidence of pre-treatment enrichment for pathways such as oxidative phosphorylation, MYC targets, and mTORC1 signaling. While blast non-response was associated with TNFa signaling and leukemia stem cell signature, hematological non-response was associated with cell-cycle related pathways. AZA induced similar transcriptional responses in MDS patients regardless of response type. Comparison of blast responders and non-responders to normal controls, allowed us to generate a transcriptional classifier that could predict AZA response and survival. This classifier outperformed a previously developed gene signature in a second MDS patient cohort, but signatures of hematological responses were unable to predict survival. Overall, these studies characterize the molecular consequences of AZA treatment in MDS HSPCs and identify a potential tool for predicting AZA therapy responses and overall survival prior to initiation of therapy.
PMCID:11456566
PMID: 39376992
ISSN: 2234-943x
CID: 5705972