CRF (6-33)
Need Assistance?
  • US & Canada:
    +
  • UK: +

CRF (6-33)

* Please kindly note that our products are not to be used for therapeutic purposes and cannot be sold to patients.

CRF (6-33) has been found to be a CRFBP inhibitor peptide and could probably suppress body weight gain.

Category
Peptide Inhibitors
Catalog number
BAT-010260
CAS number
120066-38-8
Molecular Formula
C141H231N41O43S
Molecular Weight
3220.68
CRF (6-33)
IUPAC Name
(4S)-5-[[(2S)-5-amino-1-[[(2S)-1-[[(2S)-1-[[(2S)-5-amino-1-[[(2S)-5-amino-1-[[(2S)-1-[[(2S)-1-[[(1S)-1-carboxy-2-hydroxyethyl]amino]-3-(1H-imidazol-4-yl)-1-oxopropan-2-yl]amino]-1-oxopropan-2-yl]amino]-1,5-dioxopentan-2-yl]amino]-1,5-dioxopentan-2-yl]amino]-1-oxopropan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-1,5-dioxopentan-2-yl]amino]-4-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S,3R)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S,3S)-2-amino-3-methylpentanoyl]amino]-3-hydroxypropanoyl]amino]-4-methylpentanoyl]amino]-3-carboxypropanoyl]amino]-4-methylpentanoyl]amino]-3-hydroxybutanoyl]amino]-3-phenylpropanoyl]amino]-3-(1H-imidazol-4-yl)propanoyl]amino]-4-methylpentanoyl]amino]-4-methylpentanoyl]amino]-5-carbamimidamidopentanoyl]amino]-4-carboxybutanoyl]amino]-3-methylbutanoyl]amino]-4-methylpentanoyl]amino]-4-carboxybutanoyl]amino]-4-methylsulfanylbutanoyl]amino]propanoyl]amino]-5-carbamimidamidopentanoyl]amino]propanoyl]amino]-5-oxopentanoic acid
Synonyms
Corticotropin-Releasing Factor (6-33)
Appearance
White Lyophilized Solid
Purity
>98%
Density
1.5±0.1 g/cm3
Sequence
ISLDLTFHLLREVLEMARAEQLAQQAHS
Storage
Store at -20°C
InChI
InChI=1S/C141H231N41O43S/c1-23-72(16)109(145)136(221)179-100(61-183)135(220)174-93(52-68(8)9)129(214)176-99(58-108(195)196)133(218)173-95(54-70(12)13)134(219)182-111(77(21)185)138(223)178-96(55-78-29-25-24-26-30-78)130(215)175-98(57-80-60-151-64-155-80)131(216)172-92(51-67(6)7)128(213)171-91(50-66(4)5)126(211)163-82(32-28-47-153-141(148)149)119(204)166-88(38-44-107(193)194)124(209)181-110(71(14)15)137(222)177-94(53-69(10)11)127(212)167-87(37-43-106(191)192)122(207)168-89(45-48-226-22)118(203)158-73(17)112(197)160-81(31-27-46-152-140(146)147)116(201)156-74(18)113(198)162-86(36-42-105(189)190)121(206)165-85(35-41-104(144)188)123(208)170-90(49-65(2)3)125(210)159-75(19)114(199)161-84(34-40-103(143)187)120(205)164-83(33-39-102(142)186)117(202)157-76(20)115(200)169-97(56-79-59-150-63-154-79)132(217)180-101(62-184)139(224)225/h24-26,29-30,59-60,63-77,81-101,109-111,183-185H,23,27-28,31-58,61-62,145H2,1-22H3,(H2,142,186)(H2,143,187)(H2,144,188)(H,150,154)(H,151,155)(H,156,201)(H,157,202)(H,158,203)(H,159,210)(H,160,197)(H,161,199)(H,162,198)(H,163,211)(H,164,205)(H,165,206)(H,166,204)(H,167,212)(H,168,207)(H,169,200)(H,170,208)(H,171,213)(H,172,216)(H,173,218)(H,174,220)(H,175,215)(H,176,214)(H,177,222)(H,178,223)(H,179,221)(H,180,217)(H,181,209)(H,182,219)(H,189,190)(H,191,192)(H,193,194)(H,195,196)(H,224,225)(H4,146,147,152)(H4,148,149,153)/t72-,73-,74-,75-,76-,77+,81-,82-,83-,84-,85-,86-,87-,88-,89-,90-,91-,92-,93-,94-,95-,96-,97-,98-,99-,100-,101-,109-,110-,111-/m0/s1
InChI Key
XHHPINGDTRCKNN-QZXFXOMNSA-N
Canonical SMILES
CCC(C)C(C(=O)NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC(=O)O)C(=O)NC(CC(C)C)C(=O)NC(C(C)O)C(=O)NC(CC1=CC=CC=C1)C(=O)NC(CC2=CNC=N2)C(=O)NC(CC(C)C)C(=O)NC(CC(C)C)C(=O)NC(CCCNC(=N)N)C(=O)NC(CCC(=O)O)C(=O)NC(C(C)C)C(=O)NC(CC(C)C)C(=O)NC(CCC(=O)O)C(=O)NC(CCSC)C(=O)NC(C)C(=O)NC(CCCNC(=N)N)C(=O)NC(C)C(=O)NC(CCC(=O)O)C(=O)NC(CCC(=O)N)C(=O)NC(CC(C)C)C(=O)NC(C)C(=O)NC(CCC(=O)N)C(=O)NC(CCC(=O)N)C(=O)NC(C)C(=O)NC(CC3=CNC=N3)C(=O)NC(CO)C(=O)O)N
1. Involvement of CRFR 1 in the Basolateral Amygdala in the Immediate Fear Extinction Deficit
