1
|
Liu SJ, Fu JJ, Liao ZY, Liu YX, He J, He LY, Bai J, Yang JY, Niu SQ, Guo JL. Z-ligustilide alleviates atherosclerosis by reconstructing gut microbiota and sustaining gut barrier integrity through activation of cannabinoid receptor 2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156117. [PMID: 39426255 DOI: 10.1016/j.phymed.2024.156117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/16/2024] [Accepted: 07/01/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Z-Ligustilide (ZL) is an essential phthalide found in Ligusticum chuanxiong Hort, a commonly used traditional Chinese medicine for treating atherosclerosis (AS) clinically. ZL has been shown to be effective in treating AS. However, the underlying mechanism of ZL against AS and its potential targets remain elusive. PURPOSE The purpose of this research was to assess the influence of ZL on AS and explore the role of the gut microbiome in mediating this effect. METHODS A well-established AS mouse model, apolipoprotein E deficient (ApoE-/-) mice was used to examine the effects of ZL on AS, inflammation, and the intestinal barrier. To analyze the changes in gut microbial community, we employed the 16S rRNA gene sequencing. Antibiotic cocktail and fecal microbiota transplantation (FMT) were employed to clarify the contribution of the gut microbiota to the anti-AS effects of ZL. The mechanism through which ZL provided protective effects on AS and the intestinal barrier was explored by untargeted metabolomics, as well as by validating the involvement of cannabinoid receptor 2 (CB2R) in mice and Caco-2 cells. RESULTS Oral administration of ZL inhibited the development of atherosclerotic lesions, improved plaque stability, inhibited the increase in serum and atherosclerotic inflammation, and improved intestinal barrier function. Fecal bacteria from ZL-treated mice induced similar beneficial effects on AS and the intestinal barrier. We used 16S RNA gene sequencing to reveal a significant increase in Rikenella abundance in both ZL-treated mice and ZL-FMT mice, which was associated with the beneficial effects of ZL. Further function prediction analysis of the gut microbiota and CB2R antagonist intervention experiment in mice and Caco-2 cells showed that the activation of CB2R resulted in the enhancement of the intestinal barrier by ZL. Furthermore, the analysis of metabolomic profiling revealed the enrichment of capsaicin upon ZL treatment, which induced the activation of CB2R in human colon epithelial cells. CONCLUSION Our study is the first to demonstrate that oral treatment with ZL has the potential to alleviate AS by reducing inflammation levels and enhancing the intestinal barrier function. This mechanism relies on the gut microbiota in a CB2R-dependent manner, suggesting promising strategies and ideas for managing AS. This study provides insights into a novel mechanism for treating AS with ZL.
Collapse
Affiliation(s)
- Si-Jing Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Jiao-Jiao Fu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Zhen-Yue Liao
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yi-Xin Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Jing He
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Li-Ying He
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Jing Bai
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Jing-Yan Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China.
| | - Shu-Qi Niu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China.
| | - Jin-Lin Guo
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China; Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China.
