P-glycoprotein

Source: Wikipedia, the free encyclopedia.
ABCB1
Gene ontology
Molecular function
Cellular component
Biological process
Sources:Amigo / QuickGO
Ensembl
UniProt
RefSeq (mRNA)

NM_000927

NM_011076

RefSeq (protein)

NP_000918
NP_001335873
NP_001335874
NP_001335875

NP_035206

Location (UCSC)n/aChr 5: 8.71 – 8.8 Mb
PubMed search[2][3]
Wikidata
View/Edit HumanView/Edit Mouse
ABCB1 is differentially expressed in 97 experiments [93 up/106 dn]: 26 organism parts: kidney [2 up/0 dn], bone marrow [0 up/2 dn], ...; 29 disease states: normal [10 up/3 dn], glioblastoma [0 up/2 dn], ...; 30 cell types, 22 cell lines, 11 compound treatments and 16 other conditions.
Factor Value Factor Up/Down
Legend: – number of studies the gene is up/down in
Normal Disease state 10/3
None Compound treatment 3/0
Stromal cell Cell type 1/2
Kidney Cell type 2/0
MDA-MB-231
Cell line 0/2
Glioblastoma Disease state 0/2
Epithelial cell Cell type 0/2
HeLa Cell line 0/2
Primary Disease staging 2/0
Bone marrow Organism part 0/2
ABCB1 expression data in ATLAS

P-glycoprotein 1 (permeability glycoprotein, abbreviated as P-gp or Pgp) also known as multidrug resistance protein 1 (MDR1) or ATP-binding cassette sub-family B member 1 (ABCB1) or cluster of differentiation 243 (CD243) is an important protein of the

substrate
specificity. It exists in animals, fungi, and bacteria, and it likely evolved as a defense mechanism against harmful substances.

P-gp is extensively distributed and expressed in the

proximal tubule of the kidney where it pumps them into urinary filtrate (in the proximal tubule), and in the capillary endothelial cells composing the blood–brain barrier and blood–testis barrier
, where it pumps them back into the capillaries.

P-gp is a

pharmaceutical drugs (which are said to be P-gp substrates). In addition, some cancer cells also express large amounts of P-gp, further amplifying that effect and rendering these cancers multidrug resistant. Many drugs inhibit P-gp, typically incidentally rather than as their main mechanism of action; some foods do as well.[6]
Any such substance can sometimes be called a P-gp inhibitor.

P-gp was discovered in 1971 by Victor Ling.

Gene

A 2015 review of polymorphisms in ABCB1 found that "the effect of ABCB1 variation on P-glycoprotein expression (messenger RNA and protein expression) and/or activity in various tissues (e.g. the liver, gut and heart) appears to be small. Although polymorphisms and haplotypes of ABCB1 have been associated with alterations in drug disposition and drug response, including adverse events with various ABCB1 substrates in different ethnic populations, the results have been majorly conflicting, with limited clinical relevance."[7]

Protein

P-gp is a 170 kDa transmembrane glycoprotein, which includes 10-15 kDa of N-terminal glycosylation. The N-terminal half of the protein contains 6 transmembrane helixes, followed by a large cytoplasmic domain with an ATP-binding site, and then a second section with 6 transmembrane helixes and an ATP-binding domain that shows over 65% of amino acid similarity with the first half of the polypeptide.[8] In 2009, the first structure of a mammalian P-glycoprotein was solved (3G5U).[9] The structure was derived from the mouse MDR3 gene product heterologously expressed in Pichia pastoris yeast. The structure of mouse P-gp is similar to structures of the bacterial ABC transporter MsbA (3B5W and 3B5X)[10] that adopt an inward facing conformation that is believed to be important for binding substrate along the inner leaflet of the membrane. Additional structures (3G60 and 3G61) of P-gp were also solved revealing the binding site(s) of two different cyclic peptide substrate/inhibitors. The promiscuous binding pocket of P-gp is lined with aromatic amino acid side chains.

Through Molecular Dynamic (MD) simulations, this sequence was proved to have a direct impact in the transporter's structural stability (in the nucleotide-binding domains) and defining a lower boundary for the internal drug-binding pocket.[11]

Species, tissue, and subcellular distribution

P-gp is expressed primarily in certain cell types in the liver, pancreas, kidney, colon, and

endothelial cells.[13]

Function

Substrate enters P-gp either from an opening within the inner leaflet of the membrane or from an opening at the cytoplasmic side of the protein. ATP binds at the cytoplasmic side of the protein. Following binding of each, ATP hydrolysis shifts the substrate into a position to be excreted from the cell. Release of the phosphate (from the original ATP molecule) occurs concurrently with substrate excretion. ADP is released, and a new molecule of ATP binds to the secondary ATP-binding site. Hydrolysis and release of ADP and a phosphate molecule resets the protein, so that the process can start again.

