27 results on '"Kurmi K"'
Search Results
2. Integrated nutrient management in maize (Zea mays) under different microclimatic regimes
- Author
-
GOGOI, TAPAN, primary, DUTTA, RINJUMONI, additional, SARMAH, A K, additional, and KURMI, K, additional
- Published
- 2022
- Full Text
- View/download PDF
3. Economic Size of Water Harvesting Structures in High Rainfall Areas-An Optimization Approach
- Author
-
Bora, Pradip K, Kurmi, K., Barua, N. G., and Karmakar, R.M.
- Published
- 2010
4. Double transplanting of late transplanted sali rice under lowland situation
- Author
-
Ashem, S.S., Thakuria, K., and Kurmi, K.*
- Published
- 2010
5. Response of niger (Guizotia abyssinica) to different levels of nitrogen, phosphorus, potassium and sulphur
- Author
-
Bora, P., primary, Bora, P. C., additional, Kurmi, K., additional, and Kalita, S., additional
- Published
- 2021
- Full Text
- View/download PDF
6. Yield and nutrient uptake in niger (Guizotia abyssinica) as influenced by different levels of nitrogen, phosphorus, potassium and sulphur
- Author
-
Bora, P., primary, Bora, P. C., additional, Kurmi, K., additional, and Kalita, S., additional
- Published
- 2021
- Full Text
- View/download PDF
7. Conservation of Soil Moisture and Sustenance of Yield in Late Sown toria in Sali Rice Fallows through Moisture Conservation and INM Practices
- Author
-
Mahanta, Nitumoni, primary, Kurmi, K., additional, and Das, J.C., additional
- Published
- 2019
- Full Text
- View/download PDF
8. Efficacy of Organically Managed Cropping System in Improvement of Soil Health in Ne Hill Region
- Author
-
Ezung, N. Khumdemo, primary, Choudhary, J.K., additional, Kurmi, K., additional, Kikon, Noyingthung, additional, and sunep, Moa, additional
- Published
- 2018
- Full Text
- View/download PDF
9. Kinetics of Boron Adsorption in Soils of Assam State, India
- Author
-
Tamuli, Babita, primary, Bhattacharyya, D., additional, Borua, N.G., additional, Basumatary, A., additional, Das, K.N., additional, and Kurmi, K., additional
- Published
- 2017
- Full Text
- View/download PDF
10. Evaluation of CERES-Rice model for Upper Brahmaputra Valley Zone of Assam.
- Author
-
MEDHI, KULDIP, NEOG, P., GOSWAMI, B., DEKA, R. L., KURMI, K., SARMAH, K., and KHANIKAR, P. G.
- Subjects
RICE farming ,CROP yields ,MATHEMATICAL models ,RICE varieties ,AGRICULTURE ,AGRICULTURAL climatology - Abstract
The article presents a study of the crop environment resource synthesis-rice (CERES-rice) model for two rice cultivars in the Upper Brahmaputra valley zone in Assam, India. It discusses the calibration and validation of the CERES-rice model by comparing observed and simulated phenological events and grain yields in both rice varieties, demonstrating that the model is good enough to predict the phenological events of both varieties and can be usefull for farmers in crop management operations.
- Published
- 2017
- Full Text
- View/download PDF
11. Effect of Tillage on Soil Moisture and Energy Conservation in Production ofToriain an Inceptisol of Assam
- Author
-
Barua, N.G., primary, Bora, P.K., additional, Kurmi, K., additional, Karmakar, R.M., additional, and Pathak, P.K., additional
- Published
- 2014
- Full Text
- View/download PDF
12. Performance of Lowland Transplanted Sali (Winter) Rice Varieties Under Late Planting in Assam, India
- Author
-
Choudhary, J. K., Kurmi, K., Baruah, R. K., and Das, G. R.
- Subjects
Performance Testing ,India ,Variety Trials ,Transplanting - Abstract
This article 'Performance of Lowland Transplanted Sali (Winter) Rice Varieties Under Late Planting in Assam, India' appeared in the International Rice Research Notes series, created by the International Rice Research Institute (IRRI) to expedite communication among scientists concerned with the development of improved technology for rice and rice-based systems. The series is a mechanism to help scientists keep each other informed of current rice research findings. The concise scientific notes are meant to encourage rice scientists to communicate with one another to obtain details on the research reported.
