PreprintPDF Available

Human blood plasma proteins modeling and binding affinities with Δ9-tetrahydrocannabinol active metabolites: In silico approach

Authors:
Preprints and early-stage research may not have been peer reviewed yet.

Abstract and Figures

Tetrahydrocannabinol (THC) is a key psychotropic constituent of cannabis sativa. It is also known as Δ9-tetrahydrocannabinol (Δ9-THC). Previous study suggested that owing to its high lipophilicity, it piles up in adipose tissue and it is disseminated into blood stream for prolonged time. Research suggests that numerous diseases such as multiple sclerosis, neurodegenerative disorders, epilepsy, schizophrenia, osteoporosis, cancer, glaucoma and cardiovascular disorders can be treated using this substance. However, apart from having therapeutic potential, many studies have reported detrimental outcomes along with addiction of Δ9-THC for short-term and long-term consumption. Thus, in this study, we determined the binding affinities of Δ9-THC and its two active metabolites, 11-Hydroxy-Δ9-tetrahydrocannabinol (11-OH-Δ9-THC) and 8beta,11-dihydroxy-Δ9-tetrahydrocannabinol (8β,11-diOH-Δ9-THC) with 401 human blood plasma proteins using molecular docking analysis. Results show that Δ9-THC has greater binding potential with plasma proteins as compared to other two metabolites. Overall, ADGRE5, ALB, APOA5, APOD, CP, PON1 and PON3 proteins showed the highest binding affinities with three cannabis metabolites.
Content may be subject to copyright.
Human blood plasma proteins modeling and binding
affinities with Δ9-tetrahydrocannabinol active metabolites:
In silico approach
Shravan B. Rathoda,*, Jinal C. Sonib, Priyanshu Vermac,#, Yogita Rawatd,#, Neha
Periwald, Pooja Arorac, Vikas Soodd , Mohmedyasin F. Mansurib
aDepartment of Chemistry, Smt. S. M. Panchal Science College, Talod, Gujarat, India
bDepartment of Microbiology, Smt. S. M. Panchal Science College, Talod, Gujarat, India
cDepartment of Zoology, Hansraj College, University of Delhi, New Delhi, India
dDepartment of Biochemistry, School of Chemical & Life Sciences, Jamia Hamdard, New
Delhi, India
#Equal contribution
*Corresponding author
Shravan B. Rathod: E-mail: shravanathorizon93@gmail.com
Phone: +91-8200040941
ORCID: https://orcid.org/0000-0002-1870-2508
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Abstract
Tetrahydrocannabinol (THC) is a key psychotropic constituent of cannabis
sativa. It is also known as Δ9-tetrahydrocannabinol 9-THC). Previous study
suggested that owing to its high lipophilicity, it piles up in adipose tissue and it is
disseminated into blood stream for prolonged time. Research suggests that
numerous diseases such as multiple sclerosis, neurodegenerative disorders,
epilepsy, schizophrenia, osteoporosis, cancer, glaucoma and cardiovascular
disorders can be treated using this substance. However, apart from having
therapeutic potential, many studies have reported detrimental outcomes along
with addiction of Δ9-THC for short-term and long-term consumption. Thus, in
this study, we determined the binding affinities of Δ9-THC and its two active
metabolites, 11-Hydroxy-Δ9-tetrahydrocannabinol (11-OH-Δ9-THC) and
8beta,11-dihydroxy-Δ9-tetrahydrocannabinol (8β,11-diOH-Δ9-THC) with 401
human blood plasma proteins using molecular docking analysis. Results show
that Δ9-THC has greater binding potential with plasma proteins as compared to
other two metabolites. Overall, ADGRE5, ALB, APOA5, APOD, CP, PON1 and
PON3 proteins showed the highest binding affinities with three cannabis
metabolites.
Keywords: Tetrahydrocannabinol (THC), Psychoactive, Plasma proteins,
Molecular docking, Blind docking, Binding affinity
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Introduction
The consumption of cannabis in teenagers has surged significantly and, its
adverse effects on mental health and political debate on whether make it legalise
or not need urgent molecular level study of cannabis metabolites [1]. Even though
epidemiological studies reported the risk of overconsumption of cannabis during
adolescence leads to neuropsychiatric disorders in their later life, it still remains
widely used drug illicit [24]. Researchers have shown that administration of a
key ingredient, Δ9-tetrahydrocannabinol (Δ9-THC) of cannabis in animals during
their adolescence, induced biochemical and behavioural signs of psychosis and
depression later in life [510].
Interestingly, it was noted that the exposure of THC to female rats during
adolescence is linked with epigenetic modification of histone H3 in Prefrontal
cortex (PFC). Specifically, trimethylation at Lys9 position in N-terminus of
histone H3. This alternation in histone H3 causes impacts on genes which are
closely linked to the neuroplasticity and Endocannabinoid system (ECS)
mechanisms that ultimately leads to diseases. However, THC exposure to adults
results into minor effects on epigenome[1]. Thus, these findings suggest that the
risks of neuropsychiatric disorders, CNS alternation and the cognitive impairment
are escalated during adulthood due to the consumption of cannabis in adolescence
[11]. Another study revealed that Δ9-THC activates the presynaptic Cannabinoid
receptor type 1 (CB1) receptor which is key player in CNS development during
prenatal, postnatal and adolescence periods [1214]. The CB1 is G protein-
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
coupled receptors (GPCR) and found in glial cells and neurons and cells [15].
Another, Cannabinoid receptors type 2 (CB2) is also GPCR which is generally
located in hematopoietic cells and some specific regions of the brain and
peripheral cells have CB2 receptor. CB2 plays an important role in immunity
pathways and its activation leads to immunomodulatory and anti-inflammatory
response. CB1 pathways are activated through the stimulation of CB2 [16,17].
Further, Abey N. O. investigated the effects of Δ9-THC on blood chemistry and
organs cytoarchitecture and, it was observed significant decline in brain cognitive
function, brain total proteins, and nitric oxide. Additionally, statistically
significant difference were observed in various tissues and blood plasma [18].