Fiona Hollis,Jocelyn Grosse,Olivia Zanoletti,Yannick Sevelinges,Carmen Sandi eNeuro . 2016 Nov 2;3(5):ENEURO.0084-16.2016. doi: 10.1523/ENEURO.0084-16.2016.
Several animal and clinical studies have highlighted the ineffectiveness of fear extinction sessions delivered shortly after trauma exposure. This phenomenon, termed the immediate extinction deficit, refers to situations in which extinction programs applied shortly after fear conditioning may result in the reduction of fear behaviors (in rodents, frequently measured as freezing responses to the conditioned cue) during extinction training, but failure to consolidate this reduction in the long term. The molecular mechanisms driving this immediate extinction resistance remain unclear. Here we present evidence for the involvement of the corticotropin releasing factor (CRF) system in the basolateral amygdala (BLA) in male Wistar rats. Intra-BLA microinfusion of the CRFR1antagonist NBI30775 enhances extinction recall, whereas administration of the CRF agonist CRF6-33before delayed extinction disrupts recall of extinction. We link the immediate fear extinction deficit with dephosphorylation of GluA1 glutamate receptors at Ser845and enhanced activity of the protein phosphatase calcineurin in the BLA. Their reversal after treatment with the CRFR1antagonist indicates their dependence on CRFR1actions. These findings can have important implications for the improvement of therapeutic approaches to trauma, as well as furthering our understanding of the neurobiological mechanisms underlying fear-related disorders.
2. Stress-induced relapse to cocaine seeking: roles for the CRF(2) receptor and CRF-binding protein in the ventral tegmental area of the rat
Zhi-Bing You,Roy A Wise,Kenner C Rice,Bin Wang Psychopharmacology (Berl) . 2007 Aug;193(2):283-94. doi: 10.1007/s00213-007-0782-3.
Rationale:Footshock reinstates cocaine seeking in cocaine-experienced rats by inducing corticotropin-releasing factor (CRF) and glutamate release in the ventral tegmental area (VTA) and thus activating VTA dopaminergic neurons. Footshock-induced VTA glutamate release, dopamine activation and reinstatements are blocked by VTA administration of a alpha-helical CRF, a nonselective CRF receptor antagonist. The effects of selective CRF antagonists have not yet been reported.Objective:The present studies were designed to explore the roles of VTA CRF receptor subtypes and CRF-BP in these effects induced by footshock.Methods:Rats were first trained to lever-press for intravenous cocaine (1 mg/infusion/0.13 ml, FR-1 schedule), and then tested under extinction conditions until response rates returned to the pretraining baseline. Reinstatements, VTA glutamate and dopamine levels [microdialysis with high performance liquid chromatography (HPLC)] were then assessed, under various pharmacological conditions, after mild inescapable footshock.Results:Footshock-induced reinstatement of cocaine seeking and release of VTA glutamate and dopamine were blocked by selective blockade of VTA CRF(2) receptors (CRF(2)Rs) but not CRF(1)Rs. VTA perfusion of CRF or CRF(2)R agonists that have strong affinity for CRF-BP mimicked the effects induced by footshock while CRFR agonists that do not bind CRF-BP were ineffective. CRF(6-33), which competes for the CRF binding site on CRF-BP, attenuated the effects of CRF or urocortin I on VTA glutamate and dopamine release and on reinstatement of cocaine seeking.Conclusions:The present studies revealed a role of VTA CRF-BP and suggest an involvement of CRF(2)R in the effectiveness of stress in triggering glutamate and dopamine release and cocaine seeking in drug-experienced animals.