- Published
- 1994
- Full Text
- View/download PDF
13. Nutrient control of growth and metabolism through mTORC1 regulation of mRNA splicing.
- Author
-
Ogawa T, Isik M, Wu Z, Kurmi K, Meng J, Cho S, Lee G, Fernandez-Cardenas LP, Mizunuma M, Blenis J, Haigis MC, and Blackwell TK
- Abstract
Cellular growth and organismal development are remarkably complex processes that require the nutrient-responsive kinase mechanistic target of rapamycin complex 1 (mTORC1). Anticipating that important mTORC1 functions remained to be identified, we employed genetic and bioinformatic screening in C. elegans to uncover mechanisms of mTORC1 action. Here, we show that during larval growth, nutrients induce an extensive reprogramming of gene expression and alternative mRNA splicing by acting through mTORC1. mTORC1 regulates mRNA splicing and the production of protein-coding mRNA isoforms largely independently of its target p70 S6 kinase (S6K) by increasing the activity of the serine/arginine-rich (SR) protein RSP-6 (SRSF3/7) and other splicing factors. mTORC1-mediated mRNA splicing regulation is critical for growth; mediates nutrient control of mechanisms that include energy, nucleotide, amino acid, and other metabolic pathways; and may be conserved in humans. Although mTORC1 inhibition delays aging, mTORC1-induced mRNA splicing promotes longevity, suggesting that when mTORC1 is inhibited, enhancement of this splicing might provide additional anti-aging benefits., Competing Interests: Declaration of interests M.C.H. is a member of the scientific advisory board of Alixia Therapeutics., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Profiling Proteins and Phosphorylation Sites During T Cell Activation Using an Integrated Thermal Shift Assay.
- Author
-
Gassaway BM, Huttlin EL, Huntsman EM, Yaron-Barir TM, Johnson JL, Kurmi K, Cantley LC, Paulo JA, Ringel AE, Gygi SP, and Haigis MC
- Subjects
- Phosphorylation, Phosphoproteins metabolism, Animals, Humans, Protein Stability, Signal Transduction, Tandem Mass Spectrometry, Mice, T-Lymphocytes metabolism, Lymphocyte Activation, Proteomics methods
- Abstract
T cell activation is a complex biological process of naive cells maturing into effector cells. Proteomic and phospho-proteomic approaches have provided critical insights into this process, yet it is not always clear how changes in individual proteins or phosphorylation sites have functional significance. Here, we developed the Phosphorylation Integrated Thermal Shift Assay (PITSA) that combines the measurement of protein or phosphorylation site abundance and thermal stability into a single tandem mass tags experiment and apply this method to study T cell activation. We quantified the abundance and thermal stability of over 7500 proteins and 5000 phosphorylation sites and identified significant differences in chromatin-related, TCR signaling, DNA repair, and proliferative phosphoproteins. PITSA may be applied to a wide range of biological contexts to generate hypotheses as to which proteins or phosphorylation sites are functionally regulated in a given system as well as the mechanisms by which this regulation may occur., Competing Interests: Conflict of interests The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: S. P. G. is a member of the scientific advisory boards of Cell Signaling Technologies and ThermoFisher Scientific. L. C. C. is a founder and member of the board of directors of Agios Pharmaceuticals and is a founder and receives research support from Petra Pharmaceuticals; is listed as an inventor on a patent (WO2019232403A1, Weill Cornell Medicine) for combination therapy for PI3K-associated disease or disorder, and the identification of therapeutic interventions to improve response to PI3K inhibitors for cancer treatment; is a co-founder and shareholder in Faeth Therapeutics; has equity in and consults for Cell Signaling Technologies, Volastra, Larkspur and 1 Base Pharmaceuticals; and consults for Loxo-Lilly. J. L. J has received consulting fees from Scorpion Therapeutics and Volastra Therapeutics. T. M. Y.-B. is a co-founder of DeStroke., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Metabolic modulation of mitochondrial mass during CD4 + T cell activation.