Furthermore, computational and in vitro studies of Δ9-THC unveiled that this
constituent strongly binds to three Fatty acid-binding proteins (FABPs), FABP3,
FABP5 and FABP7 [19]. According to Schenk S. et al. analysis, they have
validated and reported that there are 1193 different proteins are present in human
blood plasma [20]. Literature review suggests that significant research has not
been yet done on the binding profile of Δ9-THC and its metabolites with human
blood plasma proteins. Hence, in this study, we determined the binding affinities
of Δ9-THC and its two active metabolites, 11-OH-Δ9-THC and 8β,11-diOH-Δ9-
THC with 401 plasma proteins using molecular docking approach. Fig. 1 shows
the chemical structures of these three compounds.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Materials and methods
Protein modeling and ligand preparation
Initially, the database of human blood plasma proteins detected by immunoassay
was searched from The Human Protein Atlas
(https://www.proteinatlas.org/humanproteome/blood+protein/proteins+detected
+by+immunoassay) web portal. The total number of proteins were 419 on the
database. Many protein structures were available on the Protein data bank
(https://www.rcsb.org/) but large number of proteins had missing residues thus,
we employed newly developed neural network-based RoseTTAFold [21] protein
prediction tool (https://robetta.bakerlab.org/). This web tool has limitation that it
accepts only proteins which have residues range between 26 and 1201. Thus, out
of 419 plasma proteins, 18 proteins were excluded due to the lack of structure
prediction. These 401 predicted structures were downloaded and further used to
perform molecular docking analysis. The PDB structures of 401 human blood
plasma proteins are given in Supplementary file 1.
In case of ligands, we considered Δ9-tetrahydrocannabinol 9-THC: PubChem
CID- 16078) and its two active metabolites, 11-Hydroxy-Δ9-
tetrahydrocannabinol (11-OH-Δ9-THC: PubChem CID- 37482) and 8,11-
dihydroxy-Δ9-tetrahydrocannabinol (8β,11-diOH-Δ9-THC: PubChem CID-
126961369). Firstly, 3D structures of these ligands were retrieved from the
PubChem Database (https://pubchem.ncbi.nlm.nih.gov/) in sdf format. Then, for
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
the energy minimization, geometry optimization was performed on these ligands
using Auto Optimization Tool available in Avogadro software [22]. The
optimization was carried out by employing MMFF94s [23] force field and
Steepest descent [24] algorithm. Finally, the structures were saved as mol2 format
for further docking study.
Molecular docking
To determine the binding affinities of three THC metabolites with 401 human
blood plasma proteins, we utilized protein cavity-find guided Autodock Vina-
based blind docker, CB-DOCK [25] web server available at
http://clab.labshare.cn/cb-dock/php/. To calculate the binding affinity (Docking
score) of these three metabolites with 401 plasma proteins, protein as a PDB and
ligand as a mol2 format were uploaded to the server. We ran total 1203 (3*401)
jobs at the server. Initially, this tool searches top five binding cavities inside the
protein and orders them on the basis of cavity size. Then, it starts docking of
ligand with receptor and calculate the binding affinity for best pose inside each
cavity. Additionally, server provides details of cavity (volume, centre and size
coordinates) along with binding affinity (Vina score). Further, the interactions
analysis was carried out using Discovery Studio v.20.1 tool and UCSF Chimera
v.1.15 was used to make figures of top two complexes. The Electrostatic potential
(ESP) surface analysis of proteins was performed using PyMOL APBS
Electrostatics plugin.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Results and discussion
Molecular docking is a powerful computational approach to make drug discovery
processes fast and less expensive. Molecular docking has many success stories
for the designing inhibitors against HIV-1, cancer and bacterial targets [2629].
Hence, for the primary investigation of binding affinities of Δ9-THC and its two
active metabolites (11-OH-Δ9-THC and 8β,11-diOH-Δ9-THC) with human blood
plasma proteins, we used blind docking approach.
Docking results show that binding affinities of Δ9-THC, 11-OH-Δ9-THC and
8β,11-diOH-Δ9-THC with 401 human blood plasma proteins vary between -5.0
and -10.5 kcal/mol. However, Δ9-THC showed the greater binding affinities as
compared to other two metabolites (Fig. 2). This indicates that plasma proteins
have overall hydrophobic nature as they have highest binding affinity with Δ9-
THC. Δ9-THC is less polar (more hydrophobic) than 11-OH-Δ9-THC and 8β,11-
diOH-Δ9-THC (Fig. 1). The binding affinities of all these three metabolites are
given in Supplementary Table S1. Binding affinities of top 20 complexes are
shown in Table 1.
Additionally, we have carried out interaction analysis of top-two complexes of
each metabolite. In case of Δ9-THC, Apolipoprotein A5 (APOA5) &
Apolipoprotein D (APOD) have the large value of docking score, -10.3 & -10.1
kcal/mol respectively (Table 1). The structure and interaction plot of APOA5:Δ9-
THC and APOD:Δ9-THC complexes are illustrated in Fig. 3. It has been reported
that APOA5 plays vital role to metabolize triglyceride and triglyceride-rich
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
lipoproteins. Current findings suggest the link between APOA5 and obesity [30].
Apolipoprotein D (APOD) is structurally different from the other apolipoproteins
and it is lipocalin family member. It is widely expressed in glial cells and neurons
of peripheral and central nervous system and carrier of tiny lipophilic candidates.
It is not only transporter but has vital role in modulating the oxidation condition
and stability of lipophilic molecules. Previous studies suggest that APOD was
upregulated in stroke, schizophrenia and Alzheimer’s disease conditions. Thus, it
has neuroprotective functions [31]. APOD forms barrel with eight antiparallel β-
strands and has α-helices at the side. It has around ~15 Å deep hydrophobic cavity
and entry diameter is also 15 Å (Fig. 3C) [32].