| |
Collapse
|
2
|
Perez PA, Wiley MB, Makriyannis A, DiPatrizio NV. Cannabinoids Block Fat-induced Incretin Release via CB 1-dependent and CB 1-independent Pathways in Intestinal Epithelium. GASTRO HEP ADVANCES 2024; 3:931-941. [PMID: 39318720 PMCID: PMC11419882 DOI: 10.1016/j.gastha.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/11/2024] [Indexed: 09/26/2024]
Abstract
Background and Aims Glucose homeostasis is regulated by a dynamic interplay between hormones along the gastro-insular axis. For example, enteroendocrine L- and K- cells that line the intestine produce the incretins glucagon-like peptide-1 (GLP1) and glucose-dependent insulinotropic polypeptide (GIP), respectively, which are secreted following a meal. Broadly, incretin signaling enhances insulin release from the endocrine pancreas and participates in the control of food intake, and therapeutics that mimic their activity have recently been developed for the treatment of type-2 diabetes and obesity. Notably, genes for cannabinoid subtype-1 receptor (CB1R) are expressed in these cell subpopulations; however, roles for CB1Rs in controlling fat-induced incretin release are unclear. To address this gap in our understanding, we tested the hypothesis that intestinal epithelial CB1Rs control fat-induced incretin secretion. Methods We treated mice with conditional deletion of CB1Rs in the intestinal epithelium (IntCB1-/-) or controls (IntCB1+/+) with oil gavage to stimulate incretin release in the presence of the cannabinoid receptor agonists, WIN55,212-2 or Δ9 tetrahydrocannabinol (THC), and the peripherally-restricted CB1R antagonist AM6545. Circulating incretin levels were measured in plasma. Results Oral gavage of corn oil increased levels of bioactive GLP1 and GIP in IntCB1+/+ mouse plasma. Pretreatment with the WIN55,212-2 or THC blocked this response, which was largely reversed by coadministration with AM6545. WIN55,212-2 failed to inhibit fat-induced GIP release, but not GLP1, in IntCB1-/- mice. In contrast, THC inhibited the secretion of incretins irrespective of CB1R expression in intestinal epithelial cells. Conclusion These results indicate that cannabinoid receptor agonists can differentially inhibit incretin release via mechanisms that include intestinal epithelial CB1R-dependent and CB1R-independent mechanisms.
Collapse
Affiliation(s)
- Pedro Antonio Perez
- Center for Cannabinoid Research (UCRCCR), School of Medicine, University of California, Riverside, Riverside, California
- Department of Neuroscience and The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California
| | - Mark Benjamin Wiley
- Center for Cannabinoid Research (UCRCCR), School of Medicine, University of California, Riverside, Riverside, California
| | | | - Nicholas Vincent DiPatrizio
- Center for Cannabinoid Research (UCRCCR), School of Medicine, University of California, Riverside, Riverside, California
| |
Collapse
|
3
|
Crowley K, Kiraga Ł, Miszczuk E, Skiba S, Banach J, Latek U, Mendel M, Chłopecka M. Effects of Cannabinoids on Intestinal Motility, Barrier Permeability, and Therapeutic Potential in Gastrointestinal Diseases. Int J Mol Sci 2024; 25:6682. [PMID: 38928387 PMCID: PMC11203611 DOI: 10.3390/ijms25126682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Cannabinoids and their receptors play a significant role in the regulation of gastrointestinal (GIT) peristalsis and intestinal barrier permeability. This review critically evaluates current knowledge about the mechanisms of action and biological effects of endocannabinoids and phytocannabinoids on GIT functions and the potential therapeutic applications of these compounds. The results of ex vivo and in vivo preclinical data indicate that cannabinoids can both inhibit and stimulate gut peristalsis, depending on various factors. Endocannabinoids affect peristalsis in a cannabinoid (CB) receptor-specific manner; however, there is also an important interaction between them and the transient receptor potential cation channel subfamily V member 1 (TRPV1) system. Phytocannabinoids such as Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) impact gut motility mainly through the CB1 receptor. They were also found to improve intestinal barrier integrity, mainly through CB1 receptor stimulation but also via protein kinase A (PKA), mitogen-associated protein kinase (MAPK), and adenylyl cyclase signaling pathways, as well as by influencing the expression of tight junction (TJ) proteins. The anti-inflammatory effects of cannabinoids in GIT disorders are postulated to occur by the lowering of inflammatory factors such as myeloperoxidase (MPO) activity and regulation of cytokine levels. In conclusion, there is a prospect of utilizing cannabinoids as components of therapy for GIT disorders.