- Author
-
Kurmi K, Liang D, van de Ven R, Georgiev P, Gassaway BM, Han S, Notarangelo G, Harris IS, Yao CH, Park JS, Hu SH, Peng J, Drijvers JM, Boswell S, Sokolov A, Dougan SK, Sorger PK, Gygi SP, Sharpe AH, and Haigis MC
- Subjects
- Mice, Animals, T-Lymphocytes, Lymphocyte Activation, Receptors, Antigen, T-Cell, CD4-Positive T-Lymphocytes, Encephalomyelitis, Autoimmune, Experimental drug therapy
- Abstract
Mitochondrial biogenesis initiates within hours of T cell receptor (TCR) engagement and is critical for T cell activation, function, and survival; yet, how metabolic programs support mitochondrial biogenesis during TCR signaling is not fully understood. Here, we performed a multiplexed metabolic chemical screen in CD4
+ T lymphocytes to identify modulators of metabolism that impact mitochondrial mass during early T cell activation. Treatment of T cells with pyrvinium pamoate early during their activation blocks an increase in mitochondrial mass and results in reduced proliferation, skewed CD4+ T cell differentiation, and reduced cytokine production. Furthermore, administration of pyrvinium pamoate at the time of induction of experimental autoimmune encephalomyelitis, an experimental model of multiple sclerosis in mice, prevented the onset of clinical disease. Thus, modulation of mitochondrial biogenesis may provide a therapeutic strategy for modulating T cell immune responses., Competing Interests: Declaration of interests M.C.H. serves on the advisory boards for Alixia, Minovia, and MitoQ. M.C.H. has received funding from Agilent and Roche in the last five years. M.C.H. and K.K. have patents pending on the role of PP in T cells and mitochondria. A.S. is an employee of Flagship Labs 84, Inc, a subsidiary of Flagship Pioneering. J.M.D. reports grants from NIH during the conduct of the study, as well as personal fees from ElevateBio (consulting) and Third Rock Ventures (consulting) outside the submitted work. S.H. has consulted for Merck KGaA. I.S.H. reports financial support from Kojin Therapeutics and consulting fees for Ono Pharma USA. Fundings and fees from these companies are outside the scope of the current work. P.G. reports personal fees from RA Capital for consulting outside the submitted work. P.K.S. is a member of the SAB or BOD member of Applied Biomath, RareCyte Inc., and Glencoe Software; P.K.S. is also a member of the NanoString SAB, and the Sorger laboratory has received research funding from Novartis and Merck in the last five years. P.K.S. declares that none of these relationships have influenced the content of this manuscript. S.K.D. receives research funding unrelated to this project from Novartis and BMS and is a founder and scientific advisory board member for Kojin. A.H.S. has funding from Quark, Merck, AbbVie, Moderna, and Vertex unrelated to the submitted work. A.H.S. serves on advisory boards for SQZ Biotechnologies, Selecta, Elpiscience, Monopteros, Bicara, Fibrogen, Alixia, IOME, Corner Therapeutics, Glaxo Smith Kline, Amgen, and Janssen. She also is on scientific advisory boards for the Massachusetts General Cancer Center, Program in Cellular and Molecular Medicine at Boston Children’s Hospital, the Human Oncology and Pathogenesis Program at Memorial Sloan Kettering Cancer Center, the Gladstone Institute, and the Johns Hopkins Bloomberg-Kimmel Institute for Cancer Immunotherapy. She is an academic editor for the Journal of Experimental Medicine. A.H.S. has patents/pending royalties on the PD-1 pathway from Roche and Novartis., (Copyright © 2023 Elsevier Ltd. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
16. Uncoupled glycerol-3-phosphate shuttle in kidney cancer reveals that cytosolic GPD is essential to support lipid synthesis.
- Author
-
Yao CH, Park JS, Kurmi K, Hu SH, Notarangelo G, Crowley J, Jacobson H, Hui S, Sharpe AH, and Haigis MC
- Subjects
- Humans, Glycerol metabolism, Glycerolphosphate Dehydrogenase genetics, Glycerolphosphate Dehydrogenase metabolism, NAD metabolism, Oxidation-Reduction, Phosphates metabolism, Glycerol-3-Phosphate Dehydrogenase (NAD+) genetics, Glycerol-3-Phosphate Dehydrogenase (NAD+) metabolism, Kidney Neoplasms genetics, Kidney Neoplasms metabolism, Lipids biosynthesis
- Abstract
The glycerol-3-phosphate shuttle (G3PS) is a major NADH shuttle that regenerates reducing equivalents in the cytosol and produces energy in the mitochondria. Here, we demonstrate that G3PS is uncoupled in kidney cancer cells where the cytosolic reaction is ∼4.5 times faster than the mitochondrial reaction. The high flux through cytosolic glycerol-3-phosphate dehydrogenase (GPD) is required to maintain redox balance and support lipid synthesis. Interestingly, inhibition of G3PS by knocking down mitochondrial GPD (GPD2) has no effect on mitochondrial respiration. Instead, loss of GPD2 upregulates cytosolic GPD on a transcriptional level and promotes cancer cell proliferation by increasing glycerol-3-phosphate supply. The proliferative advantage of GPD2 knockdown tumor can be abolished by pharmacologic inhibition of lipid synthesis. Taken together, our results suggest that G3PS is not required to run as an intact NADH shuttle but is instead truncated to support complex lipid synthesis in kidney cancer., Competing Interests: Declaration of interests M.C.H. and A.H.S. received unrelated research funding from Roche Pharmaceuticals. M.C.H. received funding from Agilent Technologies. M.C.H. and A.H.S. are advisors to Alixia Therapeutics. A.H.S. is on advisory boards for Surface Oncology, SQZ Biotechnologies, Elpiscience, Selecta, Bicara and Monopteros, Bicara, and Fibrogen. She also is on scientific advisory boards for the Massachusetts General Cancer Center, Program in Cellular and Molecular Medicine at Boston Children’s Hospital, the Human Oncology and Pathogenesis Program at Memorial Sloan Kettering Cancer Center, Glaxo Smith Kline, and Janssen. She is an academic editor for the Journal of Experimental Medicine. A.H.S. has received unrelated funding from Novartis, Merck, AbbVie, Moderna, Vertex, and Erasca. M.C.H. is a Cell Metabolism and Molecular Cell advisory board member., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Oncometabolite d-2HG alters T cell metabolism to impair CD8 + T cell function.