Fig. 3A & 3C represent the structure of APOA5:Δ9-THC and APOD:Δ9-THC
complexes. It can be seen from the Fig. 3B that in APOA5:Δ9-THC complex,
Val7, Trp10, Ala11, Gly26, Phe27, Leu56, Leu60, Leu96, Leu100 and Val103
residues have hydrophobic interactions such as π-alkyl and alkyl-alkyl. A single
hydrogen bond is formed by Asp63 and hydroxyl group of Δ9-THC. Whereas, in
APOD:Δ9-THC complex, Val, Phe, Leu, Tyr and Ile are the common residues
which have hydrophobic interaction with ligand (Fig. 3D). The residues from the
APOD interact with Δ9-THC are similar to the previously reported catalytic site
residues. However, the number of residues interacting with Δ9-THC in APOD:Δ9-
THC complex is slightly higher as compared to residues in APOA59-THC
complex.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Considering 11-OH-Δ9-THC metabolite, it can be observed (Table 1) that
Adhesion G protein-coupled receptor E5 (ADGRE5) and Ceruloplasmin (CP)
have better affinities around -9.5 kcal/mol in comparison with other proteins. The
ADGRE5 is the subclass of G-protein coupled receptors (GPCRs) that has
pharmacological significance [33]. It is also known as Cluster of differentiation
97 (CD97). This protein is broadly expressed muscle, immune, stem and
progenitor cells [34-36]. Additionally, recent study reported that ADGRE5/CD97
helps to suppress Nuclear factor kappa B (NF-κB) through upregulating the
Peroxisome proliferator- activated receptor gamma (PPAR-γ) in leukocytes [37].
Some studies also revealed that CD97 was found overexpressed in various
tumours and attunes tumorigenesis [38].
The Ceruloplasmin (CP) belongs to oxidase family and copper enzyme present in
serum [39]. In neurodegenerative diseases such as Parkinson's and Alzheimer's,
CP was found in Cerebrospinal fluid (CSF) with modified structure and state [40].
Like APOD, CP has also neuroprotective role [41]. In CP, six cupredoxin
domains are arranged side-by-side and form a cavity. CP has trinuclear copper
cluster and mononuclear copper centre with a 12-13 Å distance between them.
The trinuclear copper cluster is formed by three copper ions which are located in
three even domains. This cluster has also one type II (T2) and two type III (T3)
copper ions [42].
Complex structures of 11-OH-Δ9-THC with ADGRE5 and CP are given in Fig.
4A and C respectively. While the interactions maps are shown in Fig. 4B and D
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
respectively. In ADGRE5 complex, none of the hydroxyl groups have polar
interactions with surrounding residues. All interactions are hydrophobic in nature.
Phe623 and Phe771 have π-π stacking interactions with aromatic ring in ligand.
Remaining interactions come from hydrophobic amino acids such as Phe597,
Leu686, Leu697, Val700, Leu757, Ile759 and Phe760 (Fig. 4B). However, 11-
OH-Δ9-THC has polar interactions with CP. Lys288 forms hydrogen bond with
hydroxyl group. Arg649 interacts with ligand through π-cation interaction.
Phe659, Leu664 and Tyr986 have hydrophobic -alkyl & alkyl-alkyl)
interactions (Fig. 4D). Additionally, Asn287 has unfavourable contact with
hydroxyl group of aromatic ring due to its donor nature and steric repulsion.
Finally, APOA5 and ADGRE5 were observed to have the large binding affinities
(~-9.0 kcal/mol) with third metabolite, 8β,11-diOH-Δ9-THC. The structures of
APOA5:8β,11-diOH-Δ9-THC and ADGRE5:8β,11-diOH-Δ9-THC complexes
are illustrated in Fig. 5A and 5C respectively whereas their interactions maps are
shown in Fig. 5B and 5D respectively. In these complexes, hydrophobic
interactions have been found dominant but both have polar interactions also. In
APOA5:8β,11-diOH-Δ9-THC, Lys48 and Asp63 are forming hydrogen bonds
with the hydroxyl groups of ligand. And, Trp10, Ala11, Leu14, Tyr30 and Leu96
interact through alkyl-alkyl hydrophobic interactions (Fig. 5B). Also, it has a
single unfavourable interaction with Asp63. But, in ADGRE5:8β,11-diOH-Δ9-
THC, the hydrogen bond is formed between Asn775 and aromatic hydroxyl group
and, carbon-hydrogen (C-H) polar hydrogen bond between Thr772 and
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
methylene group carbon where hydroxyl group is attached (Fig. 5D). Two
aromatic residues, Phe623 and Phe771 show π-π stacking interactions with
aromatic ring of ligand. And, large contribution comes from hydrophobic
residues, Leu686, Leu697, Val700, Ile759 and Phe760 (Fig. 5D).
Furthermore, we have also carried out ESP analysis for APOA5, APOD,
ADGRE5 and CP to probe the ligand binding cavity electrostatic properties. Fig.
6A-D illustrate the hydrophobic nature of cavities in each four plasma proteins,
APOA5, APOD, ADGRE5 and CP respectively.
Conclusion
The principal constituent of cannabis, Δ9-THC has therapeutic significance as
well as some obscurity in adverse impacts on health. Owing to the lipophilic
nature of Δ9-THC, it can bind various human blood plasma proteins. Hence, we
investigated the bindings of Δ9-THC and its two active metabolites (11-OH-Δ9-
THC & 8β,11-diOH-Δ9-THC) with 401 human blood plasma proteins using blind
docking CB-DOCK tool. Further, we performed interactions analysis of top-two
complexes of each constituent with plasma proteins. Results suggest that Δ9-THC
showed higher tendency towards plasma protein binding as compared to its two
metabolites. The blood plasma proteins such as, ADGRE5, ALB, APOA5,
APOD, CP, PON1 and PON3 have suitable binding cavities to adapt Δ9-THC and
its active metabolites in comparison with remaining plasma proteins. Our study
is a primary investigation of binding affinity between Δ9-THC and plasma
proteins. However, our study provides input to further probe the structural,
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
functional and dynamic impacts of these metabolites on blood plasma proteins
leading to adverse impacts of these cannabinoids on human health by performing
further studies.
Declaration of competing interest
No potential conflict of interest was reported by the author(s).
Acknowledgements
SBR is thankful to his Chemistry Department for providing computational
facilities and infrastructure.
References
[1] P. Prini, F. Rusconi, E. Zamberletti, M. Gabaglio, F. Penna, M. Fasano, E.