Collapse
Affiliation(s)
- Kijan Crowley
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.C.); (E.M.); (U.L.); (M.M.)
| | - Łukasz Kiraga
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.C.); (E.M.); (U.L.); (M.M.)
| | - Edyta Miszczuk
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.C.); (E.M.); (U.L.); (M.M.)
| | - Sergiusz Skiba
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.C.); (E.M.); (U.L.); (M.M.)
| | - Joanna Banach
- Department of Research and Processing Seed, Institute of Natural Fibers and Medicinal Plants—National Research Institute, Wojska Polskiego 71b, 60-630 Poznan, Poland;
| | - Urszula Latek
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.C.); (E.M.); (U.L.); (M.M.)
| | - Marta Mendel
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.C.); (E.M.); (U.L.); (M.M.)
| | - Magdalena Chłopecka
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (K.C.); (E.M.); (U.L.); (M.M.)
| |
Collapse
|
4
|
Wood CP, Alvarez C, DiPatrizio NV. Cholinergic Neurotransmission Controls Orexigenic Endocannabinoid Signaling in the Gut in Diet-Induced Obesity. J Neurosci 2024; 44:e0813232024. [PMID: 38594069 PMCID: PMC11097264 DOI: 10.1523/jneurosci.0813-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 03/12/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
The brain bidirectionally communicates with the gut to control food intake and energy balance, which becomes dysregulated in obesity. For example, endocannabinoid (eCB) signaling in the small-intestinal (SI) epithelium is upregulated in diet-induced obese (DIO) mice and promotes overeating by a mechanism that includes inhibiting gut-brain satiation signaling. Upstream neural and molecular mechanism(s) involved in overproduction of orexigenic gut eCBs in DIO, however, are unknown. We tested the hypothesis that overactive parasympathetic signaling at the muscarinic acetylcholine receptors (mAChRs) in the SI increases biosynthesis of the eCB, 2-arachidonoyl-sn-glycerol (2-AG), which drives hyperphagia via local CB1Rs in DIO. Male mice were maintained on a high-fat/high-sucrose Western-style diet for 60 d, then administered several mAChR antagonists 30 min prior to tissue harvest or a food intake test. Levels of 2-AG and the activity of its metabolic enzymes in the SI were quantitated. DIO mice, when compared to those fed a low-fat/no-sucrose diet, displayed increased expression of cFos protein in the dorsal motor nucleus of the vagus, which suggests an increased activity of efferent cholinergic neurotransmission. These mice exhibited elevated levels of 2-AG biosynthesis in the SI, that was reduced to control levels by mAChR antagonists. Moreover, the peripherally restricted mAChR antagonist, methylhomatropine bromide, and the peripherally restricted CB1R antagonist, AM6545, reduced food intake in DIO mice for up to 24 h but had no effect in mice conditionally deficient in SI CB1Rs. These results suggest that hyperactivity at mAChRs in the periphery increases formation of 2-AG in the SI and activates local CB1Rs, which drives hyperphagia in DIO.
Collapse
Affiliation(s)
- Courtney P Wood
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California 92521
- University of California Riverside Center for Cannabinoid Research, Riverside, California 92521
| | - Camila Alvarez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California 92521
- University of California Riverside Center for Cannabinoid Research, Riverside, California 92521
| | - Nicholas V DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California 92521
- University of California Riverside Center for Cannabinoid Research, Riverside, California 92521
| |
Collapse
|
5
|
Wiley MB, Bauer J, Alvarez V, Mehrotra K, Cheng W, Kolics Z, Giarrizzo M, Ingle K, Bialkowska AB, Jung B. Activin A signaling stimulates neutrophil activation and macrophage migration in pancreatitis. Sci Rep 2024; 14:9382. [PMID: 38654064 PMCID: PMC11039671 DOI: 10.1038/s41598-024-60065-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024] Open
Abstract
Acute Pancreatitis (AP) is associated with high mortality and current treatment options are limited to supportive care. We found that blockade of activin A (activin) in mice improves outcomes in two murine models of AP. To test the hypothesis that activin is produced early in response to pancreatitis and is maintained throughout disease progression to stimulate immune cells, we first performed digital spatial profiling (DSP) of human chronic pancreatitis (CP) patient tissue. Then, transwell migration assays using RAW264.7 mouse macrophages and qPCR analysis of "neutrophil-like" HL-60 cells were used for functional correlation. Immunofluorescence and western blots on cerulein-induced pancreatitis samples from pancreatic acinar cell-specific Kras knock-in (Ptf1aCreER™; LSL-KrasG12D) and functional WT Ptf1aCreER™ mouse lines mimicking AP and CP to allow for in vivo confirmation. Our data suggest activin promotes neutrophil and macrophage activation both in situ and in vitro, while pancreatic activin production is increased as early as 1 h in response to pancreatitis and is maintained throughout CP in vivo. Taken together, activin is produced early in response to pancreatitis and is maintained throughout disease progression to promote neutrophil and macrophage activation.