- Author
-
Notarangelo G, Spinelli JB, Perez EM, Baker GJ, Kurmi K, Elia I, Stopka SA, Baquer G, Lin JR, Golby AJ, Joshi S, Baron HF, Drijvers JM, Georgiev P, Ringel AE, Zaganjor E, McBrayer SK, Sorger PK, Sharpe AH, Wucherpfennig KW, Santagata S, Agar NYR, Suvà ML, and Haigis MC
- Subjects
- Animals, Gain of Function Mutation, Humans, Interferon-gamma metabolism, L-Lactate Dehydrogenase antagonists & inhibitors, L-Lactate Dehydrogenase metabolism, Mice, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Carcinogenesis genetics, Carcinogenesis metabolism, Glutarates metabolism, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase metabolism, Neoplasms genetics, Neoplasms immunology, Neoplasms metabolism
- Abstract
Gain-of-function mutations in isocitrate dehydrogenase (IDH) in human cancers result in the production of d-2-hydroxyglutarate (d-2HG), an oncometabolite that promotes tumorigenesis through epigenetic alterations. The cancer cell-intrinsic effects of d-2HG are well understood, but its tumor cell-nonautonomous roles remain poorly explored. We compared the oncometabolite d-2HG with its enantiomer, l-2HG, and found that tumor-derived d-2HG was taken up by CD8
+ T cells and altered their metabolism and antitumor functions in an acute and reversible fashion. We identified the glycolytic enzyme lactate dehydrogenase (LDH) as a molecular target of d-2HG. d-2HG and inhibition of LDH drive a metabolic program and immune CD8+ T cell signature marked by decreased cytotoxicity and impaired interferon-γ signaling that was recapitulated in clinical samples from human patients with IDH1 mutant gliomas.- Published
- 2022
- Full Text
- View/download PDF
18. Tumor cells dictate anti-tumor immune responses by altering pyruvate utilization and succinate signaling in CD8 + T cells.
- Author
-
Elia I, Rowe JH, Johnson S, Joshi S, Notarangelo G, Kurmi K, Weiss S, Freeman GJ, Sharpe AH, and Haigis MC
- Subjects
- CD8-Positive T-Lymphocytes metabolism, Humans, Immunity, Lactic Acid, Pyruvate Carboxylase metabolism, Succinic Acid, Tumor Microenvironment, Neoplasms, Pyruvic Acid metabolism, Pyruvic Acid pharmacology
- Abstract
The tumor microenvironment (TME) is a unique metabolic niche that can inhibit T cell metabolism and cytotoxicity. To dissect the metabolic interplay between tumors and T cells, we establish an in vitro system that recapitulates the metabolic niche of the TME and allows us to define cell-specific metabolism. We identify tumor-derived lactate as an inhibitor of CD8
+ T cell cytotoxicity, revealing an unexpected metabolic shunt in the TCA cycle. Metabolically fit cytotoxic T cells shunt succinate out of the TCA cycle to promote autocrine signaling via the succinate receptor (SUCNR1). Cytotoxic T cells are reliant on pyruvate carboxylase (PC) to replenish TCA cycle intermediates. By contrast, lactate reduces PC-mediated anaplerosis. The inhibition of pyruvate dehydrogenase (PDH) is sufficient to restore PC activity, succinate secretion, and the activation of SUCNR1. These studies identify PDH as a potential drug target to allow CD8+ T cells to retain cytotoxicity and overcome a lactate-enriched TME., Competing Interests: Declaration of interests M.C.H. and A.H.S. received research funding from Roche Pharmaceuticals. M.C.H. received funding from Agilent Technologies. M.C.H. and A.H.S. are advisers to Guided Clarity., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
