N-Methyl-D-histidine

N-Methyl-D-histidine

* Please be kindly noted products are not for therapeutic use. We do not sell to patients.

N-Methyl-D-histidine
Category Others
Catalog number BBF-05236
CAS 738564-96-0
Molecular Weight 169.18
Molecular Formula C7H11N3O2
Purity >95% by HPLC

Online Inquiry

Specification

Related CAS 24886-03-1 (L-configuration)
Synonyms Me-D-His-OH; r-methylhistidine; (R)-3-(1H-Imidazol-4-yl)-2-(methylamino)propanoic acid; D-Histidine, N-methyl-; methyl-D-histidine
Storage Store at -20°C
IUPAC Name (2R)-3-(1H-imidazol-5-yl)-2-(methylamino)propanoic acid
Canonical SMILES CNC(CC1=CN=CN1)C(=O)O
InChI InChI=1S/C7H11N3O2/c1-8-6(7(11)12)2-5-3-9-4-10-5/h3-4,6,8H,2H2,1H3,(H,9,10)(H,11,12)/t6-/m1/s1
InChI Key CYZKJBZEIFWZSR-ZCFIWIBFSA-N

Properties

Boiling Point 432.8±35.0°C at 760 mmHg
Density 1.3±0.1 g/cm3

Reference Reading

1. Small interfering RNA for cancer treatment: overcoming hurdles in delivery
Nitin Bharat Charbe, Nikhil D Amnerkar, B Ramesh, Murtaza M Tambuwala, Hamid A Bakshi, Alaa A A Aljabali, Saurabh C Khadse, Rajendran Satheeshkumar, Saurabh Satija, Meenu Metha, Dinesh Kumar Chellappan, Garima Shrivastava, Gaurav Gupta, Poonam Negi, Kamal Dua, Flavia C Zacconi Acta Pharm Sin B. 2020 Nov;10(11):2075-2109. doi: 10.1016/j.apsb.2020.10.005. Epub 2020 Oct 13.
In many ways, cancer cells are different from healthy cells. A lot of tactical nano-based drug delivery systems are based on the difference between cancer and healthy cells. Currently, nanotechnology-based delivery systems are the most promising tool to deliver DNA-based products to cancer cells. This review aims to highlight the latest development in the lipids and polymeric nanocarrier for siRNA delivery to the cancer cells. It also provides the necessary information about siRNA development and its mechanism of action. Overall, this review gives us a clear picture of lipid and polymer-based drug delivery systems, which in the future could form the base to translate the basic siRNA biology into siRNA-based cancer therapies.
2. How Does Replacement of the Axial Histidine Ligand in Cytochrome c Peroxidase by Nδ-Methyl Histidine Affect Its Properties and Functions? A Computational Study
Calvin W Z Lee, M Qadri E Mubarak, Anthony P Green, Sam P de Visser Int J Mol Sci. 2020 Sep 27;21(19):7133. doi: 10.3390/ijms21197133.
Heme peroxidases have important functions in nature related to the detoxification of H2O2. They generally undergo a catalytic cycle where, in the first stage, the iron(III)-heme-H2O2 complex is converted into an iron(IV)-oxo-heme cation radical species called Compound I. Cytochrome c peroxidase Compound I has a unique electronic configuration among heme enzymes where a metal-based biradical is coupled to a protein radical on a nearby Trp residue. Recent work using the engineered Nδ-methyl histidine-ligated cytochrome c peroxidase highlighted changes in spectroscopic and catalytic properties upon axial ligand substitution. To understand the axial ligand effect on structure and reactivity of peroxidases and their axially Nδ-methyl histidine engineered forms, we did a computational study. We created active site cluster models of various sizes as mimics of horseradish peroxidase and cytochrome c peroxidase Compound I. Subsequently, we performed density functional theory studies on the structure and reactivity of these complexes with a model substrate (styrene). Thus, the work shows that the Nδ-methyl histidine group has little effect on the electronic configuration and structure of Compound I and little changes in bond lengths and the same orbital occupation is obtained. However, the Nδ-methyl histidine modification impacts electron transfer processes due to a change in the reduction potential and thereby influences reactivity patterns for oxygen atom transfer. As such, the substitution of the axial histidine by Nδ-methyl histidine in peroxidases slows down oxygen atom transfer to substrates and makes Compound I a weaker oxidant. These studies are in line with experimental work on Nδ-methyl histidine-ligated cytochrome c peroxidases and highlight how the hydrogen bonding network in the second coordination sphere has a major impact on the function and properties of the enzyme.
3. Protein Histidine Methylation
Sebastian Kwiatkowski, Jakub Drozak Curr Protein Pept Sci. 2020;21(7):675-689. doi: 10.2174/1389203721666200318161330.
Protein histidine methylation is a rarely studied posttranslational modification in eukaryotes. Although the presence of N-methylhistidine was demonstrated in actin in the early 1960s, so far, only a limited number of proteins containing N-methylhistidine have been reported, including S100A9, myosin, skeletal muscle myosin light chain kinase (MLCK 2), and ribosomal protein Rpl3. Furthermore, the role of histidine methylation in the functioning of the protein and in cell physiology remains unclear due to a shortage of studies focusing on this topic. However, the molecular identification of the first two distinct histidine-specific protein methyltransferases has been established in yeast (Hpm1) and in metazoan species (actin-histidine N-methyltransferase), giving new insights into the phenomenon of protein methylation at histidine sites. As a result, we are now beginning to recognize protein histidine methylation as an important regulatory mechanism of protein functioning whose loss may have deleterious consequences in both cells and in organisms. In this review, we aim to summarize the recent advances in the understanding of the chemical, enzymological, and physiological aspects of protein histidine methylation.

Recommended Products

Bio Calculators

Stock concentration: *
Desired final volume: *
Desired concentration: *

L

* Our calculator is based on the following equation:
Concentration (start) x Volume (start) = Concentration (final) x Volume (final)
It is commonly abbreviated as: C1V1 = C2V2

* Total Molecular Weight:
g/mol
Tip: Chemical formula is case sensitive. C22H30N4O c22h30n40
g/mol
g

Recently viewed products

Online Inquiry

Verification code
cartIcon
Inquiry Basket