Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Redox Biol ; 73: 103220, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38838551

ABSTRACT

Temozolomide (TMZ) is a widely utilized chemotherapy treatment for patients with glioblastoma (GBM), although drug resistance constitutes a major therapeutic hurdle. Emerging evidence suggests that ferroptosis-mediated therapy could offer an appropriate alternative treatment option against cancer cells that are resistant to certain drugs. However, recurrent gliomas display robust ferroptosis resistance, although the precise mechanism of resistance remains elusive. In the present work, we report that proline rich protein 11 (PRR11) depletion significantly sensitizes GBM cells to TMZ by inducing ferroptosis. Mechanistically, PRR11 directly binds to and stabilizes dihydroorotate dehydrogenase (DHODH), which leads to glioma ferroptosis-resistant in a DHODH-dependent manner in vivo and in vitro. Furthermore, PRR11 inhibits HERC4 and DHODH binding, by suppressing the recruitment of E3 ubiquitin ligase HERC4 and polyubiquitination degradation of DHODH at the K306 site, which maintains DHODH protein stability. Importantly, downregulated PRR11 increases lipid peroxidation and alters DHODH-mediated mitochondrial morphology, thereby promoting ferroptosis and increasing TMZ chemotherapy sensitivity. In conclusion, our results reveal a mechanism via which PRR11 drives ferroptosis resistance and identifies ferroptosis induction and TMZ as an attractive combined therapeutic strategy for GBM.


Subject(s)
Dihydroorotate Dehydrogenase , Drug Resistance, Neoplasm , Ferroptosis , Glioblastoma , Temozolomide , Humans , Ferroptosis/drug effects , Ferroptosis/genetics , Glioblastoma/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Temozolomide/pharmacology , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Mice , Dihydroorotate Dehydrogenase/metabolism , Animals , Gene Expression Regulation, Neoplastic/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics
2.
Redox Biol ; 73: 103207, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38805974

ABSTRACT

Although 5-fluorouracil (5-FU) is the primary chemotherapy treatment for colorectal cancer (CRC), its efficacy is limited by drug resistance. Ferroptosis activation is a promising treatment for 5-FU-resistant cancer cells; however, potential therapeutic targets remain elusive. This study investigated ferroptosis vulnerability and dihydroorotate dehydrogenase (DHODH) activity using stable, 5-FU-resistant CRC cell lines and xenograft models. Ferroptosis was characterized by measuring malondialdehyde levels, assessing lipid metabolism and peroxidation, and using mitochondrial imaging and assays. DHODH function is investigated through gene knockdown experiments, tumor behavior assays, mitochondrial import reactions, intramitochondrial localization, enzymatic activity analyses, and metabolomics assessments. Intracellular lipid accumulation and mitochondrial DHODH deficiency led to lipid peroxidation overload, weakening the defense system of 5-FU-resistant CRC cells against ferroptosis. DHODH, primarily located within the inner mitochondrial membrane, played a crucial role in driving intracellular pyrimidine biosynthesis and was redistributed to the cytosol in 5-FU-resistant CRC cells. Cytosolic DHODH, like its mitochondrial counterpart, exhibited dihydroorotate catalytic activity and participated in pyrimidine biosynthesis. This amplified intracellular pyrimidine pools, thereby impeding the efficacy of 5-FU treatment through molecular competition. These findings contribute to the understanding of 5-FU resistance mechanisms and suggest that ferroptosis and DHODH are promising therapeutic targets for patients with CRC exhibiting resistance to 5-FU.


Subject(s)
Colorectal Neoplasms , Dihydroorotate Dehydrogenase , Drug Resistance, Neoplasm , Fluorouracil , Mitochondria , Oxidoreductases Acting on CH-CH Group Donors , Dihydroorotate Dehydrogenase/metabolism , Fluorouracil/pharmacology , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Mitochondria/metabolism , Mitochondria/drug effects , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Mice , Animals , Cell Line, Tumor , Xenograft Model Antitumor Assays , Lipid Peroxidation/drug effects
3.
Biomed Pharmacother ; 166: 115314, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37579695