19. Intercellular nanotubes mediate mitochondrial trafficking between cancer and immune cells.
- Author
-
Saha T, Dash C, Jayabalan R, Khiste S, Kulkarni A, Kurmi K, Mondal J, Majumder PK, Bardia A, Jang HL, and Sengupta S
- Subjects
- Animals, Base Sequence, Cell Line, Tumor, Humans, Immunity, Mice, Inbred C57BL, Mitochondrial Proteins metabolism, Nanotubes ultrastructure, Mice, Leukocytes pathology, Mitochondria metabolism, Nanotubes chemistry, Neoplasms pathology
- Abstract
Cancer progresses by evading the immune system. Elucidating diverse immune evasion strategies is a critical step in the search for next-generation immunotherapies for cancer. Here we report that cancer cells can hijack the mitochondria from immune cells via physical nanotubes. Mitochondria are essential for metabolism and activation of immune cells. By using field-emission scanning electron microscopy, fluorophore-tagged mitochondrial transfer tracing and metabolic quantification, we demonstrate that the nanotube-mediated transfer of mitochondria from immune cells to cancer cells metabolically empowers the cancer cells and depletes the immune cells. Inhibiting the nanotube assembly machinery significantly reduced mitochondrial transfer and prevented the depletion of immune cells. Combining a farnesyltransferase and geranylgeranyltransferase 1 inhibitor, namely, L-778123, which partially inhibited nanotube formation and mitochondrial transfer, with a programmed cell death protein 1 immune checkpoint inhibitor improved the antitumour outcomes in an aggressive immunocompetent breast cancer model. Nanotube-mediated mitochondrial hijacking can emerge as a novel target for developing next-generation immunotherapy agents for cancer., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
20. Enzymatic activation of pyruvate kinase increases cytosolic oxaloacetate to inhibit the Warburg effect.
- Author
-
Wiese EK, Hitosugi S, Loa ST, Sreedhar A, Andres-Beck LG, Kurmi K, Pang YP, Karnitz LM, Gonsalves WI, and Hitosugi T
- Subjects
- Animals, Cell Line, Tumor, Cytosol metabolism, Enzyme Activation, Enzyme Inhibitors pharmacology, Gene Expression Regulation, Glucose metabolism, Glycolysis, Humans, Lactate Dehydrogenase 5 antagonists & inhibitors, Lactate Dehydrogenase 5 metabolism, Mice, Pyruvate Kinase genetics, Rabbits, Oxaloacetic Acid metabolism, Pyruvate Kinase metabolism
- Abstract
Pharmacological activation of the glycolytic enzyme PKM2 or expression of the constitutively active PKM1 isoform in cancer cells results in decreased lactate production, a phenomenon known as the PKM2 paradox in the Warburg effect. Here we show that oxaloacetate (OAA) is a competitive inhibitor of human lactate dehydrogenase A (LDHA) and that elevated PKM2 activity increases de novo synthesis of OAA through glutaminolysis, thereby inhibiting LDHA in cancer cells. We also show that replacement of human LDHA with rabbit LDHA, which is relatively resistant to OAA inhibition, eliminated the paradoxical correlation between the elevated PKM2 activity and the decreased lactate concentration in cancer cells treated with a PKM2 activator. Furthermore, rabbit LDHA-expressing tumours, compared to human LDHA-expressing tumours in mice, displayed resistance to the PKM2 activator. These findings describe a mechanistic explanation for the PKM2 paradox by showing that OAA accumulates and inhibits LDHA following PKM2 activation., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
21. Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8 + T Cells.
