General Information of Drug Combination (ID: DCHPYIJ)

Drug Combination Name
Mirtazapine Methamphetamine
Indication
Disease Entry Status REF
Methamphetamine Use Disorder Phase 1 [1]
Component Drugs Mirtazapine   DML53ZJ Methamphetamine   DMPM4SK
Small molecular drug Small molecular drug
2D MOL 2D MOL
3D MOL 3D MOL

Molecular Interaction Atlas of This Drug Combination

Molecular Interaction Atlas (MIA)
Indication(s) of Mirtazapine
Disease Entry ICD 11 Status REF
Depression 6A70-6A7Z Approved [2]
Mirtazapine Interacts with 1 DTT Molecule(s)
DTT Name DTT ID UniProt ID Mode of Action REF
5-HT 2C receptor (HTR2C) TTWJBZ5 5HT2C_HUMAN Antagonist [6]
------------------------------------------------------------------------------------
Mirtazapine Interacts with 5 DME Molecule(s)
DME Name DME ID UniProt ID Mode of Action REF
Cytochrome P450 3A4 (CYP3A4) DE4LYSA CP3A4_HUMAN Metabolism [7]
Cytochrome P450 1A2 (CYP1A2) DEJGDUW CP1A2_HUMAN Metabolism [8]
Cytochrome P450 2D6 (CYP2D6) DECB0K3 CP2D6_HUMAN Metabolism [7]
Cytochrome P450 2C8 (CYP2C8) DES5XRU CP2C8_HUMAN Metabolism [9]
Cytochrome P450 2C9 (CYP2C9) DE5IED8 CP2C9_HUMAN Metabolism [9]
------------------------------------------------------------------------------------
Mirtazapine Interacts with 5 DOT Molecule(s)
DOT Name DOT ID UniProt ID Mode of Action REF
Stearoyl-CoA desaturase (SCD) OTB1073G SCD_HUMAN Increases Expression [10]
Prolactin (PRL) OTWFQGX7 PRL_HUMAN Increases Expression [11]
Solute carrier family 2, facilitated glucose transporter member 4 (SLC2A4) OTJZG0LD GLUT4_HUMAN Increases Expression [12]
Solute carrier family 2, facilitated glucose transporter member 5 (SLC2A5) OTZNYEKF GTR5_HUMAN Increases Expression [12]
Hydroxymethylglutaryl-CoA synthase, cytoplasmic (HMGCS1) OTCO26FV HMCS1_HUMAN Increases Expression [10]
------------------------------------------------------------------------------------
Indication(s) of Methamphetamine
Disease Entry ICD 11 Status REF
Anxiety N.A. Approved [3]
Attention deficit hyperactivity disorder 6A05.Z Approved [4]
Depression 6A70-6A7Z Approved [3]
Methamphetamine dependence 6C46.2 Approved [3]
Obesity 5B81 Approved [3]
Pain MG30-MG3Z Approved [5]
Sexually transmitted infection 1A9Z Approved [3]
Substance dependence N.A. Approved [3]
Methamphetamine Interacts with 2 DTT Molecule(s)
DTT Name DTT ID UniProt ID Mode of Action REF
Adrenergic receptor alpha-1B (ADRA1B) TTBRKXS ADA1B_HUMAN Antagonist [16]
Dopamine D2 receptor (D2R) TTEX248 DRD2_HUMAN Agonist [5]
------------------------------------------------------------------------------------
Methamphetamine Interacts with 1 DTP Molecule(s)
DTP Name DTP ID UniProt ID Mode of Action REF
Vesicular amine transporter 2 (SLC18A2) DTT7VPB VMAT2_HUMAN Substrate [17]
------------------------------------------------------------------------------------
Methamphetamine Interacts with 1 DME Molecule(s)
DME Name DME ID UniProt ID Mode of Action REF
Cytochrome P450 2D6 (CYP2D6) DECB0K3 CP2D6_HUMAN Metabolism [18]
------------------------------------------------------------------------------------
Methamphetamine Interacts with 169 DOT Molecule(s)
DOT Name DOT ID UniProt ID Mode of Action REF
Synaptic vesicular amine transporter (SLC18A2) OTUOMMM6 VMAT2_HUMAN Decreases Activity [19]
D(2) dopamine receptor (DRD2) OTBLXKEG DRD2_HUMAN Increases Response To Substance [20]
PRKCA-binding protein (PICK1) OT8QE6EU PICK1_HUMAN Increases Response To Substance [21]
Zeta-sarcoglycan (SGCZ) OTVGTEOD SGCZ_HUMAN Affects Response To Substance [15]
Cytosolic carboxypeptidase 4 (AGBL1) OT8NQLH0 CBPC4_HUMAN Increases ADR [22]
Catalase (CAT) OTHEBX9R CATA_HUMAN Decreases Expression [23]
Superoxide dismutase (SOD1) OT39TA1L SODC_HUMAN Increases Activity [24]