Collapse
Affiliation(s)
- Mark B Wiley
- Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Jessica Bauer
- Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Valentina Alvarez
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Kunaal Mehrotra
- Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Wenxuan Cheng
- Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Zoe Kolics
- Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Michael Giarrizzo
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Komala Ingle
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Barbara Jung
- Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA.
| |
Collapse
|
6
|
Levine AA, Liktor-Busa E, Balasubramanian S, Palomino SM, Burtman AM, Couture SA, Lipinski AA, Langlais PR, Largent-Milnes TM. Depletion of Endothelial-Derived 2-AG Reduces Blood-Endothelial Barrier Integrity via Alteration of VE-Cadherin and the Phospho-Proteome. Int J Mol Sci 2023; 25:531. [PMID: 38203706 PMCID: PMC10778805 DOI: 10.3390/ijms25010531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Mounting evidence supports the role of the endocannabinoid system in neurophysiology, including blood-brain barrier (BBB) function. Recent work has demonstrated that activation of endocannabinoid receptors can mitigate insults to the BBB during neurological disorders like traumatic brain injury, cortical spreading depression, and stroke. As alterations to the BBB are associated with worsening clinical outcomes in these conditions, studies herein sought to examine the impact of endocannabinoid depletion on BBB integrity. Barrier integrity was investigated in vitro via bEnd.3 cell monolayers to assess endocannabinoid synthesis, barrier function, calcium influx, junctional protein expression, and proteome-wide changes. Inhibition of 2-AG synthesis using DAGLα inhibition and siRNA inhibition of DAGLα led to loss of barrier integrity via altered expression of VE-cadherin, which could be partially rescued by exogenous application of 2-AG. Moreover, the deleterious effects of DAGLα inhibition on BBB integrity showed both calcium and PKC (protein kinase C)-dependency. These data indicate that disruption of 2-AG homeostasis in brain endothelial cells, in the absence of insult, is sufficient to disrupt BBB integrity thus supporting the role of the endocannabinoid system in neurovascular disorders.
Collapse
Affiliation(s)
- Aidan A. Levine
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Erika Liktor-Busa
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Shreya Balasubramanian
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Seph M. Palomino
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Anya M. Burtman
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Sarah A. Couture
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Austin A. Lipinski
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (P.R.L.)
| | - Paul R. Langlais
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (P.R.L.)