The protein belongs to the superfamily of

multidrug resistance. P-gp is an ATP-dependent drug efflux pump for xenobiotic compounds with broad substrate specificity. It is responsible for decreased drug accumulation in multidrug-resistant cells and often mediates the development of resistance to anticancer drugs. This protein also functions as a transporter in the blood–brain barrier. Mutations in this gene are associated with colchicine resistance and Inflammatory bowel disease 13. Alternative splicing and the use of alternative promoters results in multiple transcript variants. [14]

P-gp transports various substrates across the cell membrane including:

Its ability to transport the above substrates accounts for the many roles of P-gp including:

  • Regulating the distribution and bioavailability of drugs
    • Increased intestinal expression of P-glycoprotein can reduce the absorption of drugs that are substrates for P-glycoprotein. Thus, there is a reduced bioavailability, and therapeutic plasma concentrations are not attained. On the other hand, supratherapeutic plasma concentrations and drug toxicity may result because of decreased P-glycoprotein expression
    • multidrug resistance
      to these drugs
  • The removal of toxic metabolites and xenobiotics from cells into urine, bile, and the intestinal lumen
  • The transport of compounds out of the brain across the blood–brain barrier
  • Digoxin uptake
  • Prevention of ivermectin and loperamide entry into the central nervous system
  • The migration of
    dendritic cells
  • Protection of hematopoietic
    stem cells from toxins.[5]

It is inhibited by many drugs, such as

Regulation of expression and function of P-gp in cancer cells

At the

c-Jun N-terminal kinase (JNK) pathway, all of which were reported to have implications in the regulation of the expression of P-gp. Studies suggested that the MAPK/ERK pathway is involved in the positive regulation of P-gp;[21] the p38 MAPK pathway negatively regulates the expression of the P-gp gene;[22] and the JNK pathway was reported to be involved in both positive regulation and negative regulation of P-gp.[23][24]

After 2008, microRNAs (miRNAs) were identified as new players in regulating the expression of P-gp in both transcriptional and post-transcriptional levels. Some miRNAs decrease the expression of P-gp. For example, miR-200c down-regulates the expression of P-gp through the JNK signaling pathway[23] or ZEB1 and ZEB2;[25] miR-145 down-regulates the mRNA of P-gp by directly binding to the 3'-UTR of the gene of P-gp and thus suppresses the translation of P-gp.[26] Some other miRNAs increase the expression of P-gp. For example, miR-27a up-regulates P-gp expression by suppressing the Raf kinase inhibitor protein (RKIP);[27] alternatively, miR-27a can also directly bind to the promoter of the P-gp gene, which works in a similar way with the mechanism of action of transcriptional factors.[28]

The expression of P-gp is also regulated by post-translational events, such as

intracellular compartments to the cell membrane, and therefore decreases the functional P-gp level on the cell membrane.[31]

Clinical significance

Drug interactions

Some common pharmacological inhibitors of P-glycoprotein include: amiodarone, clarithromycin, ciclosporin, colchicine, diltiazem, erythromycin, felodipine, ketoconazole,[32] lansoprazole, omeprazole and other proton-pump inhibitors, nifedipine, paroxetine, reserpine,[33] saquinavir,[32] sertraline, quinidine, tamoxifen, verapamil,[34] and duloxetine.[35] Elacridar and CP 100356 are other common[citation needed] P-gp inhibitors. Zosuquidar and tariquidar were also developed with this in mind.[clarification needed] Lastly, valspodar and reversan are other examples of such agents. ABCB1 is linked to the daily dose of warfarin required to maintain the INR to a target of 2.5. Patients with the GT or TT genotypes of the 2677G>T SNP require around 20% more warfarin daily.[36]

Common pharmacological inducers of P-glycoprotein include

Substrates of P-glycoprotein are susceptible to changes in pharmacokinetics due to drug interactions with P-gp inhibitors or inducers. Some of these substrates include colchicine, ciclosporin, dabigatran,[33] digoxin, diltiazem,[38] fexofenadine, indinavir, morphine, and sirolimus.[32]

Diseases (non-cancer)

Decreased P-gp expression has been found in Alzheimer's disease brains.[39]

Altered P-gp function has also been linked to

inflammatory bowel diseases (IBD);[40] however, due to its ambivalent effects in intestinal inflammation many questions remain so far unanswered.[41] While decreased efflux activity may promote disease susceptibility and drug toxicity, increased efflux activity may confer resistance to therapeutic drugs in IBD.[41] Mice deficient in MDR1A develop chronic intestinal inflammation spontaneously, which appears to resemble human ulcerative colitis.[42]