Metallothionein-2 (MT2A) OTHOACHD MT2_HUMAN Increases Expression [25]
Activator of apoptosis harakiri (HRK) OTR4GWJ0 HRK_HUMAN Increases Expression [26]
Angiopoietin-2 (ANGPT2) OTEQK65P ANGP2_HUMAN Increases Expression [26]
Baculoviral IAP repeat-containing protein 5 (BIRC5) OTILXZYL BIRC5_HUMAN Decreases Expression [26]
Cell division control protein 45 homolog (CDC45) OT6NNLOD CDC45_HUMAN Decreases Expression [26]
Retinal dehydrogenase 2 (ALDH1A2) OTJB560Z AL1A2_HUMAN Increases Expression [26]
BAI1-associated protein 3 (BAIAP3) OTS1H0J0 BAIP3_HUMAN Increases Expression [26]
Dickkopf-related protein 1 (DKK1) OTRDLUSP DKK1_HUMAN Increases Expression [26]
G2/mitotic-specific cyclin-B2 (CCNB2) OTIEXTDK CCNB2_HUMAN Decreases Expression [26]
Kinesin-like protein KIF20A (KIF20A) OTXOQHE0 KI20A_HUMAN Decreases Expression [26]
ATP-binding cassette sub-family C member 6 (ABCC6) OTZT0LKT MRP6_HUMAN Decreases Expression [26]
Keratin, type I cytoskeletal 14 (KRT14) OTUVZ1DW K1C14_HUMAN Increases Expression [26]
C-X-C motif chemokine 10 (CXCL10) OTTLQ6S0 CXL10_HUMAN Increases Expression [26]
Fatty acid-binding protein, heart (FABP3) OT562DA1 FABPH_HUMAN Increases Expression [26]
DNA topoisomerase 2-alpha (TOP2A) OT6LPS08 TOP2A_HUMAN Decreases Expression [26]
High mobility group protein HMG-I/HMG-Y (HMGA1) OTQUSHPX HMGA1_HUMAN Increases Expression [26]
C-X-C motif chemokine 2 (CXCL2) OTEJCYMY CXCL2_HUMAN Increases Expression [26]
Galanin peptides (GAL) OTB3VPTO GALA_HUMAN Increases Expression [26]
Ribonucleoside-diphosphate reductase subunit M2 (RRM2) OTOB6J6R RIR2_HUMAN Decreases Expression [26]
Prostaglandin G/H synthase 2 (PTGS2) OT75U9M4 PGH2_HUMAN Increases Expression [26]
Phospholipase A2 group V (PLA2G5) OTQRSKCZ PA2G5_HUMAN Increases Expression [26]
Thrombopoietin (THPO) OTO73DZ2 TPO_HUMAN Increases Expression [26]
DNA-binding protein inhibitor ID-1 (ID1) OTKGNZN5 ID1_HUMAN Decreases Expression [26]
Proliferation marker protein Ki-67 (MKI67) OTA8N1QI KI67_HUMAN Decreases Expression [26]
N-acylglucosamine 2-epimerase (RENBP) OT8P1FP6 RENBP_HUMAN Increases Expression [26]
Actin, alpha cardiac muscle 1 (ACTC1) OTJU04B1 ACTC_HUMAN Decreases Expression [26]
Mevalonate kinase (MVK) OTHJCUKT KIME_HUMAN Increases Expression [26]
Origin recognition complex subunit 1 (ORC1) OTHWU8IJ ORC1_HUMAN Decreases Expression [26]
Transmembrane glycoprotein NMB (GPNMB) OTB7KQKV GPNMB_HUMAN Increases Expression [26]
PCNA-associated factor (PCLAF) OTMVIOUU PAF15_HUMAN Decreases Expression [26]
Disks large-associated protein 5 (DLGAP5) OTWCN39U DLGP5_HUMAN Decreases Expression [26]
Centrosomal protein of 55 kDa (CEP55) OTGSG2PA CEP55_HUMAN Decreases Expression [26]
Borealin (CDCA8) OT17D55D BOREA_HUMAN Decreases Expression [26]
Solute carrier organic anion transporter family member 4C1 (SLCO4C1) OTVILUUN SO4C1_HUMAN Increases Expression [26]
Protein MCM10 homolog (MCM10) OTV0O3JN MCM10_HUMAN Decreases Expression [26]
Ras guanyl-releasing protein 3 (RASGRP3) OTEMEV3P GRP3_HUMAN Increases Expression [26]
Abnormal spindle-like microcephaly-associated protein (ASPM) OTKXQMNA ASPM_HUMAN Decreases Expression [26]
CD177 antigen (CD177) OTS79FNF CD177_HUMAN Increases Expression [26]
Holliday junction recognition protein (HJURP) OTWMV16B HJURP_HUMAN Decreases Expression [26]
SHC SH2 domain-binding protein 1 (SHCBP1) OT3A21CM SHCBP_HUMAN Decreases Expression [26]
Interferon-stimulated gene 20 kDa protein (ISG20) OTCWRJJW ISG20_HUMAN Increases Expression [26]
RAD51-associated protein 1 (RAD51AP1) OTXM7UTD R51A1_HUMAN Decreases Expression [26]
Lymphokine-activated killer T-cell-originated protein kinase (PBK) OT5Z27TW TOPK_HUMAN Decreases Expression [26]
Growth/differentiation factor 15 (GDF15) OTWQN50N GDF15_HUMAN Increases Expression [26]