| | - Tally M. Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| |
Collapse
|
7
|
Wood CP, Avalos B, Alvarez C, DiPatrizio NV. A Sexually Dimorphic Role for Intestinal Cannabinoid Receptor Subtype-1 in the Behavioral Expression of Anxiety. Cannabis Cannabinoid Res 2023; 8:1045-1059. [PMID: 37862126 PMCID: PMC10771877 DOI: 10.1089/can.2023.0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Abstract
Background: Increasing evidence suggests that the endocannabinoid system (ECS) in the brain controls anxiety and may be a therapeutic target for the treatment of anxiety disorders. For example, both pharmacological and genetic disruption of cannabinoid receptor subtype-1 (CB1R) signaling in the central nervous system is associated with increased anxiety-like behaviors in rodents, while activating the system is anxiolytic. Sex is also a critical factor that controls the behavioral expression of anxiety; however, roles for the ECS in the gut in these processes and possible differences between sexes are largely unknown. Objective: In this study, we aimed to determine if CB1Rs in the intestinal epithelium exert control over anxiety-like behaviors in a sex-dependent manner. Methods: We subjected male and female mice with conditional deletion of CB1Rs in the intestinal epithelium (intCB1-/-) and controls (intCB1+/+) to the elevated plus maze (EPM), light/dark box, and open field test. Corticosterone (CORT) levels in plasma were measured at baseline and immediately after EPM exposure. Results: When compared with intCB1+/+ male mice, intCB1-/- male mice exhibited reduced levels of anxiety-like behaviors in the EPM and light/dark box. In contrast to male mice, no differences were found between female intCB1+/+ and intCB1-/- mice. Circulating CORT was higher in female versus male mice for both genotype groups at baseline and after EPM exposure; however, there was no effect of genotype on CORT levels. Conclusions: Collectively, these results indicate that genetic deletion of CB1Rs in the intestinal epithelium is associated with an anxiolytic phenotype in a sex-dependent manner.
Collapse
Affiliation(s)
- Courtney P. Wood
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Bryant Avalos
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Camila Alvarez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Nicholas V. DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
- University of California Riverside Center for Cannabinoid Research, Riverside, California, USA
| |
Collapse
|
8
|
Mboumba Bouassa RS, Comeau E, Alexandrova Y, Pagliuzza A, Yero A, Samarani S, Needham J, Singer J, Lee T, Bobeuf F, Vertzagias C, Sebastiani G, Margolese S, Mandarino E, Klein MB, Lebouché B, Routy JP, Chomont N, Costiniuk CT, Jenabian MA. Effects of Oral Cannabinoids on Systemic Inflammation and Viral Reservoir Markers in People with HIV on Antiretroviral Therapy: Results of the CTN PT028 Pilot Clinical Trial. Cells 2023; 12:1811. [PMID: 37508476 PMCID: PMC10378564 DOI: 10.3390/cells12141811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/23/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Chronic HIV infection is characterized by persistent inflammation despite antiretroviral therapy (ART). Cannabinoids may help reduce systemic inflammation in people with HIV (PWH). To assess the effects of oral cannabinoids during HIV, ten PWH on ART were randomized (n = 5/group) to increasing doses of oral Δ9-tetrahydrocannabinol (THC): cannabidiol (CBD) combination (2.5:2.5-15:15 mg/day) capsules or CBD-only (200-800 mg/day) capsules for 12 weeks. Blood specimens were collected prospectively 7-21 days prior to treatment initiation and at weeks 0 to 14. Plasma cytokine levels were determined via Luminex and ELISA. Immune cell subsets were characterized by flow cytometry. HIV DNA/RNA were measured in circulating CD4 T-cells and sperm by ultra-sensitive qPCR. Results from both arms were combined for statistical analysis. Plasma levels of IFN-γ, IL-1β, sTNFRII, and REG-3α were significantly reduced at the end of treatment (p ˂ 0.05). A significant decrease in frequencies of PD1+ memory CD4 T-cells, CD73+ regulatory CD4 T-cells, and M-DC8+ intermediate monocytes was also observed (p ˂ 0.05), along with a transient decrease in CD28-CD57+ senescent CD4 and CD8 T-cells. Ki-67+ CD4 T-cells, CCR2+ non-classical monocytes, and myeloid dendritic cells increased over time (p ˂ 0.05). There were no significant changes in other inflammatory markers or HIV DNA/RNA levels. These findings can guide future large clinical trials investigating cannabinoid anti-inflammatory properties.