Battaglioli, D. Parolaro, T. Rubino, Adolescent THC exposure in female
rats leads to cognitive deficits through a mechanism involving chromatin
modifications in the prefrontal cortex, J. Psychiatry Neurosci. 43 (2018)
87101. https://doi.org/10.1503/jpn.170082.
[2] S. Lev-Ran, M. Roerecke, B. Le Foll, T.P. George, K. McKenzie, J.
Rehm, The association between cannabis use and depression: A
systematic review and meta-analysis of longitudinal studies, Psychol.
Med. 44 (2014) 797810. https://doi.org/10.1017/S0033291713001438.
[3] M. Di Forti, H. Sallis, F. Allegri, A. Trotta, L. Ferraro, S.A. Stilo, A.
Marconi, C. La Cascia, T.R. Marques, C. Pariante, P. Dazzan, V.
Mondelli, A. Paparelli, A. Kolliakou, D. Prata, F. Gaughran, A.S. David,
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
C. Morgan, D. Stahl, M. Khondoker, J.H. MacCabe, R.M. Murray, Daily
use, especially of high-potency cannabis, drives the earlier onset of
psychosis in cannabis users, Schizophr. Bull. 40 (2014) 15091517.
https://doi.org/10.1093/schbul/sbt181.
[4] S.T. Wilkinson, R. Radhakrishnan, D.C. D’Souza, Impact of Cannabis
Use on the Development of Psychotic Disorders, Curr. Addict. Reports. 1
(2014) 115128. https://doi.org/10.1007/s40429-014-0018-7.
[5] T. Rubino, D. Vigano’, N. Realini, C. Guidali, D. Braida, V. Capurro, C.
Castiglioni, F. Cherubino, P. Romualdi, S. Candeletti, M. Sala, D.
Parolaro, Chronic Δ9-tetrahydrocannabinol during adolescence provokes
sex-dependent changes in the emotional profile in adult rats: Behavioral
and biochemical correlates, Neuropsychopharmacology. 33 (2008) 2760
2771. https://doi.org/10.1038/sj.npp.1301664.
[6] T. Rubino, N. Realini, D. Braida, T. Alberio, V. Capurro, D. Viganò, C.
Guidali, M. Sala, M. Fasano, D. Parolaro, The Depressive Phenotype
Induced in Adult Female Rats by Adolescent Exposure to THC is
Associated with Cognitive Impairment and Altered Neuroplasticity in the
Prefrontal Cortex, Neurotox. Res. 15 (2009) 291302.
https://doi.org/10.1007/s12640-009-9031-3.
[7] T. Rubino, P. Prini, F. Piscitelli, E. Zamberletti, M. Trusel, M. Melis, C.
Sagheddu, A. Ligresti, R. Tonini, V. Di Marzo, D. Parolaro, Adolescent
exposure to THC in female rats disrupts developmental changes in the
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
prefrontal cortex, Neurobiol. Dis. 73 (2015) 6069.
https://doi.org/10.1016/j.nbd.2014.09.015.
[8] N. Realini, D. Vigano’, C. Guidali, E. Zamberletti, T. Rubino, D.
Parolaro, Chronic URB597 treatment at adulthood reverted most
depressive-like symptoms induced by adolescent exposure to THC in
female rats, Neuropharmacology. 60 (2011) 235243.
https://doi.org/10.1016/j.neuropharm.2010.09.003.
[9] E. Zamberletti, S. Beggiato, L. Steardo, P. Prini, T. Antonelli, L. Ferraro,
T. Rubino, D. Parolaro, Neurobiology of Disease Alterations of prefrontal
cortex GABAergic transmission in the complex psychotic-like phenotype
induced by adolescent delta-9-tetrahydrocannabinol exposure in rats,
Neurobiol. Dis. 63 (2014) 3547.
https://doi.org/10.1016/j.nbd.2013.10.028.
[10] E. Zamberletti, M. Gabaglio, P. Prini, T. Rubino, D. Parolaro, Cortical
neuroin fl ammation contributes to long-term cognitive dysfunctions
following adolescent delta-9-tetrahydrocannabinol treatment in female
rats, Eur. Neuropsychopharmacol. 25 (2015) 24042415.
https://doi.org/10.1016/j.euroneuro.2015.09.021.
[11] J. Renard, M.O. Krebs, G. Le Pen, T.M. Jay, Long-term consequences of
adolescent cannabinoid exposure in adult psychopathology, Front.
Neurosci. 8 (2014) 114. https://doi.org/10.3389/fnins.2014.00361.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
[12] V. Di Marzo, The endocannabinoid system: Its general strategy of action,
tools for its pharmacological manipulation and potential therapeutic
exploitation, Pharmacol. Res. 60 (2009) 7784.
https://doi.org/10.1016/j.phrs.2009.02.010.
[13] T. Harkany, M. Guzmán, I. Galve-Roperh, P. Berghuis, L.A. Devi, K.
Mackie, The emerging functions of endocannabinoid signaling during
CNS development, Trends Pharmacol. Sci. 28 (2007) 8392.
https://doi.org/10.1016/j.tips.2006.12.004.
[14] M. Ellgren, A. Artmann, O. Tkalych, A. Gupta, H.S. Hansen, S.H.
Hansen, L.A. Devi, Y.L. Hurd, Dynamic changes of the endogenous
cannabinoid and opioid mesocorticolimbic systems during adolescence:
THC effects, Eur. Neuropsychopharmacol. 18 (2008) 826834.
https://doi.org/10.1016/j.euroneuro.2008.06.009.
[15] L.E. Klumpers, D.L. Thacker, A brief background on cannabis: From
plant to medical indications, J. AOAC Int. 102 (2019) 412420.
https://doi.org/10.5740/jaoacint.18-0208.
[16] R.G. Pertwee, Receptors and channels targeted by synthetic cannabinoid
receptor agonists and antagonists. Curr Med Chem. 17 (2010) 1360-1381.
http://dx.doi.org/10.2174/092986710790980050
[17] E.D. Gonçalves, R.C. Dutra, Cannabinoid receptors as therapeutic targets
for autoimmune diseases: where do we stand?, Drug Discov. Today. 24
(2019) 18451853. https://doi.org/10.1016/j.drudis.2019.05.023.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
[18] N.O. Abey, Cannabis sativa (Marijuana) alters blood chemistry and the
cytoarchitecture of some organs in Sprague Dawley rat models, Food
Chem. Toxicol. 116 (2018) 292297.
https://doi.org/10.1016/j.fct.2018.04.023.
