ω-Conotoxin MVIIC

Ca2+ channel blocker (N, P and Q-type) CAS# 147794-23-8

ω-Conotoxin MVIIC

Catalog No. BCC5699----Order now to get a substantial discount!

Product Name & Size Price Stock
ω-Conotoxin MVIIC:100µg $254.00 In stock
ω-Conotoxin MVIIC:200µg $432.00 In stock
ω-Conotoxin MVIIC:500µg $1016.00 In stock
ω-Conotoxin MVIIC:1000µg $1778.00 In stock
Related Products

Quality Control of ω-Conotoxin MVIIC

Number of papers citing our products

Chemical structure

ω-Conotoxin MVIIC

3D structure

Chemical Properties of ω-Conotoxin MVIIC

Cas No. 147794-23-8 SDF Download SDF
PubChem ID 56841670 Appearance Powder
Formula C106H178N40O32S7 M.Wt 2749.3
Type of Compound N/A Storage Desiccate at -20°C
Solubility Soluble to 1 mg/ml in water
Sequence CKGKGAPCRKTMYDCCSGSCGRRGKC

(Modifications: Disulfide bridge between 1 - 16, 8 - 20, 15 - 26, Cys-26 = C-terminal amide)

Chemical Name 2-[(1R,4S,7S,10S,13S,16S,19S,22R,27R,30S,36S,39S,45R,48S,54S,57R,62R,65S,71S,77S,83S)-62-amino-7,30,65,71-tetrakis(4-aminobutyl)-4,36,39-tris(3-carbamimidamidopropyl)-27-carbamoyl-10-[(1R)-1-hydroxyethyl]-48,54-bis(hydroxymethyl)-16-[(4-hydroxyphenyl)methyl]-77-methyl-13-(2-methylsulfanylethyl)-2,5,8,11,14,17,20,29,32,35,38,41,44,47,50,53,56,63,66,69,72,75,78,84,91-pentacosaoxo-24,25,59,60,87,88-hexathia-3,6,9,12,15,18,21,28,31,34,37,40,43,46,49,52,55,64,67,70,73,76,79,85,90-pentacosazatetracyclo[43.40.4.222,57.079,83]hennonacontan-19-yl]acetic acid
SMILES CC1C(=O)N2CCCC2C(=O)NC3CSSCC4C(=O)NCC(=O)NC(C(=O)NC(C(=O)NCC(=O)NC(C(=O)NC(CSSCC(C(=O)NC(CSSCC(C(=O)NC(C(=O)NCC(=O)NC(C(=O)NCC(=O)N1)CCCCN)CCCCN)N)C(=O)NC(C(=O)NCC(=O)NC(C(=O)N4)CO)CO)NC(=O)C(NC(=O)C(NC(=O)C(NC(=O)C(NC(=O)C(NC(=O)C(NC3=O)CCCNC(=N)N)CCCCN)C(C)O)CCSC)CC5=CC=C(C=C5)O)CC(=O)O)C(=O)N)CCCCN)CCCNC(=N)N)CCCNC(=N)N
Standard InChIKey FHVUTHWUIUXZBY-QLANQDRJSA-N
Standard InChI InChI=1S/C106H178N40O32S7/c1-53-103(178)146-36-15-23-75(146)101(176)144-74-52-185-183-49-71-89(164)126-43-79(154)130-62(21-13-34-120-105(115)116)90(165)133-60(20-12-33-119-104(113)114)87(162)124-42-78(153)129-61(18-6-10-31-109)91(166)140-70(83(112)158)48-181-184-51-73(100(175)143-72(99(174)139-68(45-147)88(163)125-44-80(155)131-69(46-148)97(172)141-71)50-182-180-47-57(111)84(159)132-59(17-5-9-30-108)86(161)123-41-77(152)128-58(16-4-8-29-107)85(160)122-40-76(151)127-53)142-96(171)67(39-81(156)157)138-95(170)66(38-55-24-26-56(150)27-25-55)137-93(168)65(28-37-179-3)136-102(177)82(54(2)149)145-94(169)63(19-7-11-32-110)134-92(167)64(135-98(74)173)22-14-35-121-106(117)118/h24-27,53-54,57-75,82,147-150H,4-23,28-52,107-111H2,1-3H3,(H2,112,158)(H,122,160)(H,123,161)(H,124,162)(H,125,163)(H,126,164)(H,127,151)(H,128,152)(H,129,153)(H,130,154)(H,131,155)(H,132,159)(H,133,165)(H,134,167)(H,135,173)(H,136,177)(H,137,168)(H,138,170)(H,139,174)(H,140,166)(H,141,172)(H,142,171)(H,143,175)(H,144,176)(H,145,169)(H,156,157)(H4,113,114,119)(H4,115,116,120)(H4,117,118,121)/t53-,54+,57-,58-,59-,60-,61-,62-,63-,64-,65-,66-,67-,68-,69-,70-,71-,72-,73-,74-,75-,82-/m0/s1
General tips For obtaining a higher solubility , please warm the tube at 37 ℃ and shake it in the ultrasonic bath for a while.Stock solution can be stored below -20℃ for several months.
We recommend that you prepare and use the solution on the same day. However, if the test schedule requires, the stock solutions can be prepared in advance, and the stock solution must be sealed and stored below -20℃. In general, the stock solution can be kept for several months.
Before use, we recommend that you leave the vial at room temperature for at least an hour before opening it.
About Packaging 1. The packaging of the product may be reversed during transportation, cause the high purity compounds to adhere to the neck or cap of the vial.Take the vail out of its packaging and shake gently until the compounds fall to the bottom of the vial.
2. For liquid products, please centrifuge at 500xg to gather the liquid to the bottom of the vial.
3. Try to avoid loss or contamination during the experiment.
Shipping Condition Packaging according to customer requirements(5mg, 10mg, 20mg and more). Ship via FedEx, DHL, UPS, EMS or other couriers with RT, or blue ice upon request.