ABSTRACT

Although all-trans retinoic acid (ATRA)-induced differentiation has transformed acute promyelocytic leukemia (APL) from the most fatal to the most curable hematological disease, resistance to ATRA in high-risk APL patients remains a clinical challenge. In this paper, we discovered that dihydroorotate dehydrogenase (DHODH) inhibition overcame ATRA resistance. 416, a potent DHODH inhibitor previously obtained in our group, inhibited the occurrence of APL in cells and model mice. Excitingly, 416 effectively overcame ATRA resistance in vitro and in vivo by inducing apoptosis and differentiation. Further mechanistic studies showed that PML/RARα lost the regulation of Bcl-2 and c-Myc in NB4-R1 cells, which probably contributed to ATRA resistance. Notably, 416 maintained its Bcl-2 and c-Myc down-regulation effect in NB4-R1 cells and overcome ATRA resistance by inhibiting DHODH. In conclusion, our study highlights the potential of 416 for APL therapy and overcoming ATRA resistance, supporting the further development of DHODH inhibitors for clinical use in refractory and relapsed APL.


Subject(s)
Antineoplastic Agents , Dihydroorotate Dehydrogenase , Drug Resistance, Neoplasm , Leukemia, Promyelocytic, Acute , Tretinoin , Animals , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Differentiation , Dihydroorotate Dehydrogenase/antagonists & inhibitors , Dihydroorotate Dehydrogenase/genetics , Dihydroorotate Dehydrogenase/metabolism , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Tretinoin/pharmacology , Tretinoin/therapeutic use
4.
Int J Mol Sci ; 23(5)2022 Feb 23.
Article in English | MEDLINE | ID: mdl-35269583

ABSTRACT

The fourth enzymatic reaction in the de novo pyrimidine biosynthesis, the oxidation of dihydroorotate to orotate, is catalyzed by dihydroorotate dehydrogenase (DHODH). Enzymes belonging to the DHODH Class II are membrane-bound proteins that use ubiquinones as their electron acceptors. We have designed this study to understand the interaction of an N-terminally truncated human DHODH (HsΔ29DHODH) and the DHODH from Escherichia coli (EcDHODH) with ubiquinone (Q10) in supported lipid membranes using neutron reflectometry (NR). NR has allowed us to determine in situ, under solution conditions, how the enzymes bind to lipid membranes and to unambiguously resolve the location of Q10. Q10 is exclusively located at the center of all of the lipid bilayers investigated, and upon binding, both of the DHODHs penetrate into the hydrophobic region of the outer lipid leaflet towards the Q10. We therefore show that the interaction between the soluble enzymes and the membrane-embedded Q10 is mediated by enzyme penetration. We can also show that EcDHODH binds more efficiently to the surface of simple bilayers consisting of 1-palmitoyl, 2-oleoyl phosphatidylcholine, and tetraoleoyl cardiolipin than HsΔ29DHODH, but does not penetrate into the lipids to the same degree. Our results also highlight the importance of Q10, as well as lipid composition, on enzyme binding.


Subject(s)
Dihydroorotate Dehydrogenase/chemistry , Dihydroorotate Dehydrogenase/metabolism , Escherichia coli/enzymology , Lipid Bilayers/metabolism , Ubiquinone/metabolism , Cardiolipins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Phosphatidylcholines/metabolism , Protein Conformation , Protein Domains
5.
Science ; 374(6572): 1227-1237, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34855504

ABSTRACT

For electrons to continuously enter and flow through the mitochondrial electron transport chain (ETC), they must ultimately land on a terminal electron acceptor (TEA), which is known to be oxygen in mammals. Paradoxically, we find that complex I and dihydroorotate dehydrogenase (DHODH) can still deposit electrons into the ETC when oxygen reduction is impeded. Cells lacking oxygen reduction accumulate ubiquinol, driving the succinate dehydrogenase (SDH) complex in reverse to enable electron deposition onto fumarate. Upon inhibition of oxygen reduction, fumarate reduction sustains DHODH and complex I activities. Mouse tissues display varying capacities to use fumarate as a TEA, most of which net reverse the SDH complex under hypoxia. Thus, we delineate a circuit of electron flow in the mammalian ETC that maintains mitochondrial functions under oxygen limitation.