[19] M.W. Elmes, M. Kaczocha, W.T. Berger, K.N. Leung, B.P. Ralph, L.
Wang, J.M. Sweeney, J.T. Miyauchi, S.E. Tsirka, I. Ojima, D.G. Deutsch,
Fatty acid-binding proteins (FABPs) are intracellular carriers for Δ9-
tetrahydrocannabinol (THC) and cannabidiol (CBD), J. Biol. Chem. 290
(2015) 87118721. https://doi.org/10.1074/jbc.M114.618447.
[20] S. Schenk, G.J. Schoenhals, G. de Souza, M. Mann, A high confidence,
manually validated human blood plasma protein reference set, BMC Med.
Genomics. 1 (2008). https://doi.org/10.1186/1755-8794-1-41.
[21] M. Baek, F. DiMaio, I. Anishchenko, J. Dauparas, S. Ovchinnikov, G.R.
Lee, J. Wang, Q. Cong, L.N. Kinch, R. Dustin Schaeffer, C. Millán, H.
Park, C. Adams, C.R. Glassman, A. DeGiovanni, J.H. Pereira, A. V.
Rodrigues, A.A. Van Dijk, A.C. Ebrecht, D.J. Opperman, T. Sagmeister,
C. Buhlheller, T. Pavkov-Keller, M.K. Rathinaswamy, U. Dalwadi, C.K.
Yip, J.E. Burke, K. Christopher Garcia, N. V. Grishin, P.D. Adams, R.J.
Read, D. Baker, Accurate prediction of protein structures and interactions
using a three-track neural network, Science. 373 (2021) 871876.
https://doi.org/10.1126/science.abj8754.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
[22] M.D. Hanwell, D.E. Curtis, D.C. Lonie, T. Vandermeersch, E. Zurek, G.R.
Hutchison, Avogadro: an advanced semantic chemical editor,
visualization, and analysis platform, J Cheminform. 4 (2012) 1-17.
https://doi.org/10.1186/1758-2946-4-17.
[23] T.A. Halgren, Performance of MMFF94*, J. Comput. Chem. 17 (1996)
490519. http://journals.wiley.com/jcc.
[24] C. Chen, Y. Huang, X. Ji, Y. Xiao, Efficiently finding the minimum free
energy path from steepest descent path, J. Chem. Phys. 138 (2013) 19.
https://doi.org/10.1063/1.4799236.
[25] Y. Liu, M. Grimm, W. tao Dai, M. chun Hou, Z.X. Xiao, Y. Cao, CB-
Dock: a web server for cavity detection-guided proteinligand blind
docking, Acta Pharmacol. Sin. 41 (2020) 138144.
https://doi.org/10.1038/s41401-019-0228-6.
[26] R. Dayam, N. Neamati, Active site binding modes of the β-diketoacids: A
multi-active site approach in HIV-1 integrase inhibitor design, Bioorganic
Med. Chem. 12 (2004) 63716381.
https://doi.org/10.1016/j.bmc.2004.09.035.
[27] A. Kazi, H. Lawrence, W.C. Guida, M.L. McLaughlin, G.M. Springett, N.
Berndt, R.M.L. Yip, S.M. Sebti, Discovery of a novel proteasome
inhibitor selective for cancer cells over non-transformed cells, Cell Cycle.
8 (2009) 19401951. https://doi.org/10.4161/cc.8.12.8798.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
[28] N.A. Kahlous, M.A.M. Bawarish, M.A. Sarhan, M. Küpper, A. Hasaba,
M. Rajab, Using Chemoinformatics, Bioinformatics, and Bioassay to
Predict and Explain the Antibacterial Activity of Nonantibiotic Food and
Drug Administration Drugs, Assay Drug Dev. Technol. 15 (2017) 89105.
https://doi.org/10.1089/adt.2016.771.
[29] C.R. Oliva, W. Zhang, C. Langford, M.J. Suto, C.E. Griguer,
Repositioning chlorpromazine for treating chemoresistant glioma through
the inhibition of cytochrome c oxidase bearing the COX4-1 regulatory
subunit, Oncotarget. 8 (2017) 3756837583.
https://doi.org/10.18632/oncotarget.17247.
[30] X. Su, Y. Kong, D.Q. Peng, New insights into apolipoprotein A5 in
controlling lipoprotein metabolism in obesity and the metabolic syndrome
patients, Lipids Health Dis. 17 (2018) 110.
https://doi.org/10.1186/s12944-018-0833-2.
[31] S. Dassati, A. Waldner, R. Schweigreiter, Apolipoprotein D takes center
stage in the stress response of the aging and degenerative brain,
Neurobiol. Aging. 35 (2014) 16321642.
https://doi.org/10.1016/j.neurobiolaging.2014.01.148.
[32] A. Eichinger, A. Nasreen, J.K. Hyun, A. Skerra, Structural insight into the
dual ligand specificity and mode of high density lipoprotein association of
apolipoprotein D, J. Biol. Chem. 282 (2007) 3106831075.
https://doi.org/10.1074/jbc.M703552200.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
[33] J. Hamann, G. Aust, D. Araç, F.B. Engel, C. Formstone, R. Fredriksson,
R.A. Hall, B.L. Harty, C. Kirchhoff, B. Knapp, A. Krishnan, I. Liebscher,
H.H. Lin, D.C. Martinelli, K.R. Monk, M.C. Peeters, X. Piao, S. Prömel,
T. Schöneberg, T.W. Schwartz, K. Singer, M. Stacey, Y.A. Ushkaryov,
M. Vallon, U. Wolfrum, M.W. Wright, L. Xu, T. Langenhan, H.B.
Schiöth, International union of basic and clinical pharmacology. XCIV.
adhesion G protein-coupled receptors, Pharmacol. Rev. 67 (2015) 338
367. https://doi.org/10.1124/pr.114.009647.
[34] M. Van Pel, H. Hagoort, J. Hamann, W.E. Fibbe, CD97 is differentially
expressed on murine hematopoietic stem- and progenitor-cells,
Haematologica. 93 (2008) 11371144.
https://doi.org/10.3324/haematol.12838.