- Author
-
Drijvers JM, Gillis JE, Muijlwijk T, Nguyen TH, Gaudiano EF, Harris IS, LaFleur MW, Ringel AE, Yao CH, Kurmi K, Juneja VR, Trombley JD, Haigis MC, and Sharpe AH
- Subjects
- Animals, Autophagy drug effects, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Line, Tumor, Endoplasmic Reticulum drug effects, Female, Ferroptosis genetics, Humans, Mice, Mice, Inbred C57BL, Neoplasms drug therapy, Antineoplastic Agents pharmacology, CD8-Positive T-Lymphocytes drug effects, Ferroptosis drug effects, Tumor Cells, Cultured drug effects
- Abstract
Metabolic constraints in the tumor microenvironment constitute a barrier to effective antitumor immunity and similarities in the metabolic properties of T cells and cancer cells impede the specific therapeutic targeting of metabolism in either population. To identify distinct metabolic vulnerabilities of CD8
+ T cells and cancer cells, we developed a high-throughput in vitro pharmacologic screening platform and used it to measure the cell type-specific sensitivities of activated CD8+ T cells and B16 melanoma cells to a wide array of metabolic perturbations during antigen-specific killing of cancer cells by CD8+ T cells. We illustrated the applicability of this screening platform by showing that CD8+ T cells were more sensitive to ferroptosis induction by inhibitors of glutathione peroxidase 4 (GPX4) than B16 and MC38 cancer cells. Overexpression of ferroptosis suppressor protein 1 (FSP1) or cytosolic GPX4 yielded ferroptosis-resistant CD8+ T cells without compromising their function, while genetic deletion of the ferroptosis sensitivity-promoting enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) protected CD8+ T cells from ferroptosis but impaired antitumor CD8+ T-cell responses. Our screen also revealed high T cell-specific vulnerabilities for compounds targeting NAD+ metabolism or autophagy and endoplasmic reticulum (ER) stress pathways. We focused the current screening effort on metabolic agents. However, this in vitro screening platform may also be valuable for rapid testing of other types of compounds to identify regulators of antitumor CD8+ T-cell function and potential therapeutic targets., (©2020 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
22. Nitrogen Metabolism in Cancer and Immunity.
- Author
-
Kurmi K and Haigis MC
- Subjects
- Animals, Humans, Purines metabolism, Pyrimidines metabolism, Tumor Microenvironment, Immunity, Neoplasms metabolism, Nitrogen metabolism
- Abstract
As one of the fundamental requirements for cell growth and proliferation, nitrogen acquisition and utilization must be tightly regulated. Nitrogen can be generated from amino acids (AAs) and utilized for biosynthetic processes through transamination and deamination reactions. Importantly, limitations of nitrogen availability in cells can disrupt the synthesis of proteins, nucleic acids, and other important nitrogen-containing compounds. Rewiring cellular metabolism to support anabolic processes is a feature common to both cancer and proliferating immune cells. In this review, we discuss how nitrogen is utilized in biosynthetic pathways and highlight different metabolic and oncogenic programs that alter the flow of nitrogen to sustain biomass production and growth, an important emerging feature of cancer and immune cell proliferation., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
23. BRCA1 Deficiency Upregulates NNMT, Which Reprograms Metabolism and Sensitizes Ovarian Cancer Cells to Mitochondrial Metabolic Targeting Agents.
- Author
-
Kanakkanthara A, Kurmi K, Ekstrom TL, Hou X, Purfeerst ER, Heinzen EP, Correia C, Huntoon CJ, O'Brien D, Wahner Hendrickson AE, Dowdy SC, Li H, Oberg AL, Hitosugi T, Kaufmann SH, Weroha SJ, and Karnitz LM
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, BRCA1 Protein deficiency, Carcinoma, Ovarian Epithelial genetics, Carcinoma, Ovarian Epithelial pathology, Cell Line, Tumor, Cyclin-Dependent Kinases genetics, DNA Methylation, Energy Metabolism drug effects, Female, Gene Expression Regulation, Neoplastic, Humans, Hydrazones pharmacology, Hydrazones therapeutic use, Hydroxybenzoates pharmacology, Hydroxybenzoates therapeutic use, Mice, Mitochondria drug effects, Mitochondria metabolism, Mutation, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Ovary pathology, Oxidative Phosphorylation drug effects, Promoter Regions, Genetic genetics, Tigecycline pharmacology, Tigecycline therapeutic use, Triazoles pharmacology, Triazoles therapeutic use, Up-Regulation, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, BRCA1 Protein genetics, Carcinoma, Ovarian Epithelial drug therapy, Nicotinamide N-Methyltransferase metabolism, Ovarian Neoplasms drug therapy
- Abstract
BRCA1 plays a key role in homologous recombination (HR) DNA repair. Accordingly, changes that downregulate BRCA1, including BRCA1 mutations and reduced BRCA1 transcription, due to promoter hypermethylation or loss of the BRCA1 transcriptional regulator CDK12, disrupt HR in multiple cancers. In addition, BRCA1 has also been implicated in the regulation of metabolism. Here, we show that reducing BRCA1 expression, either by CDK12 or BRCA1 depletion, led to metabolic reprogramming of ovarian cancer cells, causing decreased mitochondrial respiration and reduced ATP levels. BRCA1 depletion drove this reprogramming by upregulating nicotinamide N-methyltransferase (NNMT). Notably, the metabolic alterations caused by BRCA1 depletion and NNMT upregulation sensitized ovarian cancer cells to agents that inhibit mitochondrial metabolism (VLX600 and tigecycline) and to agents that inhibit glucose import (WZB117). These observations suggest that inhibition of energy metabolism may be a potential strategy to selectively target BRCA1-deficient high-grade serous ovarian cancer, which is characterized by frequent BRCA1 loss and NNMT overexpression. SIGNIFICANCE: Loss of BRCA1 reprograms metabolism, creating a therapeutically targetable vulnerability in ovarian cancer., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