Kinesin-like protein KIFC1 (KIFC1) OTNQDS00 KIFC1_HUMAN Decreases Expression [26]
Denticleless protein homolog (DTL) OTC29RYE DTL_HUMAN Decreases Expression [26]
Adherens junction-associated protein 1 (AJAP1) OTAAW121 AJAP1_HUMAN Increases Expression [26]
2-oxoglutarate dehydrogenase-like, mitochondrial (OGDHL) OT1AZK6R OGDHL_HUMAN Increases Expression [26]
Lymphocyte antigen 96 (LY96) OTN4HXA2 LY96_HUMAN Increases Expression [26]
Protein kinase C delta type (PRKCD) OTSEH90E KPCD_HUMAN Affects Localization [27]
Bcl-2-like protein 11 (BCL2L11) OTNQQWFJ B2L11_HUMAN Increases Expression [28]
E3 ubiquitin-protein ligase parkin (PRKN) OTJBN41W PRKN_HUMAN Decreases Expression [29]
Serine/threonine-protein kinase/endoribonuclease IRE1 (ERN1) OTY9R6FZ ERN1_HUMAN Increases Phosphorylation [13]
Prolactin (PRL) OTWFQGX7 PRL_HUMAN Affects Expression [30]
Tumor necrosis factor (TNF) OT4IE164 TNFA_HUMAN Increases Expression [31]
T-cell surface glycoprotein CD3 delta chain (CD3D) OTRBLP0R CD3D_HUMAN Increases Expression [32]
Amyloid-beta precursor protein (APP) OTKFD7R4 A4_HUMAN Increases Expression [33]
Transcription factor Jun (JUN) OTCYBO6X JUN_HUMAN Increases Activity [31]
Insulin receptor (INSR) OTTY341H INSR_HUMAN Decreases Phosphorylation [33]
Tyrosine 3-monooxygenase (TH) OT6ZORKP TY3H_HUMAN Decreases Expression [34]
Synaptophysin (SYP) OTFJKMO4 SYPH_HUMAN Decreases Expression [35]
Poly polymerase 1 (PARP1) OT310QSG PARP1_HUMAN Decreases Cleavage [36]
Apoptosis regulator Bcl-2 (BCL2) OT9DVHC0 BCL2_HUMAN Decreases Expression [28]
Microtubule-associated protein tau (MAPT) OTMTP2Z7 TAU_HUMAN Increases Phosphorylation [32]
Endoplasmic reticulum chaperone BiP (HSPA5) OTFUIRAO BIP_HUMAN Increases Expression [13]
Cyclic AMP-responsive element-binding protein 1 (CREB1) OT1MDLA1 CREB1_HUMAN Increases Expression [37]
Protein kinase C alpha type (PRKCA) OT5UWNRD KPCA_HUMAN Increases Expression [37]
Cyclic AMP-dependent transcription factor ATF-4 (ATF4) OTRFV19J ATF4_HUMAN Increases Expression [13]
Cyclic AMP-dependent transcription factor ATF-6 alpha (ATF6) OTAFHAVI ATF6A_HUMAN Increases Cleavage [13]
Troponin I, cardiac muscle (TNNI3) OT65E12V TNNI3_HUMAN Increases Expression [38]
Nuclear factor NF-kappa-B p105 subunit (NFKB1) OTNRRD8I NFKB1_HUMAN Increases Activity [31]
D(1A) dopamine receptor (DRD1) OTLZPBT7 DRD1_HUMAN Increases Expression [39]
Solute carrier family 2, facilitated glucose transporter member 5 (SLC2A5) OTZNYEKF GTR5_HUMAN Increases Expression [40]
NF-kappa-B inhibitor alpha (NFKBIA) OTFT924M IKBA_HUMAN Increases Degradation [41]
Mitogen-activated protein kinase 3 (MAPK3) OTCYKGKO MK03_HUMAN Affects Phosphorylation [33]
Choline O-acetyltransferase (CHAT) OTEJWQ45 CLAT_HUMAN Decreases Activity [42]
Mitogen-activated protein kinase 1 (MAPK1) OTH85PI5 MK01_HUMAN Affects Phosphorylation [33]
Sodium-dependent serotonin transporter (SLC6A4) OT6FGDLW SC6A4_HUMAN Decreases Expression [43]
D(3) dopamine receptor (DRD3) OT0OFFKB DRD3_HUMAN Decreases Expression [44]
Insulin receptor substrate 1 (IRS1) OTNO82CU IRS1_HUMAN Decreases Phosphorylation [33]
DNA damage-inducible transcript 3 protein (DDIT3) OTI8YKKE DDIT3_HUMAN Increases Expression [13]
Serine/threonine-protein kinase mTOR (MTOR) OTHH8KU7 MTOR_HUMAN Decreases Phosphorylation [45]
Caspase-3 (CASP3) OTIJRBE7 CASP3_HUMAN Decreases Cleavage [36]
Neuropeptide Y receptor type 2 (NPY2R) OTEWQJ6B NPY2R_HUMAN Increases Expression [46]
Tuberin (TSC2) OT47LWI9 TSC2_HUMAN Increases Phosphorylation [29]
Glycogen synthase kinase-3 alpha (GSK3A) OT0F6DWV GSK3A_HUMAN Affects Phosphorylation [33]
Glycogen synthase kinase-3 beta (GSK3B) OTL3L14B GSK3B_HUMAN Affects Phosphorylation [33]
BH3-interacting domain death agonist (BID) OTOSHSHU BID_HUMAN Increases Expression [28]