[35] G. Aust, E. Wandel, C. Boltze, D. Sittig, A. Schütz, L.C. Horn, M. Wobus,
Diversity of CD97 in smooth muscle cells, Cell Tissue Res. 324 (2006)
139147. https://doi.org/10.1007/s00441-005-0103-2.
[36] L.H. Jaspars, W. Vos, G. Aust, R.A.W. Van Lier, J. Hamann, Tissue
distribution of the human CD97 EGF-TM7 receptor, Tissue Antigens. 57
(2001) 325331. https://doi.org/10.1034/j.1399-0039.2001.057004325.x.
[37] S. Wang, Z. Sun, W. Zhao, Z. Wang, M. Wu, Y. Pan, H. Yan, J. Zhu,
CD97/ADGRE5 Inhibits LPS Induced NF- B Activation through PPAR- γ
Upregulation in Macrophages, Mediators Inflamm. 2016 (2016) 1-10.
https://doi.org/10.1155/2016/1605948.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
[38] W.Y. Tjong, H.H. Lin, The role of the RGD motif in CD97/ADGRE5-and
EMR2/ADGRE2-modulated tumor angiogenesis, Biochem. Biophys. Res.
Commun. 520 (2019) 243249.
https://doi.org/10.1016/j.bbrc.2019.09.113.
[39] N.E. Hellman, J.D. Gitlin, Ceruloplasmin metabolism and function, Annu.
Rev. Nutr. 22 (2002) 439458.
https://doi.org/10.1146/annurev.nutr.22.012502.114457.
[40] A. Zanardi, M. Alessio, Ceruloplasmin deamidation in neurodegeneration:
From loss to gain of function, Int. J. Mol. Sci. 22 (2021) 113.
https://doi.org/10.3390/ijms22020663.
[41] B. Wang, X.-P. Wang, Does Ceruloplasmin Defend Against
Neurodegenerative Diseases?, Curr. Neuropharmacol. 17 (2018) 539549.
https://doi.org/10.2174/1570159x16666180508113025.
[42] I. Bento, C. Peixoto, V.N. Zaitsev, P.F. Lindley, Ceruloplasmin revisited:
Structural and functional roles of various metal cation-binding sites, Acta
Crystallogr. Sect. D Biol. Crystallogr. 63 (2007) 240248.
https://doi.org/10.1107/S090744490604947X.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Table 1
Binding affinities of top 20 complexes of Δ9-THC and its two metabolites (Ligands) with
human blood plasma proteins.
Sr.
No.
Ligands
Δ9-THC
11-OH-Δ9-THC
8β,11-diOH-Δ9-THC
Protein
Binding
affinity
(kcal/mol)
Binding
affinity
(kcal/mol)
Protein
Binding
affinity
(kcal/mol)
1
APOA5
-10.3
-9.6
APOA5
-9.3
2
APOD
-10.1
-9.5
ADGRE5
-9.1
3
PON1
-9.5
-8.9
APOD
-9.1
4
CP
-9.3
-8.9
CP
-9.1
5
ANGPTL3
-9.2
-8.9
PON3
-9.1
6
LEPR
-9.1
-8.9
VTN
-8.9
7
ADGRE5
-9
-8.9
ALB
-8.8
8
FGG
-8.9
-8.9
B2M
-8.8
9
AFM
-8.8
-8.8
BPI
-8.7
10
PLA2G2A
-8.8
-8.6
NAMPT
-8.7
11
APOF
-8.7
-8.6
SELE
-8.6
12
PDGFB
-8.7
-8.6
FGG
-8.5
13
PON3
-8.7
-8.6
HPX
-8.5
14
ALB
-8.6
-8.6
PON1
-8.5
15
BMP6
-8.6
-8.5
BMP6
-8.4
16
C9
-8.6
-8.5
FABP3
-8.4
17
IL1RL1
-8.6
-8.5
HABP2
-8.4
18
SELE
-8.6
-8.4
LBP
-8.4
19
VTN
-8.6
-8.4
MMP9
-8.4
20
BPI
-8.5
-8.4
OSM
-8.4
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Graphical abstract
The sequences of the 401 human blood plasma proteins were retrieved from the UniProt
database and RoseTTAFold protein structure prediction tool was employed to predict the
structures of proteins. Next, three metabolites, Δ9-THC, 11-OH-Δ9-THC and 8β,11-diOH-Δ9-
THC were downloaded from the PubChem database and energy minimization of these three
metabolites were done using Avogadro molecular graphic tool. Finally, the molecular docking
of these three substances was performed with 401 (3*401= 1203) using blind docker CB-
DOCK tool and, interactions analysis of top two complexes of each metabolite was carried out.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Fig. 1. (A-C) 2D structures of Δ9-THC, 11-OH-Δ9-THC and 8β,11-diOH-Δ9-THC respectively.
(D-F) 3D structures of Δ9-THC, 11-OH-Δ9-THC and 8β,11-diOH-Δ9-THC respectively. The
yellow spheres indicate the addition of new hydroxyl groups to the Δ9-THC.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Fig. 2. The heatmap of binding affinity (Vina score in kcal/mol) of Δ9-THC and its two
metabolites with 401 human blood plasma proteins. Binding affinities correspond to color bar.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Fig. 3. Complex structures and their 2D interactions. (A) APOA5:Δ9-THC complex. (B)
APOA5-Δ9-THC interactions. (C) APOD:Δ9-THC complex. (D) APOD:Δ9-THC interactions.
(APOA5: Apolipoprotein A5 & APOD: Apolipoprotein D). Orange-red circle shows the ligand
position.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Fig. 4. Complex structures and their 2D interactions. (A) ADGRE5:11-OH-Δ9-THC complex.