24. Tyrosine Phosphorylation of Mitochondrial Creatine Kinase 1 Enhances a Druggable Tumor Energy Shuttle Pathway.
- Author
-
Kurmi K, Hitosugi S, Yu J, Boakye-Agyeman F, Wiese EK, Larson TR, Dai Q, Machida YJ, Lou Z, Wang L, Boughey JC, Kaufmann SH, Goetz MP, Karnitz LM, and Hitosugi T
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Cell Transformation, Neoplastic, Creatine Kinase genetics, Creatinine analogs & derivatives, Creatinine therapeutic use, Energy Transfer, Female, Gene Knockdown Techniques, Humans, Lapatinib therapeutic use, Mice, Mice, Nude, Mitochondrial Proteins metabolism, Phosphocreatine metabolism, Phosphorylation, Trastuzumab therapeutic use, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms metabolism, Creatine Kinase metabolism, Drug Resistance, Neoplasm, Energy Metabolism, Mitochondria metabolism, Receptor, ErbB-2 metabolism
- Abstract
How mitochondrial metabolism is altered by oncogenic tyrosine kinases to promote tumor growth is incompletely understood. Here, we show that oncogenic HER2 tyrosine kinase signaling induces phosphorylation of mitochondrial creatine kinase 1 (MtCK1) on tyrosine 153 (Y153) in an ABL-dependent manner in breast cancer cells. Y153 phosphorylation, which is commonly upregulated in HER2
+ breast cancers, stabilizes MtCK1 to increase the phosphocreatine energy shuttle and promote proliferation. Inhibition of the phosphocreatine energy shuttle by MtCK1 knockdown or with the creatine analog cyclocreatine decreases proliferation of trastuzumab-sensitive and -resistant HER2+ cell lines in culture and in xenografts. Finally, we show that cyclocreatine in combination with the HER2 kinase inhibitor lapatinib reduces the growth of a trastuzumab-resistant HER2+ patient-derived xenograft. These findings suggest that activation of the phosphocreatine energy shuttle by MtCK1 Y153 phosphorylation creates a druggable metabolic vulnerability in cancer., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
25. Impaired β-cell glucokinase as an underlying mechanism in diet-induced diabetes.
- Author
-
Lu B, Kurmi K, Munoz-Gomez M, Jacobus Ambuludi EJ, Tonne JM, Rakshit K, Hitosugi T, Kudva YC, Matveyenko AV, and Ikeda Y
- Subjects
- Animals, Calcium metabolism, Cell Proliferation, Dependovirus metabolism, Diabetes Mellitus, Experimental genetics, Diet, High-Fat, Glucose metabolism, Glucose Tolerance Test, Glycolysis, Insulin metabolism, Intracellular Space metabolism, Male, Mice, Inbred C57BL, Signal Transduction, Transduction, Genetic, Up-Regulation genetics, Diabetes Mellitus, Experimental enzymology, Diabetes Mellitus, Experimental pathology, Glucokinase metabolism, Insulin-Secreting Cells enzymology, Insulin-Secreting Cells pathology
- Abstract
High-fat diet (HFD)-fed mouse models have been widely used to study early type 2 diabetes. Decreased β-cell glucokinase (GCK) expression has been observed in HFD-induced diabetes. However, owing to its crucial roles in glucose metabolism in the liver and in islet β-cells, the contribution of decreased GCK expression to the development of HFD-induced diabetes is unclear. Here, we employed a β-cell-targeted gene transfer vector and determined the impact of β-cell-specific increase in GCK expression on β-cell function and glucose handling in vitro and in vivo Overexpression of GCK enhanced glycolytic flux, ATP-sensitive potassium channel activation and membrane depolarization, and increased proliferation in Min6 cells. β-cell-targeted GCK transduction did not change glucose handling in chow-fed C57BL/6 mice. Although adult mice fed a HFD showed reduced islet GCK expression, impaired glucose tolerance and decreased glucose-stimulated insulin secretion (GSIS), β-cell-targeted GCK transduction improved glucose tolerance and restored GSIS. Islet perifusion experiments verified restored GSIS in isolated HFD islets by GCK transduction. Thus, our data identify impaired β-cell GCK expression as an underlying mechanism for dysregulated β-cell function and glycemic control in HFD-induced diabetes. Our data also imply an etiological role of GCK in diet-induced diabetes.This article has an associated First Person interview with the first author of the paper., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2018. Published by The Company of Biologists Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