Cyclin-dependent kinase 5 (CDK5) OT1YAK9F CDK5_HUMAN Increases Expression [32]
Transcription factor p65 (RELA) OTUJP9CN TF65_HUMAN Affects Localization [41]
Apoptosis regulator BAX (BAX) OTAW0V4V BAX_HUMAN Increases Expression [28]
Bcl-2-like protein 1 (BCL2L1) OTRC5K9O B2CL1_HUMAN Decreases Expression [28]
Beclin-1 (BECN1) OT4X293M BECN1_HUMAN Increases Expression [29]
Bcl-2 homologous antagonist/killer (BAK1) OTDP6ILW BAK_HUMAN Increases Expression [28]
Inactive caspase-12 (CASP12) OTY2W6FG CASPC_HUMAN Increases Expression [32]
Bcl-2-binding component 3, isoforms 3/4 (BBC3) OTUAXDAY BBC3B_HUMAN Increases Expression [28]
Tribbles homolog 3 (TRIB3) OTG5OS7X TRIB3_HUMAN Increases Expression [29]
1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase beta-1 (PLCB1) OT9HYT7A PLCB1_HUMAN Increases Expression [37]
Eukaryotic translation initiation factor 2-alpha kinase 3 (EIF2AK3) OT0DZGY4 E2AK3_HUMAN Increases Phosphorylation [13]
Long-chain-fatty-acid--CoA ligase 6 (ACSL6) OT0TT8P8 ACSL6_HUMAN Affects Response To Substance [15]
Mu-type opioid receptor (OPRM1) OT16AAT8 OPRM_HUMAN Increases Response To Substance [47]
Flavin-containing monooxygenase 3 (FMO3) OT1G2EV3 FMO3_HUMAN Increases Metabolism [48]
3',5'-cyclic-AMP phosphodiesterase 4D (PDE4D) OT1RWFV0 PDE4D_HUMAN Affects Response To Substance [15]
Coiled-coil alpha-helical rod protein 1 (CCHCR1) OT22C116 CCHCR_HUMAN Affects Response To Substance [15]
5-hydroxytryptamine receptor 1A (HTR1A) OT38K9MK 5HT1A_HUMAN Increases Response To Substance [49]
Sodium-dependent dopamine transporter (SLC6A3) OT39XG28 SC6A3_HUMAN Increases Response To Substance [50]
Leucine-rich repeat and immunoglobulin-like domain-containing nogo receptor-interacting protein 2 (LINGO2) OT3N88Q1 LIGO2_HUMAN Affects Response To Substance [15]
Prokineticin receptor 2 (PROKR2) OT51C69M PKR2_HUMAN Affects Response To Substance [51]
Rap guanine nucleotide exchange factor 5 (RAPGEF5) OT53VS75 RPGF5_HUMAN Affects Response To Substance [15]
Cubilin (CUBN) OT5A9HD3 CUBN_HUMAN Affects Response To Substance [15]
Neuropeptide Y receptor type 1 (NPY1R) OT5DA9DY NPY1R_HUMAN Increases Response To Substance [52]
Regulator of G-protein signaling 17 (RGS17) OT5RVUDS RGS17_HUMAN Affects Response To Substance [15]
Dual specificity mitogen-activated protein kinase kinase 5 (MAP2K5) OT6T408U MP2K5_HUMAN Affects Response To Substance [15]
RNA-binding protein Raly (RALY) OT78NNWY RALY_HUMAN Affects Response To Substance [15]
Collagen alpha-1(XXVIII) chain (COL28A1) OT78UJD5 COSA1_HUMAN Affects Response To Substance [15]
FK506-binding protein 15 (FKBP15) OT7NQJAU FKB15_HUMAN Affects Response To Substance [15]
Mitochondrial amidoxime reducing component 2 (MTARC2) OT8B6LVD MARC2_HUMAN Affects Response To Substance [15]
5-hydroxytryptamine receptor 6 (HTR6) OT8GTDDY 5HT6R_HUMAN Increases Response To Substance [53]
RAC-alpha serine/threonine-protein kinase (AKT1) OT8H2YY7 AKT1_HUMAN Increases Response To Substance [54]
PR domain zinc finger protein 2 (PRDM2) OT8L7CGX PRDM2_HUMAN Affects Response To Substance [15]
Histone-lysine N-methyltransferase SMYD3 (SMYD3) OT9YR20G SMYD3_HUMAN Affects Response To Substance [15]
Beta-arrestin-2 (ARRB2) OTAEJZCI ARRB2_HUMAN Increases Response To Substance [55]
D(4) dopamine receptor (DRD4) OTAJTO7N DRD4_HUMAN Increases Response To Substance [56]
Unconventional myosin-Vb (MYO5B) OTCKL3W3 MYO5B_HUMAN Affects Response To Substance [15]
Cadherin-13 (CDH13) OTD2CYM5 CAD13_HUMAN Affects Response To Substance [15]
Metabotropic glutamate receptor 2 (GRM2) OTD88Q92 GRM2_HUMAN Increases Response To Substance [57]
POU domain, class 5, transcription factor 1 (POU5F1) OTDHHN7O PO5F1_HUMAN Affects Response To Substance [15]