Cancer

P-gp efflux activity is capable of lowering intracellular concentrations of otherwise beneficial compounds, such as chemotherapeutics and other medications, to sub-therapeutic levels. Consequently, P-gp overexpression is one of the main mechanisms behind decreased intracellular drug accumulation and development of multidrug resistance in human multidrug-resistant (MDR) cancers.[43][44]

History

P-gp was first characterized in 1976. P-gp was shown to be responsible for conferring multidrug resistance upon mutant cultured cancer cells that had developed resistance to cytotoxic drugs.[5][45]

The structure of mouse P-gp, which has 87% sequence identity to human P-gp, was resolved by x-ray crystallography in 2009.[9] The first structure of human P-gp was solved in 2018, with the protein in its ATP-bound, outward-facing conformation. [46]

Research

Radioactive verapamil can be used for measuring P-gp function with positron emission tomography.[47]

P-gp is also used to differentiate

transitional B cells from naive B cells. Dyes such as rhodamine 123 and MitoTracker dyes from Invitrogen can be used to make this differentiation.[48]

MDR1 as a drug target

It has been suggested that MDR1 inhibitors might treat various diseases, especially cancers, but none have done well in clinical trials.[49]

Single nucleotide polymorphism rs1045642

Single Nucleotide Polymorphism
are important for the differential activity of the PGP pump.

Homozygous subjects, identified with the TT genotype, are usually more able to extrude xenobiotics from the cell. A Homozygous genotype for the allele ABCB1/MDR1 is capable of a higher absorption from the blood vessels and a lower extrusion into the lumen.

Xenobiotics are extruded at a lower rate with heterozygous (CT) alleles compared to homozygous ones. [50]

References

  1. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000040584 - Ensembl, May 2017
  2. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  3. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. PMID 3027054
    .
  5. ^ a b c Dean, Michael (2002-11-01). "The Human ATP-Binding Cassette (ABC) Transporter Superfamily". National Library of Medicine (US), NCBI. Archived from the original on 2006-02-12. Retrieved 2008-03-02.
  6. PMID 28414144
    .
  7. .
  8. ^ Franck Viguié (1998-03-01). "ABCB1". Atlas of Genetics and Cytogenetics in Oncology and Haematology. Retrieved 2008-03-02.
  9. ^
    PMID 19325113
    .
  10. .
  11. .
  12. .
  13. .
  14. ^ "Entrez Gene: ABCB1".
  15. ^ "Desloratadine: Drug information". UpToDate. Retrieved 2019-02-01.
  16. ^ "Drug Development and Drug Interactions: Table of Substrates, Inhibitors and Inducers". FDA. 26 May 2021.
  17. PMID 7829527
    .
  18. ^ .
  19. .
  20. .
  21. .
  22. .
  23. ^ .
  24. .
  25. .
  26. .
  27. .
  28. .
  29. .
  30. .
  31. .
  32. ^ a b c John r. Horn, Pharmd; Philip Hansten, Pharmd (December 2008). "Drug Transporters: The Final Frontier for Drug Interactions". Pharmacy Times. Retrieved 2018-11-30.
  33. ^ a b Research, Center for Drug Evaluation and. "Drug Interactions & Labeling – Drug Development and Drug Interactions: Table of Substrates, Inhibitors and Inducers". www.fda.gov. Retrieved 2018-11-30.
  34. .
  35. ^ Gopisankar MG, Hemachandren M, Surendiran A. ABCB1 266G-T single nucleotide polymorphism influences warfarin dose requirement for warfarin maintenance therapy. Br J Biomed Sci 2019:76;150-152
  36. ^ Health, Ministry of (2015). "Use of Non-Vitamin K Antagonist Oral Anticoagulants (NOAC) in Non-Valvular Atrial Fibrillation – Province of British Columbia. Appendix A (2015)". www2.gov.bc.ca. Archived from the original on 2018-12-01. Retrieved 2018-11-30.{{cite web}}: CS1 maint: bot: original URL status unknown (link)
  37. ^ "Diltiazem: Drug information". UpToDate. Retrieved 2019-02-01.
  38. PMID 27494062
    .
  39. .
  40. ^ .
  41. .
  42. .
  43. .
  44. .
  45. .
  46. .
  47. .
  48. ^ Inhibiting Cancer Drug Resistance Gene May Not Be Best Approach Apr 2020
  49. PMID 18424454
    .

Further reading

External links

This article incorporates text from the United States National Library of Medicine, which is in the public domain.