26. Carnitine Palmitoyltransferase 1A Has a Lysine Succinyltransferase Activity.
- Author
-
Kurmi K, Hitosugi S, Wiese EK, Boakye-Agyeman F, Gonsalves WI, Lou Z, Karnitz LM, Goetz MP, and Hitosugi T
- Subjects
- Animals, Humans, Carnitine O-Palmitoyltransferase metabolism, Lysine metabolism, Protein Processing, Post-Translational physiology
- Abstract
Lysine succinylation was recently identified as a post-translational modification in cells. However, the molecular mechanism underlying lysine succinylation remains unclear. Here, we show that carnitine palmitoyltransferase 1A (CPT1A) has lysine succinyltransferase (LSTase) activity in vivo and in vitro. Using a stable isotope labeling by amino acid in cell culture (SILAC)-based proteomics approach, we found that 101 proteins were more succinylated in cells expressing wild-type (WT) CPT1A compared with vector control cells. One of the most heavily succinylated proteins in this analysis was enolase 1. We found that CPT1A WT succinylated enolase 1 and reduced enolase enzymatic activity in cells and in vitro. Importantly, mutation of CPT1A Gly710 (G710E) selectively inactivated carnitine palmitoyltransferase (CPTase) activity but not the LSTase activity that decreased enolase activity in cells and promoted cell proliferation under glutamine depletion. These findings suggest that CPT1A acts as an LSTase that can regulate enzymatic activity of a substrate protein and metabolism independent of its classical CPTase activity., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. Circulating Mycobacterium bovis peptides and host response proteins as biomarkers for unambiguous detection of subclinical infection.
- Author
-
Lamont EA, Janagama HK, Ribeiro-Lima J, Vulchanova L, Seth M, Yang M, Kurmi K, Waters WR, Thacker T, and Sreevatsan S
- Subjects
- Animals, Bacterial Proteins blood, Blood Chemical Analysis, Cattle, Latent Tuberculosis diagnosis, Proteome analysis, Sensitivity and Specificity, Vitamin D-Binding Protein blood, Biomarkers blood, Clinical Laboratory Techniques methods, Latent Tuberculosis veterinary, Mycobacterium bovis chemistry, Peptides blood, Tuberculosis, Bovine diagnosis, Veterinary Medicine methods
- Abstract
Bovine tuberculosis remains one of the most damaging diseases to agriculture, and there is also a concern for human spillover. A critical need exists for rapid, thorough, and inexpensive diagnostic methods capable of detecting and differentiating Mycobacterium bovis infection from other pathogenic and environmental mycobacteria at multiple surveillance levels. In a previous study, Seth et al. (PLoS One 4:e5478, 2009, doi:10.1371/journal.pone.0005478) identified 32 host peptides that specifically increased in the blood serum of M. bovis-infected animals). In the current study, 16 M. bovis proteins were discovered in the blood serum proteomics data sets. A large-scale validation analysis was undertaken for selected host and M. bovis proteins using a cattle serum repository containing M. bovis (n = 128), Mycobacterium kansasii (n = 10), and Mycobacterium avium subsp. paratuberculosis (n = 10), cases exposed to M. bovis (n = 424), and negative controls (n = 38). Of the host biomarkers, vitamin D binding protein (VDBP) showed the greatest sensitivity and specificity for M. bovis detection. Circulating M. bovis proteins, specifically polyketide synthetase 5, detected M. bovis-infected cattle with little to no seroreactivity against M. kansasii- and M. avium subsp. paratuberculosis-infected animals. These data indicate that host and pathogen serum proteins can serve as reliable biomarkers for tracking M. bovis infection in animal populations.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.