Transient receptor potential cation channel subfamily M member 3 (TRPM3) OTE4CDOQ TRPM3_HUMAN Affects Response To Substance [15]
Cone cGMP-specific 3',5'-cyclic phosphodiesterase subunit alpha' (PDE6C) OTE7EVWQ PDE6C_HUMAN Affects Response To Substance [15]
Proenkephalin-B (PDYN) OTEJ6430 PDYN_HUMAN Increases Response To Substance [58]
Astrotactin-2 (ASTN2) OTF0W2FJ ASTN2_HUMAN Affects Response To Substance [15]
Dendritic cell-specific transmembrane protein (DCSTAMP) OTFKRP2H DCSTP_HUMAN Affects Response To Substance [15]
Gamma-aminobutyric acid receptor subunit gamma-2 (GABRG2) OTGNDWUO GBRG2_HUMAN Increases Response To Substance [59]
Bis(5'-adenosyl)-triphosphatase (FHIT) OTGWBSLA FHIT_HUMAN Affects Response To Substance [15]
Prostaglandin G/H synthase 1 (PTGS1) OTHCRLEC PGH1_HUMAN Increases Response To Substance [60]
Cytochrome P450 2E1 (CYP2E1) OTHQ17JG CP2E1_HUMAN Affects Metabolism [61]
Sodium- and chloride-dependent glycine transporter 1 (SLC6A9) OTHRUON2 SC6A9_HUMAN Increases Response To Substance [62]
CUB and sushi domain-containing protein 1 (CSMD1) OTIVDSC4 CSMD1_HUMAN Affects Response To Substance [15]
Superoxide dismutase , mitochondrial (SOD2) OTIWXGZ9 SODM_HUMAN Increases Response To Substance [63]
E3 ubiquitin-protein ligase SH3RF3 (SH3RF3) OTK9VVUU SH3R3_HUMAN Affects Response To Substance [15]
Estrogen receptor (ESR1) OTKLU61J ESR1_HUMAN Increases Response To Substance [64]
Brain-derived neurotrophic factor (BDNF) OTLGH7EW BDNF_HUMAN Increases Response To Substance [65]
Cytochrome P450 1A2 (CYP1A2) OTLLBX48 CP1A2_HUMAN Affects Metabolism [61]
Glutathione S-transferase P (GSTP1) OTLP0A0Y GSTP1_HUMAN Increases Response To Substance [66]
Mitochondrial amidoxime-reducing component 1 (MTARC1) OTNBE033 MARC1_HUMAN Affects Response To Substance [15]
Heparan sulfate glucosamine 3-O-sulfotransferase 4 (HS3ST4) OTO0UBFN HS3S4_HUMAN Affects Response To Substance [15]
3',5'-cyclic-AMP phosphodiesterase 4B (PDE4B) OTOA8WU2 PDE4B_HUMAN Affects Response To Substance [15]
Acyloxyacyl hydrolase (AOAH) OTOYBXKW AOAH_HUMAN Affects Response To Substance [15]
cGMP-dependent protein kinase 1 (PRKG1) OTPAS4LF KGP1_HUMAN Affects Response To Substance [15]
Alpha-synuclein (SNCA) OTPWC1MR SYUA_HUMAN Increases Response To Substance [67]
Catechol O-methyltransferase (COMT) OTPWKTQG COMT_HUMAN Increases Response To Substance [56]
Cytochrome P450 3A4 (CYP3A4) OTQGYY83 CP3A4_HUMAN Affects Metabolism [61]
Glutathione S-transferase theta-1 (GSTT1) OTRFMWBK GSTT1_HUMAN Affects Response To Substance [68]
Glutathione S-transferase Mu 1 (GSTM1) OTSBF2MO GSTM1_HUMAN Decreases Response To Substance [69]
Procollagen C-endopeptidase enhancer 2 (PCOLCE2) OTSUEQZ1 PCOC2_HUMAN Affects Response To Substance [15]
Adenylate kinase isoenzyme 5 (AK5) OTTGXLOT KAD5_HUMAN Affects Response To Substance [15]
PH and SEC7 domain-containing protein 3 (PSD3) OTTN9B9O PSD3_HUMAN Affects Response To Substance [15]
Plasma serine protease inhibitor (SERPINA5) OTTZXPGD IPSP_HUMAN Affects Response To Substance [15]
Xylosyl- and glucuronyltransferase LARGE1 (LARGE1) OTUH7H9F LARG1_HUMAN Affects Response To Substance [15]
Adenosine receptor A2a (ADORA2A) OTVRBZ0I AA2AR_HUMAN Increases Response To Substance [70]
Complement decay-accelerating factor (CD55) OTZ3HWTO DAF_HUMAN Affects Response To Substance [15]
Pro-neuregulin-1, membrane-bound isoform (NRG1) OTZO6F1X NRG1_HUMAN Affects Response To Substance [15]
------------------------------------------------------------------------------------
⏷ Show the Full List of 169 DOT(s)

References

1 ClinicalTrials.gov (NCT04614584) Mirtazapine and Methamphetamine Drug-drug Interaction Study
2 URL: http://www.guidetopharmacology.org Nucleic Acids Res. 2015 Oct 12. pii: gkv1037. The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. (Ligand id: 7241).