Collapse
Affiliation(s)
- Ralph-Sydney Mboumba Bouassa
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Eve Comeau
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
| | - Yulia Alexandrova
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Amélie Pagliuzza
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Alexis Yero
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
| | - Suzanne Samarani
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Judy Needham
- CIHR Canadian HIV Trials Network, Vancouver, BC V6Z 1Y6, Canada
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Joel Singer
- CIHR Canadian HIV Trials Network, Vancouver, BC V6Z 1Y6, Canada
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Terry Lee
- CIHR Canadian HIV Trials Network, Vancouver, BC V6Z 1Y6, Canada
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Florian Bobeuf
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Claude Vertzagias
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Giada Sebastiani
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Shari Margolese
- CIHR Canadian HIV Trials Network, Vancouver, BC V6Z 1Y6, Canada
| | | | - Marina B Klein
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Bertrand Lebouché
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Family Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
- Centre for Outcomes Research & Evaluation, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Jean-Pierre Routy
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Nicolas Chomont
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Cecilia T Costiniuk
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
9
|
Wiley MB, Bauer J, Mehrotra K, Zessner-Spitzenberg J, Kolics Z, Cheng W, Castellanos K, Nash MG, Gui X, Kone L, Maker AV, Qiao G, Reddi D, Church DN, Kerr RS, Kerr DJ, Grippo PJ, Jung B. Non-Canonical Activin A Signaling Stimulates Context-Dependent and Cellular-Specific Outcomes in CRC to Promote Tumor Cell Migration and Immune Tolerance. Cancers (Basel) 2023; 15:3003. [PMID: 37296966 PMCID: PMC10252122 DOI: 10.3390/cancers15113003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
We have shown that activin A (activin), a TGF-β superfamily member, has pro-metastatic effects in colorectal cancer (CRC). In lung cancer, activin activates pro-metastatic pathways to enhance tumor cell survival and migration while augmenting CD4+ to CD8+ communications to promote cytotoxicity. Here, we hypothesized that activin exerts cell-specific effects in the tumor microenvironment (TME) of CRC to promote anti-tumoral activity of immune cells and the pro-metastatic behavior of tumor cells in a cell-specific and context-dependent manner. We generated an Smad4 epithelial cell specific knockout (Smad4-/-) which was crossed with TS4-Cre mice to identify SMAD-specific changes in CRC. We also performed IHC and digital spatial profiling (DSP) of tissue microarrays (TMAs) obtained from 1055 stage II and III CRC patients in the QUASAR 2 clinical trial. We transfected the CRC cells to reduce their activin production and injected them into mice with intermittent tumor measurements to determine how cancer-derived activin alters tumor growth in vivo. In vivo, Smad4-/- mice displayed elevated colonic activin and pAKT expression and increased mortality. IHC analysis of the TMA samples revealed increased activin was required for TGF-β-associated improved outcomes in CRC. DSP analysis identified that activin co-localization in the stroma was coupled with increases in T-cell exhaustion markers, activation markers of antigen presenting cells (APCs), and effectors of the PI3K/AKT pathway. Activin-stimulated PI3K-dependent CRC transwell migration, and the in vivo loss of activin lead to smaller CRC tumors. Taken together, activin is a targetable, highly context-dependent molecule with effects on CRC growth, migration, and TME immune plasticity.