Biological Activity of ω-Conotoxin MVIIC

DescriptionPeptide neurotoxin; wide spectrum blocker of N, P and Q type calcium channels.

ω-Conotoxin MVIIC Dilution Calculator

Concentration (start)
x
Volume (start)
=
Concentration (final)
x
Volume (final)
 
 
 
C1
V1
C2
V2

calculate

ω-Conotoxin MVIIC Molarity Calculator

Mass
=
Concentration
x
Volume
x
MW*
 
 
 
g/mol

calculate

Organizitions Citing Our Products recently

 
 
 

Calcutta University

University of Minnesota

University of Maryland School of Medicine

University of Illinois at Chicago

The Ohio State University

University of Zurich

Harvard University

Colorado State University

Auburn University

Yale University

Worcester Polytechnic Institute

Washington State University

Stanford University

University of Leipzig

Universidade da Beira Interior

The Institute of Cancer Research

Heidelberg University

University of Amsterdam

University of Auckland
TsingHua University
TsingHua University
The University of Michigan
The University of Michigan
Miami University
Miami University
DRURY University
DRURY University
Jilin University
Jilin University
Fudan University
Fudan University
Wuhan University
Wuhan University
Sun Yat-sen University
Sun Yat-sen University
Universite de Paris
Universite de Paris
Deemed University
Deemed University
Auckland University
Auckland University
The University of Tokyo
The University of Tokyo
Korea University
Korea University
Featured Products
New Products
 

References on ω-Conotoxin MVIIC

Characteristics of omega-conotoxin GVI A and MVIIC binding to Cav 2.1 and Cav 2.2 channels captured by anti-Ca2+ channel peptide antibodies.[Pubmed:16076016]

Neurochem Res. 2005 Apr;30(4):457-66.

A New Binding Method (NBM) was used to investigate the characteristics of the specific binding of 125I-omega-conotoxin (omega-CTX) GVIA and 125I-omega-CTX MVIIC to Cav2.1 and Cav2.2 channels captured from chick brain membranes by antibodies against B1Nt (a peptide sequence in Car2.1 and Cav2.2 channels). The results for the NBM were as follows. (1) The ED50 values for specific binding of 125I-omega-CTX GVIA and 125I-omega-CTX MVIIC to Cav2.1 and Cav2.2 channels were about 68 and 60 pM, respectively, and very similar to those (87 and 35 pM, respectively) to crude membranes from chick brain. (2) The specific 125I-omega-CTX GVIA (100 pM) binding was inhibited by omega-CTX GVIA (0.5 nM), dynorphine A (Dyn), gentamicin (Gen), neomycin (Neo) and tobramicin (Tob) (100 microM each), but not by omega-agaconotoxin (Aga) IVA, calciseptine, omega-CTX SVIB, omega-CTX MVIIC (0.5 nM each), PN200-110 (PN), diltiazem (Dil) or verapamil (Ver) (100 microM each). Calmodulin (CaM) inhibited the specific binding in a dose-dependent manner (IC50 value of about 100 microg protein/ml). (3) The specific 125I-omega-CTX MVIIC (60 pM) binding was inhibited by omega-CTX MVIIC, omega-CTX GVIA, omega-CTX SVIB (0.5 nM each), Dyn, Neo and Tob (100 microM, each), but not by omega-Aga IVA, calciseptine (0.5 nM each), PN, Dil, Ver (100 microM each) or 100 microg protein/ml CaM. These results suggested that the characteristics of the specific binding of 125I-omega-CTX GVIA and 125I-omega-CTX MVIIC to Cav2.1 and Cav2.2 channels in the NBM were very similar to those to crude membranes from chick brain, although the IC50 values for CaM and free Ca2+ of CaM were about 33- and 5000-fold higher, respectively, than those for the specific binding of 125I-omega-CTX GVIA and 125I-omega-CTX MVIIC to crude membranes.