Subject(s)
Electron Transport , Electrons , Fumarates/metabolism , Animals , Cell Hypoxia , Cell Line , Cell Line, Tumor , Dihydroorotate Dehydrogenase/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidation-Reduction , Oxygen/metabolism , Succinate Dehydrogenase/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism
6.
Front Immunol ; 12: 730342, 2021.
Article in English | MEDLINE | ID: mdl-34721394

ABSTRACT

Background and Objectives: Inhibition of de novo pyrimidine synthesis in proliferating T and B lymphocytes by teriflunomide, a pharmacological inhibitor of dihydroorotate dehydrogenase (DHODH), has been shown to be an effective therapy to treat patients with MS in placebo-controlled phase 3 trials. Nevertheless, the underlying mechanism contributing to the efficacy of DHODH inhibition has been only partially elucidated. Here, we aimed to determine the impact of teriflunomide on the immune compartment in a longitudinal high-dimensional follow-up of patients with relapse-remitting MS (RRMS) treated with teriflunomide. Methods: High-dimensional spectral flow cytometry was used to analyze the phenotype and the function of innate and adaptive immune system of patients with RRMS before and 12 months after teriflunomide treatment. In addition, we assessed the impact of teriflunomide on the migration of memory CD8 T cells in patients with RRMS, and we defined patient immune metabolic profiles. Results: We found that 12 months of treatment with teriflunomide in patients with RRMS does not affect the B cell or CD4 T cell compartments, including regulatory TREG follicular helper TFH cell and helper TH cell subsets. In contrast, we observed a specific impact of teriflunomide on the CD8 T cell compartment, which was characterized by decreased homeostatic proliferation and reduced production of TNFα and IFNγ. Furthermore, we showed that DHODH inhibition also had a negative impact on the migratory velocity of memory CD8 T cells in patients with RRMS. Finally, we showed that the susceptibility of memory CD8 T cells to DHODH inhibition was not related to impaired metabolism. Discussion: Overall, these findings demonstrate that the clinical efficacy of teriflunomide results partially in the specific susceptibility of memory CD8 T cells to DHODH inhibition in patients with RRMS and strengthens active roles for these T cells in the pathophysiological process of MS.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Crotonates/therapeutic use , Dihydroorotate Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Hydroxybutyrates/therapeutic use , Immunologic Memory/drug effects , Immunosuppressive Agents/therapeutic use , Memory T Cells/drug effects , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Nitriles/therapeutic use , Toluidines/therapeutic use , Adult , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Crotonates/adverse effects , Dihydroorotate Dehydrogenase/metabolism , Enzyme Inhibitors/adverse effects , Female , Humans , Hydroxybutyrates/adverse effects , Immunosuppressive Agents/adverse effects , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Male , Memory T Cells/enzymology , Memory T Cells/immunology , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/enzymology , Multiple Sclerosis, Relapsing-Remitting/immunology , Nitriles/adverse effects , Phenotype , Time Factors , Toluidines/adverse effects , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism
7.
J Med Chem ; 64(18): 13918-13932, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34516133

ABSTRACT

Human dihydroorotate dehydrogenase (hDHODH) is an attractive tumor target essential to de novo pyrimidine biosynthesis. Novel potent hDHODH inhibitors with low toxicity are urgently needed. Herein, we demonstrate the isolation of 25 ascochlorin (ASC) derivatives, including 13 new ones, from the coral-derived fungus Acremonium sclerotigenum, and several of them showed pronounced inhibitions against hDHODH and triple-negative breast cancer (TNBC) cell lines, MDA-MB-231/-468. Interestingly, we found that hDHODH is required for proliferation and survival of TNBC cells, and several ASCs significantly inhibited TNBC cell growth and induced their apoptosis via hDHODH inhibition. Furthermore, the novel and potent hDHODH inhibitors (1 and 21) efficiently suppressed tumor growth in patient-derived TNBC xenograft models without obvious body weight loss or overt toxicity in mice. Collectively, our findings offered a novel lead scaffold as the hDHODH inhibitor for further development of potent anticancer agents and a potential therapeutic strategy for TNBC.