(B) ADGRE5:11-OH-Δ9-THC interactions. (C) CP:11-OH-Δ9-THC complex. (D) CP:11-OH-
Δ9-THC interactions. (ADGRE5: Adhesion G protein-coupled receptor E5 & CP:
Ceruloplasmin). Orange-red circle shows the ligand position.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Fig. 5. Complex structures and their 2D interactions. (A) APOA5:8,11-diOH-Δ9-THC
complex. (B) APOA5-8,11-diOH-Δ9-THC interactions. (C) ADGRE5:8,11-diOH-Δ9-THC
complex. (D) ADGRE5:8,11-diOH-Δ9-THC interactions. (APOA5: Apolipoprotein A5 &
ADGRE5: Adhesion G protein-coupled receptor E5). Orange-red circle shows the ligand
position.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
Fig. 6. Electrostatic potential surface (ESP) of four proteins. (A) Apolipoprotein A5 (APOA5).
(B) Apolipoprotein D (APOD). (C) Adhesion G protein-coupled receptor E5 (ADGRE5). (D)
Ceruloplasmin (CP). Ligand position is indicated and zoomed in black square box.
.CC-BY-NC-ND 4.0 International licensemade available under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is
The copyright holder for this preprintthis version posted April 14, 2022. ; https://doi.org/10.1101/2022.04.13.488197doi: bioRxiv preprint
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The three-dimensional molecular structure of human serum ceruloplasmin has been reinvestigated using X-ray synchrotron data collected at 100 K from a crystal frozen to liquid-nitrogen temperature. The resulting model, with an increase in resolution from 3.1 to 2.8 Å, gives an overall improvement of the molecular structure, in particular the side chains. In addition, it enables the clear definition of previously unidentified Ca²⁺-binding and Na⁺-binding sites. The Ca²⁺ cation is located in domain 1 in a configuration very similar to that found in the activated bovine factor Va. The Na⁺ sites appear to play a structural role in providing rigidity to the three protuberances on the top surface of the molecule. These features probably help to steer substrates towards the mononuclear copper sites prior to their oxidation and to restrict the size of the approaching substrate. The trinuclear copper centre appears to differ from the room-temperature structure in that a dioxygen moiety is bound in a similar way to that found in the endospore coat protein CotA from Bacillus subtilis.
Article
Full-text available
Neurodegenerative disorders can induce modifications of several proteins; one of which is ceruloplasmin (Cp), a ferroxidase enzyme found modified in the cerebrospinal fluid (CSF) of neurodegenerative diseases patients. Cp modifications are caused by the oxidation induced by the pathological environment and are usually associated with activity loss. Together with oxidation, deamidation of Cp was found in the CSF from Alzheimer’s and Parkinson’s disease patients. Protein deamidation is a process characterized by asparagine residues conversion in either aspartate or isoaspartate, depending on protein sequence/structure and cellular environment. Cp deamidation occurs at two Asparagine-Glycine-Arginine (NGR)-motifs which, once deamidated to isoAspartate-Glycine-Arginine (isoDGR), bind integrins, a family of receptors mediating cell adhesion. Therefore, on the one hand, Cp modifications lead to loss of enzymatic activity, while on the other hand, these alterations confer gain of function to Cp. In fact, deamidated Cp binds to integrins and triggers intracellular signaling on choroid plexus epithelial cells, changing cell functioning. Working in concert with the oxidative environment, Cp deamidation could reach different target cells in the brain, altering their physiology and causing detrimental effects, which might contribute to the pathological mechanism.
Article
Full-text available
Apolipoprotein A5 (apoA5) has been identified to play an important role in lipid metabolism, specifically in triglyceride (TG) and TG-rich lipoproteins (TRLs) metabolism. Numerous evidence has demonstrated for an association between apoA5 and the increased risk of obesity and metabolic syndrome, but the mechanism remains to be fully elucidated. Recently, several studies verified that apoA5 could significantly reduce plasma TG level by stimulating lipoprotein lipase (LPL) activity, and the intracellular role of apoA5 has also been proved since apoA5 is associated with cytoplasmic lipid droplets (LDs) and affects intrahepatic TG accumulation. Furthermore, since adipocytes provide the largest storage depot for TG and play a crucial role in the development of obesity, we could infer that apoA5 also acts as a novel regulator to modulate TG storage in adipocytes. In this review, we focus on the association of gene and protein of apoA5 with obesity and metabolic syndrome, and provide new insights into the physiological role of apoA5 in humans, giving a potential therapeutic target for obesity and associated disorders.
Article
Full-text available
Ceruloplasmin (CP) is the major copper transport protein in plasma, mainly produced by the liver. Glycosylphosphatidylinositol-linked CP (GPI-CP) is the predominant form expressed in astrocytes of the brain. A growing body of evidence has demonstrated that CP is an essential protein in body with multiple functions such as regulating the homeostasis of copper and iron ions, ferroxidase activity, oxidizing organic amines, and preventing the formation of free radicals. In addition, as an acute-phase protein, CP is induced during inflammation and infection. The fact that patients with genetic disorder aceruloplasminemia do not suffer from tissue copper deficiency, but rather from disruptions in iron metabolism shows the essential roles of CP on iron metabolism rather than copper. Furthermore, abnormal metabolism of metal ions and oxidative stress are found in other neurodegenerative diseases, such as Wilson's disease, Alzheimer's disease and Parkinson's disease. Brain iron accumulation and decreased activity of CP have been shown to be associated with neurodegeneration. We hypothesize that CP may play a protective role in neurodegenerative diseases. However, whether iron accumulation is a cause or a result of neurodegeneration remains unclear. Further research on molecular mechanisms are required before a consensus can be reached regarding a neuroprotective role for CP in neurodegeneration. This review article summarizes the main physiological functions of CP and the current knowledge on its role in neurodegenerative diseases.