3. Corticotropin-releasing factor (CRF) or CRF binding-protein ligand inhibitor administration suppresses food intake in mice and elevates body temperature in rats
S Iyengar,D L Li,S C Heinrichs Brain Res . 2001 May 11;900(2):177-85. doi: 10.1016/s0006-8993(01)02286-7.
Corticotropin-releasing factor (CRF) receptor agonist and CRF binding-protein (CRF-BP) ligand inhibitor peptides both activate CRF systems but exert very distinct functional profiles in animal models of arousal, energy balance and emotionality. The present studies were designed to extend the dissimilar efficacy profiles of central administration of a CRF agonist, r/h CRF(1-41), versus a CRF-BP ligand inhibitor, r/h CRF(6-33), into mouse and rat models of energy balance in order to further explore in vivo efficacy of these ligands in two separate animal species. In CD-1 mice, food intake was significantly attenuated 3 h after acute administration of CRF(1-41) (0.007-0.2 nmol), but not CRF(6-33). In obese Ob/Ob mice, both CRF(1-41) (0.007-0.2 nmol) and CRF(6-33) (0.02-2.3 nmol) significantly attenuated basal feeding over 3 h following acute peptide administration. In rats, CRF(1-41) (1 nmol) and CRF(6-33) (1.5-7.7 nmol) infusion significantly increased rectal temperature. In studies employing a telemetry apparatus, core temperature was also increased by CRF(1-41) (1 nmol) and CRF(6-33) (1.5 nmol), whereas only CRF(1-41) increased locomotor activity and heart rate. These results suggest that CRF receptor agonist administration is capable of producing a global profile of negative energy balance by reducing food intake in mice and increasing energy expenditure in rats. In contrast, CRF-BP ligand inhibitor administration appears to suppress food intake in a mouse strain selective manner and to elevate rectal and core temperature in rats without accompanying cardiovascular activation.
4. Brain CRF-binding protein modulates aspects of maternal behavior under stressful conditions and supports a hypo-anxious state in lactating rats
Stefanie M Klampfl,Oliver J Bosch,Milena M Schramm,Gwen S Stinnett,Doris S Bayerl,Audrey F Seasholtz Horm Behav . 2016 Aug;84:136-44. doi: 10.1016/j.yhbeh.2016.06.009.
Reduced corticotropin-releasing factor (CRF) receptor activation in the postpartum period is essential for adequate maternal behavior. One of the factors contributing to this hypo-activity might be the CRF-binding protein (CRF-BP), which likely reduces the availability of free extracellular CRF/urocortin 1. Here, we investigated behavioral effects of acute CRF-BP inhibition using 5μg of CRF(6-33) administered either centrally or locally within different parts of the bed nucleus of the stria terminalis (BNST) in lactating rats. Additionally, we assessed CRF-BP expression in the BNST comparing virgin and lactating rats. Central CRF-BP inhibition increased maternal aggression during maternal defense but did not affect maternal care or anxiety-related behavior. CRF-BP inhibition in the medial-posterior BNST had no effect on maternal care under non-stress conditions but impaired the reinstatement of maternal care following stressor exposure. Furthermore, maternal aggression, particularly threat behavior, and anxiety-related behavior were elevated by CRF-BP inhibition in the medial-posterior BNST. In the anterior-dorsal BNST, CRF-BP inhibition increased only non-maternal behaviors following stress. Finally, CRF-BP expression was higher in the anterior compared to the posterior BNST but was not different between virgin and lactating rats in either region. Our study demonstrates a key role of the CRF-BP, particularly within the BNST, in modulating CRF's impact on maternal behavior. The CRF-BP is important for the reinstatement of maternal care after stress, for modulating threat behavior during an aggressive encounter and for maintaining a hypo-anxious state during lactation. Thus, the CRF-BP likely contributes to the postpartum-associated down-regulation of the CRF system in a brain region-dependent manner.
Online Inquiry
Verification code
Inquiry Basket