Subject(s)
Alkenes/therapeutic use , Antineoplastic Agents/therapeutic use , Dihydroorotate Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Phenols/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Acremonium/chemistry , Alkenes/chemistry , Alkenes/isolation & purification , Alkenes/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dihydroorotate Dehydrogenase/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/metabolism , Humans , Male , Mice, Inbred NOD , Mice, SCID , Molecular Structure , Phenols/chemistry , Phenols/isolation & purification , Phenols/metabolism , Structure-Activity Relationship , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Cell Death Dis ; 12(9): 821, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34462431

ABSTRACT

Metabolic reprogramming is an integral part of the growth-promoting program driven by the MYC family of oncogenes. However, this reprogramming also imposes metabolic dependencies that could be exploited therapeutically. Here we report that the pyrimidine biosynthetic enzyme dihydroorotate dehydrogenase (DHODH) is an attractive therapeutic target for MYCN-amplified neuroblastoma, a childhood cancer with poor prognosis. Gene expression profiling and metabolomic analysis reveal that MYCN promotes pyrimidine nucleotide production by transcriptional upregulation of DHODH and other enzymes of the pyrimidine-synthesis pathway. Genetic and pharmacological inhibition of DHODH suppresses the proliferation and tumorigenicity of MYCN-amplified neuroblastoma cell lines. Furthermore, we obtain evidence suggesting that serum uridine is a key factor in determining the efficacy of therapeutic agents that target DHODH. In the presence of physiological concentrations of uridine, neuroblastoma cell lines are highly resistant to DHODH inhibition. This uridine-dependent resistance to DHODH inhibitors can be abrogated by dipyridamole, an FDA-approved drug that blocks nucleoside transport. Importantly, dipyridamole synergizes with DHODH inhibition to suppress neuroblastoma growth in animal models. These findings suggest that a combination of targeting DHODH and nucleoside transport is a promising strategy to overcome intrinsic resistance to DHODH-based cancer therapeutics.


Subject(s)
Dihydroorotate Dehydrogenase/metabolism , Gene Amplification , Molecular Targeted Therapy , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Nucleosides/metabolism , Animals , Biological Transport/drug effects , Biphenyl Compounds/pharmacology , Carbazoles/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Amplification/drug effects , Humans , Male , Mice, Inbred NOD , Mice, SCID , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/blood , Neuroblastoma/pathology , Transcription, Genetic/drug effects , Uridine/blood
9.
Curr Top Med Chem ; 21(23): 2134-2154, 2021.
Article in English | MEDLINE | ID: mdl-34348625

ABSTRACT

BACKGROUND: Dihydroorotate dehydrogenase (DHODH) has long been recognized as an important drug target for proliferative and parasitic diseases, including compounds that exhibit trypanocidal action and broad-spectrum antiviral activity. Despite numerous and successful efforts in structural and functional characterization of DHODHs, as well as in the development of inhibitors, DHODH hot spots remain largely unmapped and underexplored. OBJECTIVE: This review describes the tools that are currently available for the identification and characterization of hot spots in protein structures and how freely available webservers can be exploited to predict DHODH hot spots. Moreover, it provides for the first time a review of the antiviral properties of DHODH inhibitors. METHODS: X-ray structures from human (HsDHODH) and Trypanosoma cruzi DHODH (TcDHODH) had their hot spots predicted by both FTMap and Fragment Hotspot Maps web servers. RESULTS: FTMap showed that hot spot occupancy in HsDHODH is correlated with the ligand efficiency (LE) of its known inhibitors, and Fragment Hotspot Maps pointed out the contribution of selected moieties to the overall LE. The conformational flexibility of the active site loop in TcDHODH was found to have a major impact on the druggability of the orotate binding site. In addition, both FTMap and Fragment Hotspot Maps servers predict a novel pocket in TcDHODH dimer interface (S6 site). CONCLUSION: This review reports how hot spots can be exploited during hit-to-lead steps, docking studies or even to improve inhibitor binding profile and by doing so using DHODH as a model, points to new drug development opportunities.


Subject(s)
Dihydroorotate Dehydrogenase/antagonists & inhibitors , Dihydroorotate Dehydrogenase/chemistry , Drug Development/trends , Antiviral Agents , Dihydroorotate Dehydrogenase/metabolism , Humans , Trypanosoma cruzi/enzymology
10.
Bioorg Med Chem Lett ; 46: 128194, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34116160

ABSTRACT

Twenty novel 2-substituted quinoline-4-carboxylic acids bearing amide moiety were designed and synthesized by Doebner reaction. Human dihydroorotate dehydrogenase (hDHODH) was recognized as a biological target and all compounds were screened as potential hDHODH inhibitors in an enzyme inhibition assay. The prepared heterocycles were also evaluated for their cytotoxic effects on the healthy HaCaT cell line while lipophilic properties were considered on the basis of experimentally determined logD values at physiological pH. The most promising compound 5j, with chlorine at para-position of terminal phenyl ring, showed good hDHODH inhibitory activity, low cytotoxicity, and optimal lipophilicity. The bioactive conformation of 5j on the hDHODH, determined by means of molecular docking, revealed the compound's pharmacology and provide guidelines for further lead optimization.