3 Methamphetamine FDA Label
4 URL: http://www.guidetopharmacology.org Nucleic Acids Res. 2015 Oct 12. pii: gkv1037. The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. (Ligand id: 4803).
5 Pharmacotherapy for obesity. Drugs. 2005;65(10):1391-418.
6 Inverse agonist and neutral antagonist actions of antidepressants at recombinant and native 5-hydroxytryptamine2C receptors: differential modulatio... Mol Pharmacol. 2008 Mar;73(3):748-57.
7 A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001 Fall;7(3):249-64.
8 Impact of the CYP2D6 ultrarapid metabolizer genotype on mirtazapine pharmacokinetics and adverse events in healthy volunteers. J Clin Psychopharmacol. 2004 Dec;24(6):647-52.
9 Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002 Feb-May;34(1-2):83-448.
10 Antidepressant drugs activate SREBP and up-regulate cholesterol and fatty acid biosynthesis in human glial cells. Neurosci Lett. 2006 Mar 13;395(3):185-90. doi: 10.1016/j.neulet.2005.10.096. Epub 2005 Dec 1.
11 [Gynecomastia-galactorrhea during treatment with mirtazapine]. Presse Med. 2004 Apr 10;33(7):458. doi: 10.1016/s0755-4982(04)98631-9.
12 Effects of different antipsychotics and the antidepressant mirtazapine on glucose transporter mRNA levels in human blood cells. J Psychiatr Res. 2006 Jun;40(4):374-9. doi: 10.1016/j.jpsychires.2005.04.016. Epub 2005 Jul 5.
13 Methamphetamine-mediated endoplasmic reticulum (ER) stress induces type-1 programmed cell death in astrocytes via ATF6, IRE1 and PERK pathways. Oncotarget. 2016 Jul 19;7(29):46100-46119. doi: 10.18632/oncotarget.10025.
14 Thioredoxin-1 blocks methamphetamine-induced injury in brain through inhibiting endoplasmic reticulum and mitochondria-mediated apoptosis in mice. Neurotoxicology. 2020 May;78:163-169. doi: 10.1016/j.neuro.2020.03.006. Epub 2020 Mar 20.
15 Genome-wide association for methamphetamine dependence: convergent results from 2 samples. Arch Gen Psychiatry. 2008 Mar;65(3):345-55. doi: 10.1001/archpsyc.65.3.345.
16 Mirtazapine treatment after conditioning with methamphetamine alters subsequent expression of place preference. Drug Alcohol Depend. 2009 Jan 1;99(1-3):231-9.
17 Synthesis and Discovery of Arylpiperidinylquinazolines: New Inhibitors of the Vesicular Monoamine Transporter. J Med Chem. 2018 Oct 25;61(20):9121-9131.
18 Cytochrome P450 2D6.1 and cytochrome P450 2D6.10 differ in catalytic activity for multiple substrates. Pharmacogenetics. 2001 Aug;11(6):477-87.
19 Assessing Vesicular Monoamine Transport and Toxicity Using Fluorescent False Neurotransmitters. Chem Res Toxicol. 2021 May 17;34(5):1256-1264. doi: 10.1021/acs.chemrestox.0c00380. Epub 2020 Dec 30.
20 A preliminary study: novelty seeking, frontal executive function, and dopamine receptor (D2) TaqI A gene polymorphism in patients with methamphetamine dependence. Compr Psychiatry. 2008 Jul-Aug;49(4):387-92. doi: 10.1016/j.comppsych.2008.01.008. Epub 2008 Mar 21.
21 Identification of functional polymorphisms in the promoter region of the human PICK1 gene and their association with methamphetamine psychosis. Am J Psychiatry. 2007 Jul;164(7):1105-14. doi: 10.1176/ajp.2007.164.7.1105.
22 Evidence for shared genetic risk between methamphetamine-induced psychosis and schizophrenia. Neuropsychopharmacology. 2013 Sep;38(10):1864-70. doi: 10.1038/npp.2013.94. Epub 2013 Apr 12.
23 3,4-Methylenedioxymethamphetamine (MDMA) abuse may cause oxidative stress and potential free radical damage. Free Radic Res. 2003 May;37(5):491-7.
24 Brain antioxidant systems in human methamphetamine users. J Neurochem. 2004 Jun;89(6):1396-408.
25 Complex role of zinc in methamphetamine toxicity in vitro. Neuroscience. 2010 Nov 24;171(1):31-9.
26 Methamphetamine alters the normal progression by inducing cell cycle arrest in astrocytes. PLoS One. 2014 Oct 7;9(10):e109603.
27 Ginsenoside Re protects methamphetamine-induced mitochondrial burdens and proapoptosis via genetic inhibition of protein kinase C Delte in human neuroblastoma dopaminergic SH-SY5Y cell lines. J Appl Toxicol. 2015 Aug;35(8):927-44.
28 An insight into the hepatocellular death induced by amphetamines, individually and in combination: the involvement of necrosis and apoptosis. Arch Toxicol. 2013 Dec;87(12):2165-85. doi: 10.1007/s00204-013-1082-9. Epub 2013 Jul 3.