Role of Thr(11) in the binding of omega-conotoxin MVIIC to N-type Ca2+ channels.[Pubmed:11226434]

FEBS Lett. 2001 Feb 23;491(1-2):127-30.

As replacement of Thr(11) of omega-conotoxin MVIIC with Ala significantly reduced the affinity for both N- and P/Q-type calcium channels, we examined the effect of substitution at this position with other residues. Binding assays using rat cerebellar P2 membranes showed that the affinity is in the order of Leu>Val, aminobutyric acid, Thr>Asn&z.Gt;Ser, Ala, Asp, Phe, Tyr for N-type channels and Thr>Leu, Val, aminobutyric acid, Asn, Ser>Ala&z.Gt;Asp, Phe, Tyr for P/Q-type channels, suggesting that aliphatic amino acids with longer side chains are favorable for block of N-type channels. The effects of substitution were examined electrophysiologically in BHK cells expressing N-type Ca2+ channels. Inhibition of Ba2+ current by the analogs did not completely correlate with binding affinity, although binding to BHK cells was comparable to rat cerebellar membranes.

Systemic effects induced by intralesional injection of omega-conotoxin MVIIC after spinal cord injury in rats.[Pubmed:24739121]

J Venom Anim Toxins Incl Trop Dis. 2014 Apr 16;20:15.

BACKGROUND: Calcium channel blockers such as conotoxins have shown a great potential to reduce brain and spinal cord injury. MVIIC neuroprotective effects analyzed in in vitro models of brain and spinal cord ischemia suggest a potential role of this toxin in preventing injury after spinal cord trauma. However, previous clinical studies with MVIIC demonstrated that clinical side effects might limit the usefulness of this drug and there is no research on its systemic effects. Therefore, the present study aimed to investigate the potential toxic effects of MVIIC on organs and to evaluate clinical and blood profiles of rats submitted to spinal cord injury and treated with this marine toxin. Rats were treated with placebo or MVIIC (at doses of 15, 30, 60 or 120 pmol) intralesionally following spinal cord injury. Seven days after the toxin administration, kidney, brain, lung, heart, liver, adrenal, muscles, pancreas, spleen, stomach, and intestine were histopathologically investigated. In addition, blood samples collected from the rats were tested for any hematologic or biochemical changes. RESULTS: The clinical, hematologic and biochemical evaluation revealed no significant abnormalities in all groups, even in high doses. There was no significant alteration in organs, except for degenerative changes in kidneys at a dose of 120 pmol. CONCLUSIONS: These findings suggest that MVIIC at 15, 30 and 60 pmol are safe for intralesional administration after spinal cord injury and could be further investigated in relation to its neuroprotective effects. However, 120 pmol doses of MVIIC may provoke adverse effects on kidney tissue.

Omega-conotoxin MVIIC attenuates neuronal apoptosis in vitro and improves significant recovery after spinal cord injury in vivo in rats.[Pubmed:25120731]

Int J Clin Exp Pathol. 2014 Jun 15;7(7):3524-36. eCollection 2014.