Collapse
Affiliation(s)
- Mark B. Wiley
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Jessica Bauer
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Kunaal Mehrotra
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Jasmin Zessner-Spitzenberg
- Clinical Department for Gastroenterology and Hepatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Zoe Kolics
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Wenxuan Cheng
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Karla Castellanos
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Michael G. Nash
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Xianyong Gui
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Lyonell Kone
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Ajay V. Maker
- Department of Surgery, University of California-San Francisco, San Francisco, CA 94115, USA
| | - Guilin Qiao
- Department of Surgery, University of California-San Francisco, San Francisco, CA 94115, USA
| | - Deepti Reddi
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - David N. Church
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 4BH, UK
- NIHR Oxford Comprehensive Biomedical Research Center, Oxford University Hospitals NHS Foundation Trust, University of Oxford, Oxford OX1 4BH, UK
| | - Rachel S. Kerr
- Department of Oncology, University of Oxford, Oxford OX1 4BH, UK
| | - David J. Kerr
- Radcliffe Department of Medicine, University of Oxford, Oxford OX1 4BH, UK
| | - Paul J. Grippo
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Barbara Jung
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| |
Collapse
|
10
|
Kozlova EV, Chinthirla BD, Bishay AE, Pérez PA, Denys ME, Krum JM, DiPatrizio NV, Currás-Collazo MC. Glucoregulatory disruption in male mice offspring induced by maternal transfer of endocrine disrupting brominated flame retardants in DE-71. Front Endocrinol (Lausanne) 2023; 14:1049708. [PMID: 37008952 PMCID: PMC10063979 DOI: 10.3389/fendo.2023.1049708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/23/2023] [Indexed: 03/19/2023] Open
Abstract
Introduction Polybrominated diphenyl ethers (PBDEs) are commercially used flame retardants that bioaccumulate in human tissues, including breast milk. PBDEs produce endocrine and metabolic disruption in experimental animals and have been associated with diabetes and metabolic syndrome (MetS) in humans, however, their sex-specific diabetogenic effects are not completely understood. Our past works show glucolipid dysregulation resulting from perinatal exposure to the commercial penta-mixture of PBDEs, DE-71, in C57BL/6 female mice. Methods As a comparison, in the current study, the effects of DE-71 on glucose homeostasis in male offspring was examined. C57BL/6N dams were exposed to DE-71 at 0.1 mg/kg/d (L-DE-71), 0.4 mg/kg/d (H-DE-71), or received corn oil vehicle (VEH/CON) for a total of 10 wks, including gestation and lactation and their male offspring were examined in adulthood. Results Compared to VEH/CON, DE-71 exposure produced hypoglycemia after a 11 h fast (H-DE-71). An increased fast duration from 9 to 11 h resulted in lower blood glucose in both DE-71 exposure groups. In vivo glucose challenge showed marked glucose intolerance (H-DE-71) and incomplete clearance (L- and H-DE-71). Moreover, L-DE-71-exposed mice showed altered glucose responses to exogenous insulin, including incomplete glucose clearance and/or utilization. In addition, L-DE-71 produced elevated levels of plasma glucagon and the incretin, active glucagon-like peptide-1 (7-36) amide (GLP-1) but no changes were detected in insulin. These alterations, which represent criteria used clinically to diagnose diabetes in humans, were accompanied with reduced hepatic glutamate dehydrogenase enzymatic activity, elevated adrenal epinephrine and decreased thermogenic brown adipose tissue (BAT) mass, indicating involvement of several organ system targets of PBDEs. Liver levels of several endocannabinoid species were not altered. Discussion Our findings demonstrate that chronic, low-level exposure to PBDEs in dams can dysregulate glucose homeostasis and glucoregulatory hormones in their male offspring. Previous findings using female siblings show altered glucose homeostasis that aligned with a contrasting diabetogenic phenotype, while their mothers displayed more subtle glucoregulatory alterations, suggesting that developing organisms are more susceptible to DE-71. We summarize the results of the current work, generated in males, considering previous findings in females. Collectively, these findings offer a comprehensive account of differential effects of environmentally relevant PBDEs on glucose homeostasis and glucoregulatory endocrine dysregulation of developmentally exposed male and female mice.
Collapse
Affiliation(s)
- Elena V. Kozlova
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, United States
- Neuroscience Graduate Program, University of California Riverside, Riverside, CA, United States
| | - Bhuvaneswari D. Chinthirla
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, United States
| | - Anthony E. Bishay
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, United States
| | - Pedro A. Pérez
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Maximillian E. Denys
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, United States
| | - Julia M. Krum
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, United States
| | - Nicholas V. DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Margarita C. Currás-Collazo
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, United States
| |
Collapse
|