29 Involvement of C/EBP-related signaling pathway in methamphetamine-induced neuronal autophagy and apoptosis. Toxicol Lett. 2019 Sep 15;312:11-21. doi: 10.1016/j.toxlet.2019.05.003. Epub 2019 May 3.
30 Elevated plasma prolactin in abstinent methamphetamine-dependent subjects. Am J Drug Alcohol Abuse. 2011 Jan;37(1):62-7. doi: 10.3109/00952990.2010.538945. Epub 2010 Dec 10.
31 Methamphetamine induces AP-1 and NF-kappaB binding and transactivation in human brain endothelial cells. J Neurosci Res. 2001 Nov 15;66(4):583-91. doi: 10.1002/jnr.1248.
32 CDK5-mediated tau accumulation triggers methamphetamine-induced neuronal apoptosis via endoplasmic reticulum-associated degradation pathway. Toxicol Lett. 2018 Aug;292:97-107. doi: 10.1016/j.toxlet.2018.04.027. Epub 2018 Apr 26.
33 Methamphetamine exposure upregulates the amyloid precursor protein and hyperphosphorylated tau expression: The roles of insulin signaling in SH-SY5Y cell line. J Toxicol Sci. 2019;44(7):493-503. doi: 10.2131/jts.44.493.
34 Immunohistochemical investigation of dopaminergic terminal markers and caspase-3 activation in the striatum of human methamphetamine users. Int J Legal Med. 2007 May;121(3):163-8. doi: 10.1007/s00414-006-0087-9. Epub 2006 Apr 19.
35 Patterns of selective neuronal damage in methamphetamine-user AIDS patients. J Acquir Immune Defic Syndr. 2003 Dec 15;34(5):467-74. doi: 10.1097/00126334-200312150-00004.
36 Role of PUMA in methamphetamine-induced neuronal apoptosis. Toxicol Lett. 2016 Jan 5;240(1):149-60. doi: 10.1016/j.toxlet.2015.10.020. Epub 2015 Oct 30.
37 Inhibition of PLC1 signaling pathway regulates methamphetamine self-administration and neurotoxicity in rats. Food Chem Toxicol. 2021 Mar;149:111970. doi: 10.1016/j.fct.2021.111970. Epub 2021 Jan 7.
38 Methamphetamine-induced myocardial infarction with elevated troponin I. Am J Emerg Med. 2006 Jan;24(1):132-4. doi: 10.1016/j.ajem.2005.08.005.
39 Dopamine D1 receptor protein is elevated in nucleus accumbens of human, chronic methamphetamine users. Mol Psychiatry. 2000 Nov;5(6):664-72. doi: 10.1038/sj.mp.4000760.
40 Microglial and astrocytic changes in the striatum of methamphetamine abusers. Leg Med (Tokyo). 2010 Mar;12(2):57-62. doi: 10.1016/j.legalmed.2009.11.001. Epub 2010 Jan 27.
41 Melatonin attenuates methamphetamine-induced neuroinflammation through the melatonin receptor in the SH-SY5Y cell line. Neurotoxicology. 2015 Sep;50:122-30. doi: 10.1016/j.neuro.2015.08.008. Epub 2015 Aug 15.
42 Brain choline acetyltransferase activity in chronic, human users of cocaine, methamphetamine, and heroin. Mol Psychiatry. 1999 Jan;4(1):26-32. doi: 10.1038/sj.mp.4000462.
43 Brain serotonin transporter density and aggression in abstinent methamphetamine abusers. Arch Gen Psychiatry. 2006 Jan;63(1):90-100. doi: 10.1001/archpsyc.63.1.90.
44 Striatal dopamine d2/d3 receptor availability is reduced in methamphetamine dependence and is linked to impulsivity. J Neurosci. 2009 Nov 25;29(47):14734-40. doi: 10.1523/JNEUROSCI.3765-09.2009.
45 Melatonin attenuates methamphetamine-induced deactivation of the mammalian target of rapamycin signaling to induce autophagy in SK-N-SH cells. J Pineal Res. 2009 Mar;46(2):199-206. doi: 10.1111/j.1600-079X.2008.00648.x. Epub 2008 Nov 19.
46 Protective effect of neuropeptide Y2 receptor activation against methamphetamine-induced brain endothelial cell alterations. Toxicol Lett. 2020 Nov 1;334:53-59. doi: 10.1016/j.toxlet.2020.09.013. Epub 2020 Sep 18.
47 Linkage disequilibrium and association with methamphetamine dependence/psychosis of mu-opioid receptor gene polymorphisms. Pharmacogenomics J. 2006 May-Jun;6(3):179-88. doi: 10.1038/sj.tpj.6500355.
48 What is the contribution of human FMO3 in the N-oxygenation of selected therapeutic drugs and drugs of abuse?. Toxicol Lett. 2016 Sep 6;258:55-70. doi: 10.1016/j.toxlet.2016.06.013. Epub 2016 Jun 15.
49 Serotonin 1A receptor gene is associated with Japanese methamphetamine-induced psychosis patients. Neuropharmacology. 2010 Feb;58(2):452-6. doi: 10.1016/j.neuropharm.2009.09.006. Epub 2009 Sep 10.