Subject(s)
Antineoplastic Agents/pharmacology , Benzaldehydes/chemistry , Dihydroorotate Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Quinolines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line , Cell Survival/drug effects , Dihydroorotate Dehydrogenase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Quinolines/chemistry , Structure-Activity Relationship
11.
Nature ; 593(7860): 586-590, 2021 05.
Article in English | MEDLINE | ID: mdl-33981038

ABSTRACT

Ferroptosis, a form of regulated cell death that is induced by excessive lipid peroxidation, is a key tumour suppression mechanism1-4. Glutathione peroxidase 4 (GPX4)5,6 and ferroptosis suppressor protein 1 (FSP1)7,8 constitute two major ferroptosis defence systems. Here we show that treatment of cancer cells with GPX4 inhibitors results in acute depletion of N-carbamoyl-L-aspartate, a pyrimidine biosynthesis intermediate, with concomitant accumulation of uridine. Supplementation with dihydroorotate or orotate-the substrate and product of dihydroorotate dehydrogenase (DHODH)-attenuates or potentiates ferroptosis induced by inhibition of GPX4, respectively, and these effects are particularly pronounced in cancer cells with low expression of GPX4 (GPX4low). Inactivation of DHODH induces extensive mitochondrial lipid peroxidation and ferroptosis in GPX4low cancer cells, and synergizes with ferroptosis inducers to induce these effects in GPX4high cancer cells. Mechanistically, DHODH operates in parallel to mitochondrial GPX4 (but independently of cytosolic GPX4 or FSP1) to inhibit ferroptosis in the mitochondrial inner membrane by reducing ubiquinone to ubiquinol (a radical-trapping antioxidant with anti-ferroptosis activity). The DHODH inhibitor brequinar selectively suppresses GPX4low tumour growth by inducing ferroptosis, whereas combined treatment with brequinar and sulfasalazine, an FDA-approved drug with ferroptosis-inducing activity, synergistically induces ferroptosis and suppresses GPX4high tumour growth. Our results identify a DHODH-mediated ferroptosis defence mechanism in mitochondria and suggest a therapeutic strategy of targeting ferroptosis in cancer treatment.


Subject(s)
Dihydroorotate Dehydrogenase/metabolism , Ferroptosis , Mitochondria/metabolism , Neoplasms/enzymology , Animals , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Dihydroorotate Dehydrogenase/genetics , Female , Gene Deletion , Humans , Lipid Peroxidation , Metabolomics , Mice, Nude , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Xenograft Model Antitumor Assays
12.
Stem Cells ; 39(1): 33-42, 2021 01.
Article in English | MEDLINE | ID: mdl-33038285

ABSTRACT

Pluripotent stem cells (PSCs), such as embryonic stem cells and induced pluripotent stem cells, give rise to all kinds of functional cells, making them promising for successful application in regenerative medicine. However, there is concern that a PSC-derived differentiated cell population may form teratomas when used for cell therapy if the population contains undifferentiated PSCs. Therefore, for the success of regenerative medicine, it is crucial to establish methods that induce complete PSC differentiation and eliminate the contamination of PSCs. Here, I show that the dihydroorotate dehydrogenase (DHODH) inhibitor brequinar (BRQ) induced cell cycle arrest, cell death, and stemness loss in mouse PSCs (mPSCs), whereas it was less toxic against normal tissue-specific stem cells and differentiating cells. I demonstrate that BRQ-pretreated mPSCs did not form teratomas after being transplanted into NOD/SCID mice. Moreover, BRQ administration to teratoma-bearing mice prevented tumor growth and decreased PSC marker levels in the tumor without any visible effects in the differentiated germ layer cells and the mice. Collectively, these data suggested that DHODH inhibitors such as BRQ can be indispensable in the fundamental methods of PSC-based therapy.


Subject(s)
Biphenyl Compounds/pharmacology , Dihydroorotate Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mouse Embryonic Stem Cells/enzymology , Animals , Cell Line , Dihydroorotate Dehydrogenase/metabolism , Mice , Mice, Inbred NOD , Mice, SCID
SELECTION OF CITATIONS
SEARCH DETAIL
...