Article
Full-text available
Background: Increasing cannabis consumption among adolescents, studies that link its early use with mental illnesses, and the political debate on cannabis legalization together call for an urgent need to study molecular underpinnings of adolescent brain vulnerability. The emerging role of epigenetic mechanisms in psychiatric diseases led us to hypothesize that epigenetic alterations could play a role in causes and subsequent development of the depressive/psychotic-like phenotype induced by adolescent, but not adult, Δ9-tetrahydrocannabinol (THC) exposure in female rats. Methods: We performed a time-course analysis of histone modifications, chromatin remodelling enzymes and gene expression in the prefrontal cortex of female rats after adolescent and adult THC exposure. We also administered a specific epigenetic drug (chaetocin) with THC to investigate its impact on THC-induced behavioural alterations. Results: Adolescent THC exposure induced alterations of selective histone modifications (mainly H3K9me3), impacting the expression of genes closely associated with synaptic plasticity. Changes in both histone modifications and gene expression were more widespread and intense after adolescent treatment, suggesting specific adolescent susceptibility. Adolescent THC exposure significantly increased Suv39H1 levels, which could account for the enhanced H3K9me3. Pharmacological blockade of H3K9me3 during adolescent THC treatment prevented THC-induced cognitive deficits, suggesting the relevant role played by H3K9me3 in THC-induced effects. Limitations: Only female rats were investigated, and the expression studies were limited to a specific subset of genes. Conclusion: Through a mechanism involving SUV39H1, THC modifies histone modifications and, thereby, expression of plasticity genes. This pathway appears to be relevant for the development of cognitive deficits.
Article
Full-text available
Patients with glioblastoma have one of the lowest overall survival rates among patients with cancer. Standard of care for patients with glioblastoma includes temozolomide and radiation therapy, yet 30% of patients do not respond to these treatments and nearly all glioblastoma tumors become resistant. Chlorpromazine is a United States Food and Drug Administration-approved phenothiazine widely used as a psychotropic in clinical practice. Recently, experimental evidence revealed the anti-proliferative activity of chlorpromazine against colon and brain tumors. Here, we used chemoresistant patient-derived glioma stem cells and chemoresistant human glioma cell lines to investigate the effects of chlorpromazine against chemoresistant glioma. Chlorpromazine selectively and significantly inhibited proliferation in chemoresistant glioma cells and glioma stem cells. Mechanistically, chlorpromazine inhibited cytochrome c oxidase (CcO, complex IV) activity from chemoresistant but not chemosensitive cells, without affecting other mitochondrial complexes. Notably, our previous studies revealed that the switch to chemoresistance in glioma cells is accompanied by a switch from the expression of CcO subunit 4 isoform 2 (COX4-2) to COX4-1. In this study, chlorpromazine induced cell cycle arrest selectively in glioma cells expressing COX4-1, and computer-simulated docking studies indicated that chlorpromazine binds more tightly to CcO expressing COX4-1 than to CcO expressing COX4-2. In orthotopic mouse brain tumor models, chlorpromazine treatment significantly increased the median overall survival of mice harboring chemoresistant tumors. These data indicate that chlorpromazine selectively inhibits the growth and proliferation of chemoresistant glioma cells expressing COX4-1. The feasibility of repositioning chlorpromazine for selectively treating chemoresistant glioma tumors should be further explored.
Article
CD97/ADGRE5, an adhesion G protein-coupled receptor (aGPCR), is highly expressed in several tumor cell types. CD97 has been shown to modulate tumorigenesis in part by promoting HUVEC migration, invasion and angiogenesis through the interaction with integrin α5β1 via its ectodomain RGD motif. In this study, we show that CD97 could induce angiogenesis via an alternative RGD-independent mechanism. Overexpression of CD97 with the wild-type or mutant RGD motif in HT1080 cells led to up-regulated MMP-9 and induced angiogenesis as revealed by the in vitro endothelial cell tube formation assay and in ovo chick chorioallantoic membrane assay. By contrast, expression of EMR2/ADGRE2, the CD97-homologous aGPCR that contains a corresponding SGD sequence, fails to induce angiogenesis due to lower MMP-9 expression. Interestingly, a single change of the SGD to RGD sequence allowed EMR2 to up-regulate MMP-9 expression, leading to enhanced angiogenesis. MMP-9 was shown to promote the proliferation, migration, and invasion of HUVEC partly by modulating the levels of VEGF, PIGF, and bFGF. Finally, we showed that the MMP-9 expression was in turn modulated by N-cadherin that was up-regulated by CD97 and EMR2/RGD. Our results indicate that two homologous aGPCRs, CD97 and EMR2, modulate angiogenesis and HUVEC proliferation, migration, and invasion through N-cadherin-regulated MMP-9 expression by RGD-independent and -dependent mechanisms, respectively.
Article
As the number of elucidated protein structures is rapidly increasing, the growing data call for methods to efficiently exploit the structural information for biological and pharmaceutical purposes. Given the three-dimensional (3D) structure of a protein and a ligand, predicting their binding sites and affinity are a key task for computer-aided drug discovery. To address this task, a variety of docking tools have been developed. Most of them focus on docking in the preset binding sites given by users. To automatically predict binding modes without information about binding sites, we developed a user-friendly blind docking web server, named CB-Dock, which predicts binding sites of a given protein and calculates the centers and sizes with a novel curvature-based cavity detection approach, and performs docking with a popular docking program, Autodock Vina. This method was carefully optimized and achieved ~70% success rate for the top-ranking poses whose root mean square deviation (RMSD) were within 2 Å from the X-ray pose, which outperformed the state-of-the-art blind docking tools in our benchmark tests. CB-Dock offers an interactive 3D visualization of results, and is freely available at http://cao.labshare.cn/cb-dock/.
Article
There is evidence that Cannabis whose active ingredient is tetrahydrocannabinol (THC) is the most commonly abused neuroactive substance, among young adults. This work investigated the effects of Cannabis sativa on the cytoarchitecture of some key organs and the blood chemistry of rat models. Twenty-one (21) male Sprague Dawley rats were fed different percentage of cannabis chow (0%, 5% and 10%) for a period of seven (7) weeks. Rats were subjected to intermittent cognitive function test and sacrificed after the seventh week, collecting the blood, brain and other important tissues for analysis which include; brain total protein and nitric oxide concentration, blood chemistry and histopathology. Results revealed a dose-dependent decline in the cognitive function, statistically significant decrease in the brain total protein and nitric oxide. Histopathology revealed significant hypertrophy in the heart, hypercellularity in neuronal cells, prominent sinusoids cytoarchitecture of the hepatocytes and vascular congestion in the seminiferous tubules of testes. There was a statistically significant difference in the plasma ALP, ALT, AST level between controls and the cannabis test groups. Cannabis use caused cellular damage through mediation of imbalance and altered cytoarchitecture which may affects the overall health of dependent user.