50 Nine- or fewer repeat alleles in VNTR polymorphism of the dopamine transporter gene is a strong risk factor for prolonged methamphetamine psychosis. Pharmacogenomics J. 2003;3(4):242-7. doi: 10.1038/sj.tpj.6500189.
51 PROKR2 is associated with methamphetamine dependence in the Japanese population. Prog Neuropsychopharmacol Biol Psychiatry. 2010 Aug 16;34(6):1033-6. doi: 10.1016/j.pnpbp.2010.05.018. Epub 2010 May 24.
52 Association between neuropeptide Y gene and its receptor Y1 gene and methamphetamine dependence. Psychiatry Clin Neurosci. 2009 Jun;63(3):417-22. doi: 10.1111/j.1440-1819.2009.01961.x.
53 Serotonin 6 receptor gene is associated with methamphetamine-induced psychosis in a Japanese population. Drug Alcohol Depend. 2011 Jan 1;113(1):1-7. doi: 10.1016/j.drugalcdep.2010.06.021. Epub 2010 Aug 11.
54 Positive association of AKT1 haplotype to Japanese methamphetamine use disorder. Int J Neuropsychopharmacol. 2006 Feb;9(1):77-81. doi: 10.1017/S1461145705005481. Epub 2005 Jun 28.
55 Possible association of beta-arrestin 2 gene with methamphetamine use disorder, but not schizophrenia. Genes Brain Behav. 2007 Feb;6(1):107-12. doi: 10.1111/j.1601-183X.2006.00237.x.
56 Association analysis of the DRD4 and COMT genes in methamphetamine abuse. Am J Med Genet B Neuropsychiatr Genet. 2004 Aug 15;129B(1):120-4. doi: 10.1002/ajmg.b.30024.
57 Association analysis of GRM2 and HTR2A with methamphetamine-induced psychosis and schizophrenia in the Japanese population. Prog Neuropsychopharmacol Biol Psychiatry. 2010 May 30;34(4):639-44. doi: 10.1016/j.pnpbp.2010.03.002. Epub 2010 Mar 6.
58 Genetic variant of prodynorphin gene is risk factor for methamphetamine dependence. Neurosci Lett. 2006 May 29;400(1-2):158-62. doi: 10.1016/j.neulet.2006.02.038. Epub 2006 Mar 9.
59 Haplotype association between GABAA receptor gamma2 subunit gene (GABRG2) and methamphetamine use disorder. Pharmacogenomics J. 2005;5(2):89-95. doi: 10.1038/sj.tpj.6500292.
60 Human prostaglandin H synthase (hPHS)-1 and hPHS-2 in amphetamine analog bioactivation, DNA oxidation, and cytotoxicity. Toxicol Sci. 2011 Mar;120(1):154-62.
61 Epoxidation of the methamphetamine pyrolysis product, trans-phenylpropene, to trans-phenylpropylene oxide by CYP enzymes and stereoselective glutathione adduct formation. Toxicol Appl Pharmacol. 2006 Mar 1;211(2):148-56. doi: 10.1016/j.taap.2005.06.017. Epub 2005 Jul 20.
62 The glycine transporter 1 gene (GLYT1) is associated with methamphetamine-use disorder. Am J Med Genet B Neuropsychiatr Genet. 2008 Jan 5;147B(1):54-8. doi: 10.1002/ajmg.b.30565.
63 Association analysis of SOD2 variants with methamphetamine psychosis in Japanese and Taiwanese populations. Hum Genet. 2006 Sep;120(2):243-52. doi: 10.1007/s00439-006-0189-y. Epub 2006 Jun 29.
64 A functional polymorphism in estrogen receptor alpha gene is associated with Japanese methamphetamine induced psychosis. Prog Neuropsychopharmacol Biol Psychiatry. 2009 Aug 1;33(5):895-8. doi: 10.1016/j.pnpbp.2009.04.008. Epub 2009 Apr 19.
65 Association of brain-derived neurotrophic factor (Val66Met) genetic polymorphism with methamphetamine dependence in a Malaysian population. Brain Res. 2010 Oct 21;1357:91-6. doi: 10.1016/j.brainres.2010.08.053. Epub 2010 Aug 21.
66 A functional glutathione S-transferase P1 gene polymorphism is associated with methamphetamine-induced psychosis in Japanese population. Am J Med Genet B Neuropsychiatr Genet. 2005 May 5;135B(1):5-9. doi: 10.1002/ajmg.b.30164.
67 Study of association between alpha-synuclein gene polymorphism and methamphetamine psychosis/dependence. Ann N Y Acad Sci. 2004 Oct;1025:325-34. doi: 10.1196/annals.1316.040.
68 Association study between polymorphisms in glutathione-related genes and methamphetamine use disorder in a Japanese population. Am J Med Genet B Neuropsychiatr Genet. 2008 Oct 5;147B(7):1040-6. doi: 10.1002/ajmg.b.30703.
69 Association between the GST genetic polymorphisms and methamphetamine abusers in the Japanese population. Leg Med (Tokyo). 2009 Apr;11 Suppl 1:S468-70. doi: 10.1016/j.legalmed.2009.01.095. Epub 2009 Feb 28.
70 The adenosine A2A receptor is associated with methamphetamine dependence/psychosis in the Japanese population. Behav Brain Funct. 2010 Aug 30;6:50. doi: 10.1186/1744-9081-6-50.