Excessive accumulation of intracellular calcium is the most critical step after spinal cord injury (SCI). Reducing the calcium influx should result in a better recovery from SCI. Calcium channel blockers have been shown a great potential in reducing brain and spinal cord injury. In this study, we first tested the neuroprotective effect of MVIIC on slices of spinal cord subjected to ischemia evaluating cell death and caspase-3 activation. Thereafter, we evaluated the efficacy of MVIIC in ameliorating damage following SCI in rats, for the first time in vivo. The spinal cord slices subjected a pretreatment with MVIIC showed a cell protection with a reduction of dead cells in 24.34% and of caspase-3-specific protease activation. In the in vivo experiment, Wistar rats were subjected to extradural compression of the spinal cord at the T12 vertebral level using a weigh of 70 g/cm, following intralesional treatment with either placebo or MVIIC in different doses (15, 30 and 60 pmol) five minutes after injury. Behavioral testing of hindlimb function was done using the Basso Beattie Bresnahan locomotor rating scale, and revealed significant recovery with 15 pmol (G15) compared to other trauma groups. Also, histological bladder structural revealed significant outcome in G15, with no morphological alterations, and anti-NeuN and TUNEL staining showed that G15 provided neuron preservation and indicated that this group had fewer neuron cell death, similar to sham. These results showed the neuroprotective effects of MVIIC in in vitro and in vivo model of SCI with neuronal integrity, bladder and behavioral improvements.

Inhibition of calcium channels in rat central and peripheral neurons by omega-conotoxin MVIIC.[Pubmed:8786437]

J Neurosci. 1996 Apr 15;16(8):2612-23.

Inhibition of voltage-dependent calcium channels by omega-conotoxin MVIIC (omega-CTx-MVIIC) was studied in various types of rat neurons. When studied with 5 mM Ba2+ as charge carrier, omega-CTx-MVIIC block of N-type calcium channels in sympathetic neurons was potent, with half-block at 18 nM. Block of N-type channels had a rapid onset (tau approximately 1 sec at 1 microM omega-CTx-MVIIC) and quick reversibility (tau approximately 30 sec). The rate of block was proportional to toxin concentration, consistent with 1:1 binding of toxin to channels, with a rate constant (k on) of approximately 1 X 10(6) M-1. sec-1. Both potency and rate of block were reduced dramatically with increasing concentrations of extracellular Ba2+ omega-CTx-MVIIC also blocked P-type calcium channels in cerebellar Purkinje neurons, but both development and reversal of block were far slower than for N-type channels. The rate of block was proportional to toxin concentration, with k on -1.5 x 10(3) M-1. sec-1 at 5 mM Ba2+. From this value and an unblocking time constant of approximately 200 min, a dissociation constant of approximately 50 nM was estimated. Thus, block of P-type channels is potent but very slow. In hippocampal CA3 pyramidal neurons, omega-CTx-MVIIC blocked approximately 50% of the high-threshold calcium channel current; one component (approximately 20%) was blocked with the rapid kinetics expected for N-type channels, whereas the other component was blocked slowly. The component blocked slowly was reduced but not eliminated by preexposure to 200 nM or 1 microM omega-Aga-IVA.

A new Conus peptide ligand for mammalian presynaptic Ca2+ channels.[Pubmed:1352986]

Neuron. 1992 Jul;9(1):69-77.

Voltage-sensitive Ca2+ channels that control neurotransmitter release are blocked by omega-conotoxin (omega-CgTx) GVIA from the marine snail Conus geographus, the most widely used inhibitor of neurotransmitter release. However, many mammalian synapses are omega-CgTx-GVIA insensitive. We describe a new Conus peptide, omega-CgTx-MVIIC, that is an effective inhibitor of omega-CgTx-GVIA-resistant synaptic transmission. Ca2+ channel targets that are inhibited by omega-CgTx-MVIIC but not by omega-CgTx-GVIA include those mediating depolarization-induced 45Ca2+ uptake in rat synaptosome preparations, "P" currents in cerebellar Purkinje cells, and a subset of omega-CgTx-GVIA-resistant currents in CA1 hippocampal pyramidal cells. The characterization of omega-CgTx-MVIIC by a combination of molecular genetics and chemical synthesis defines a general approach for obtaining ligands with novel receptor subtype specificity from Conus.

Keywords:

ω-Conotoxin MVIIC,147794-23-8,Natural Products,Calcium Channel, buy ω-Conotoxin MVIIC , ω-Conotoxin MVIIC supplier , purchase ω-Conotoxin MVIIC , ω-Conotoxin MVIIC cost , ω-Conotoxin MVIIC manufacturer , order ω-Conotoxin MVIIC , high purity ω-Conotoxin MVIIC

Online Inquiry for:

      Fill out the information below

      • Size:Qty: - +

      * Required Fields

                                      Result: