1
|
Li W, Shen Q, Tong T, Tian H, Lian X, Wang H, Yang K, Dai Z, Li Y, Chen X, Wang Q, Yang D, Wang F, Hao F, Wang L. Sequential simulation of regeneration-specific microenvironments using scaffolds loaded with nanoplatelet vesicles enhances bone regeneration. Bioact Mater 2025; 50:475-493. [PMID: 40342486 PMCID: PMC12059598 DOI: 10.1016/j.bioactmat.2025.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 03/27/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
Bone regeneration is a complex and coordinated physiological process, and the different stages of this process have corresponding microenvironments to support cell development and physiological activities. However, biological scaffolds that provide different three-dimensional environments during different stages of bone regeneration are lacking. In this study, we report a novel composite scaffold (NPE@DCBM) inspired by the stages of bone regeneration; this scaffold was composed of a fibrin hydrogel loaded with nanoplatelet vesicles (NPVs), designated as NPE, and decellularized cancellous bone matrix (DCBM) microparticles. Initially, the NPE rapidly established a temporary microenvironment conducive to cell migration and angiogenesis. Subsequently, the DCBM simulated the molecular structure of bone and promoted new bone formation. In vitro, the NPVs regulated lipid metabolism in bone marrow mesenchymal stem cells (BMSCs), reprogramed the fate of BMSCs by activating the PI3K/AKT and MAPK/ERK positive feedback pathways, and increased BMSC functions, including proliferation, migration and proangiogenic potential. In vivo, NPV@DCBM accelerated bone tissue regeneration and repair. Initially, the NPE rapidly induced angiogenesis between DCBM microparticles, and subsequently, BMSCs differentiated into osteoblasts with DCBM microparticles at their core. In summary, the design of this composite scaffold that sequentially mimics different bone regeneration microenvironments may provide a promising strategy for bone regeneration, with clinical translational potential.
Collapse
Affiliation(s)
- Wenshuai Li
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- Hangzhou OrigO Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Qichen Shen
- Hangzhou OrigO Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Tong Tong
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Hongsen Tian
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Xiaowei Lian
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Haoli Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Ke Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Zhanqiu Dai
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Yijun Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xianhua Chen
- Zhejiang Institute of Medical Device Testing, Hangzhou, Zhejiang, 310016, China
| | - Qingqing Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315000, China
| | - Dan Yang
- Zhejiang DecellMatrix Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Feng Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Feng Hao
- Zhejiang DecellMatrix Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Linfeng Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| |
Collapse
|
2
|
Menezes J, Jakobsson JE, Bersellini Farinotti A, Krock E, Hunt MA, Simon N, Venckute Larsson S, Tanum L, Kultima K, Kosek E, Svensson CI. Comparative Analysis of Lysophosphatidic Acid Levels in Fibromyalgia and Other Painful Conditions in Female Patients. Eur J Pain 2025; 29:e70022. [PMID: 40269628 PMCID: PMC12018871 DOI: 10.1002/ejp.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/09/2025] [Accepted: 03/26/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Previous work found a decrease in lysophosphatidylcholines (LPCs) in fibromyalgia (FM) serum, prompting the hypothesis that this decrease could be due to increased conversion of LPC to lysophosphatidic acid (LPA) through autotaxin (ATX). LPA has pronociceptive functions, and increased LPA levels could modulate FM pain. METHODS This study quantified LPA levels in serum and lumbar cerebrospinal fluid (CSF) and serum ATX levels in FM patients, comparing with healthy controls (HCs), osteoarthritis (OA), degenerative disc disease (DDD) and lumbar disc herniation (LDH) patients. RESULTS We found increased serum LPA levels in FM and OA patients, with no changes in FM lumbar CSF. Unexpectedly, a positive correlation between serum LPA and conditioned pain modulation was observed in FM patients, while LPA levels were correlated with pain intensity and Knee Injury and Osteoarthritis Outcome Scores in OA. Serum ATX levels in FM patients were comparable to those in HC but correlated significantly with FM LPA levels (in one cohort), as well as with pain duration and the maximal weekly pain intensity. CONCLUSIONS This study suggests that increased LPA levels play distinct roles in FM and OA patients. In FM, LPA levels were linked to less impaired inhibitory pain pathways, while LPA levels in OA correlated with pain intensity and knee-related impairment. ATX levels in FM serum are associated with pain intensity and duration. These findings underscore the complex role of LPA and ATX in FM pathophysiology. Future studies are essential to clarify LPA's specific roles and to develop therapies. SIGNIFICANCE STATEMENT This study provides novel insights into the role of LPA in FM and other chronic pain conditions. Although ATX levels were unchanged in FM, a positive correlation between serum ATX and LPA supports the role of ATX in LPA conversion. These findings suggest complex lipid dysregulation in FM, with LPA potentially modulating pain pathways. Further research is needed to clarify LPA's role and its potential as a biomarker or therapeutic target.
Collapse
Affiliation(s)
- Joana Menezes
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Jenny E. Jakobsson
- Department of Medical SciencesUppsala UniversityUppsalaSweden
- Clinical Pain Research, Department of Surgical SciencesUppsala UniversityUppsalaSweden
| | - Alex Bersellini Farinotti
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Emerson Krock
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
- Faculty of Dental Medicine and Oral Health SciencesAlan Edwards Centre for Research on Pain, McGill UniversityMontrealCanada
| | - Matthew A. Hunt
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Nils Simon
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Sigita Venckute Larsson
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Lars Tanum
- Department of R&D in Mental HealthAkershus University HospitalLørenskogNorway
| | - Kim Kultima
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Eva Kosek
- Clinical Pain Research, Department of Surgical SciencesUppsala UniversityUppsalaSweden
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Camilla I. Svensson
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| |
Collapse
|
3
|
Lindgren ES, Yan R, Kuo YM, Gao Q, de Souza Goncalves L, Chen FY, Chan MF, Verkman AS, Cil O, Pasricha ND. Lysophosphatidic acid receptor 3 (LPAR3) regulates ocular surface chloride transport via calcium signaling. Exp Eye Res 2025; 255:110346. [PMID: 40112945 DOI: 10.1016/j.exer.2025.110346] [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: 10/28/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Dry eye is a multifactorial disease associated with impaired tear film homeostasis, damaging the ocular surface epithelium. Lysophosphatidic acid receptors (LPARs) are G-protein coupled receptors involved in Ca2+ and cAMP signaling via PLC and adenylyl cyclase activation. LPAR activation is involved in cell proliferation and wound healing in human corneal epithelial cells (HCECs) and in neuropathic pain. This study investigates the expression and functions of LPARs in ocular surface epithelial cells. Functional measurements of ocular surface potential difference (OSPD) were done in mice with topically applied, selective LPAR modulators. LPAR3 immunostaining was performed in mouse and human cornea and conjunctiva, and mouse lacrimal gland. LPAR-induced Ca2+ signaling was studied in primary and immortalized HCECs. The general LPAR agonist, linoleoyl LPA, and the LPAR3 selective agonist, 2S-OMPT, stimulated ocular surface Cl- secretion via Ca2+-activated Cl- channels (CaCCs). LPAR3 was expressed in the corneal and conjunctival epithelia of mice and humans, as well as in mouse lacrimal gland. Activation of LPAR and LPAR3 in HCECs transiently elevated intracellular Ca2+ through the Gq/PLC signaling pathway. LPAR3 agonists may potentially have therapeutic efficacy in ocular surface diseases, including dry eye disease.
Collapse
Affiliation(s)
- Ethan S Lindgren
- Department of Ophthalmology, University of California, San Francisco, USA
| | - Rongshan Yan
- Department of Ophthalmology, University of California, San Francisco, USA
| | - Yien-Ming Kuo
- Department of Ophthalmology, University of California, San Francisco, USA
| | - Qi Gao
- Department of Pediatrics, University of California, San Francisco, USA
| | | | - Feeling Y Chen
- Department of Cell & Tissue Biology, University of California, San Francisco, USA
| | - Matilda F Chan
- Department of Ophthalmology, University of California, San Francisco, USA; Francis I. Proctor Foundation, University of California, San Francisco, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, USA
| | - Onur Cil
- Department of Pediatrics, University of California, San Francisco, USA
| | - Neel D Pasricha
- Department of Ophthalmology, University of California, San Francisco, USA; Francis I. Proctor Foundation, University of California, San Francisco, USA.
| |
Collapse
|
4
|
Solís KH, Romero-Ávila MT, Alcántara-Hernández R, García-Sáinz JA. The many facets of biased signaling: Mechanisms and possible therapeutic implications. Pharmacol Ther 2025:108877. [PMID: 40383400 DOI: 10.1016/j.pharmthera.2025.108877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/08/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
Receptor-mediated cell activation frequently results in a plethora of effects, and interestingly, not all agonists that act on a given receptor activate all of those actions to the same extent. Biased agonism refers to this fact, i.e., the possibility to activate only a part of the receptor's signaling capabilities. It is worth mentioning that Biased Signaling is an integral concept that includes the system (organisms, isolated tissues, or cells), the individual receptor studied, and the ligands. It should be remembered that the system's genetic expression profile defines the type, abundance, and cellular localization of proteins that participate in signaling. This short review will be focused on G protein receptors, but biased signaling occurs in many other receptor types. Biased signaling can be related to the G proteins and β-arrestins available. Similarly, enzymes that catalyze receptor posttranslational modifications, such as phosphorylation, acylation, or ubiquitination, can play a role. G protein-coupled receptor signaling occurs at the plasma membrane, but it is well-established that endosomal signaling is a functional reality. Therefore, paying attention to cellular elements that participate in receptor endosomal traffic and destination (recycling to the plasma membrane/ degradation) is pertinent. There is still much to be known about these bias mechanisms, which are essential for basic knowledge of receptor drug action and for treating many pathological entities.
Collapse
Affiliation(s)
- K Helivier Solís
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria. Ap. Postal 70-600, Ciudad de México 04510. Mexico
| | - M Teresa Romero-Ávila
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria. Ap. Postal 70-600, Ciudad de México 04510. Mexico.
| | - Rocío Alcántara-Hernández
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria. Ap. Postal 70-600, Ciudad de México 04510. Mexico.
| | - J Adolfo García-Sáinz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria. Ap. Postal 70-600, Ciudad de México 04510. Mexico.
| |
Collapse
|
5
|
Sánchez-Marín L, Jiménez-Castilla V, Flores-López M, Navarro JA, Gavito A, Blanco-Calvo E, Santín LJ, Pavón-Morón FJ, Rodríguez de Fonseca F, Serrano A. Sex-specific alterations in emotional behavior and neurotransmitter systems in LPA 1 receptor-deficient mice. Neuropharmacology 2025; 268:110325. [PMID: 39864586 DOI: 10.1016/j.neuropharm.2025.110325] [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: 10/31/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
Lysophosphatidic acid (LPA) and the endocannabinoid system (ECS) are critical lipid signaling pathways involved in emotional regulation and behavior. Despite their interconnected roles and shared metabolic pathways, the specific contributions of LPA signaling through the LPA1 receptor to stress-related disorders remain poorly understood. This study investigates the effects of LPA1 receptor deficiency on emotional behavior and neurotransmitter-related gene expression, with a focus on sex-specific differences, using maLPA1-null mice of both sexes. We hypothesized LPA1 receptor loss disrupts the interplay between LPA and the endocannabinoid 2-arachidonoylglycerol (2-AG) signaling, resulting in distinct behavioral and molecular alterations. maLPA1-null mice exhibited increased anxiety-like behaviors and altered stress-coping responses compared to wild-type counterparts, with more pronounced effects observed in females. Female mice also displayed higher corticosterone levels, though no genotype-related differences were observed. Plasma analyses revealed elevated LPA levels in maLPA1-null mice, suggesting a compensatory mechanism, and reduced 2-AG levels, indicating impaired ECS signaling. Gene expression profiling in the amygdala and medial prefrontal cortex showed significant alterations in the gene expression of key components of LPA and 2-AG signaling pathways, as well as neuropeptide systems such as corticotropin-releasing hormone (CRH) and neuropeptide Y (NPY). Glutamatergic signaling components also exhibited sex-specific variations. These findings suggest that LPA1 receptor deficiency impacts behavioral response and disrupts sex-specific neurotransmitter signaling, emphasizing the importance of LPA-ECS crosstalk in emotional regulation. This study provides insights into the molecular mechanisms underlying stress-related disorders such as depression and anxiety, which may inform the development of sex-specific therapeutic approaches.
Collapse
Affiliation(s)
- Laura Sánchez-Marín
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Violeta Jiménez-Castilla
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - María Flores-López
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Juan A Navarro
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Servicio de Neurología, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Ana Gavito
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Servicio de Neurología, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Eduardo Blanco-Calvo
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010, Málaga, Spain
| | - Luis J Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010, Málaga, Spain
| | - Francisco J Pavón-Morón
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria de Málaga, 29010, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Servicio de Neurología, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain; Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), 29001, Malaga, Spain.
| | - Antonia Serrano
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain.
| |
Collapse
|
6
|
Jiang J, Zhang Y, Zuo Y, Bai Y, He Q. Gαq/11 Signaling Modulates Fibroblast Growth Factor 23 Production and Contributes to Acute Kidney Injury. FASEB J 2025; 39:e70544. [PMID: 40275688 DOI: 10.1096/fj.202402848rr] [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: 11/14/2024] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
Fibroblast growth factor 23 (FGF23), primarily secreted by osteocytes and osteoblasts, is essential in regulating phosphate-calcium metabolism by inhibiting renal phosphate reabsorption and vitamin D synthesis. Acute kidney injury (AKI) is characterized by a rapid deterioration in renal function, accompanied by a significant increase in FGF23 levels, which contribute to its severity and progression. This study investigated the mechanistic roles of Gαq and Gα11 proteins, integral components of the lysophosphatidic acid (LPA) signaling pathway, in the regulation of FGF23 expression during AKI. Through targeted knockdown and pharmacological inhibition of Gαq and Gα11 in the osteoblastic MC3T3-E1 and osteocytic MLO-Y4 cells, we demonstrated that individual suppression of these G proteins had minimal impact on both basal and LPA-stimulated FGF23 levels. In contrast, concurrent knockdown significantly diminished FGF23 expression, implicating a synergistic role of Gαq and Gα11 in FGF23 regulation. This hypothesis was supported by using Gαq/11-specific inhibitors, YM-254890 and FR900359, which attenuated LPA-induced FGF23 upregulation. Our findings further elucidated the downstream signaling events, highlighting the involvement of PKC phosphorylation, intracellular calcium mobilization, and the MAPK/ERK1/2 pathway in mediating FGF23 expression. Moreover, in a folic acid-induced AKI mouse model, elevated FGF23 levels in bone, bone marrow, and serum were significantly reduced following YM-254890 administration, underscoring the potential of targeting Gαq/11 signaling in managing AKI-associated FGF23 dysregulation. This study not only advances our understanding of FGF23 regulation in renal injuries but also identifies Gαq/11 signaling modulation as a promising strategy to alleviate AKI severity and other disorders associated with dysregulated FGF23 levels.
Collapse
Affiliation(s)
- Jie Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yue Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yiyi Zuo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qing He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Solís KH, Romero-Ávila MT, Rincón-Heredia R, Romero-Romero S, Correa-Basurto J, García-Sáinz JA. Multiple LPA 3 Receptor Agonist Binding Sites Evidenced Under Docking and Functional Studies. Int J Mol Sci 2025; 26:4123. [PMID: 40362362 PMCID: PMC12071260 DOI: 10.3390/ijms26094123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Comparative studies using lysophosphatidic acid (LPA) and the synthetic agonist, oleoyl-methoxy glycerophosphothionate (OMPT), in cells expressing the LPA3 receptor revealed differences in the action of these agents. The possibility that more than one recognition cavity might exist for these ligands in the LPA3 receptor was considered. We performed agonist docking studies exploring the whole protein to obtain tridimensional details of the ligand-receptor interaction. Functional in cellulo experiments using mutants were also executed. Our work includes blind docking using the unrefined and refined proteins subjected to hot spot predictions. Distinct ligand protonation (charge -1 and -2) states were evaluated. One LPA recognition cavity is located near the lower surface of the receptor close to the cytoplasm (Lower Cavity). OMPT displayed an affinity for an additional identification cavity detected in the transmembrane and extracellular regions (Upper Cavity). Docking targeted to Trp102 favored binding of both ligands in the transmembrane domain near the extracellular areas (Upper Cavity), but the associating amino acids were not identical due to close sub-cavities. A receptor model was generated using AlphaFold3, which properly identified the transmembrane regions of the sequence and co-modeled the lipid environment accordingly. These two models independently generated (with and without the membrane) and adopted essentially the same conformation, validating the data obtained. A DeepSite analysis of the model predicted two main binding pockets, providing additional confidence in the predicted ligand-binding regions and support for the relevance of the docking-based interaction models. In addition, mutagenesis was performed of the amino acids of the two detected cavities. In the in cellulo studies, LPA action was much less affected by the distinct mutations than that of OMPT (which was almost abolished). Therefore, docking and functional data indicate the presence of distinct agonist binding cavities in the LPA3 receptor.
Collapse
Affiliation(s)
- K. Helivier Solís
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico; (K.H.S.); (M.T.R.-Á.)
| | - M. Teresa Romero-Ávila
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico; (K.H.S.); (M.T.R.-Á.)
| | - Ruth Rincón-Heredia
- Unidad de Imagenología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico;
| | - Sergio Romero-Romero
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria. Ap. Postal 70-600, Ciudad de México 04510, Mexico;
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón S/N, Casco de Santo Tomas, Miguel Hidalgo, Ciudad de México 11340, Mexico;
| | - J. Adolfo García-Sáinz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico; (K.H.S.); (M.T.R.-Á.)
| |
Collapse
|
8
|
Ma D, Jiang N, Zhang J, Lei H, Zhai X. Development of potent indole-3-carboxamide autotaxin inhibitors with preferred lipophilicity for in vivo treatment of pulmonary fibrosis. Eur J Med Chem 2025; 288:117398. [PMID: 39983555 DOI: 10.1016/j.ejmech.2025.117398] [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: 01/01/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
Autotaxin (ATX), a major source of the lipid mediator lysophosphatidic acid (LPA), plays a critical role in the pathogenesis and progression of pulmonary fibrosis. In this study, with the aim of developing novel ATX inhibitors with preferred lipophilicity, structure-based optimizations of PAT-409 were carried out, leading to the identification of two novel orally active ATX inhibitors, 4 and 29, with IC50 values of 1.5 nM and 1.08 nM, respectively. Both compounds demonstrated favorable physicochemical properties and desirable ADMET profiles. Notably, compounds 4 and 29 exhibited excellent in vitro metabolic stability (t1/2 > 170 min) and negligible cytotoxicity. Furthermore, oral administration of either compound 4 or 29 (60 mg/kg) exhibited comparable anti-pulmonary fibrosis effects to PAT-409 (60 mg/kg) in a bleomycin-induced pulmonary fibrosis mouse model, suggesting their potential as promising anti-pulmonary fibrosis agents for further development.
Collapse
Affiliation(s)
- Deyi Ma
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Nan Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jiachen Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hongrui Lei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
9
|
Ma D, Zhao B, Yue L, Li S, Wei X, Jiang N, Zang L, Lei H, Zhai X. Development of Tricyclic 4,5-Dihydro-3 H-pyrrolo[2,3- c]quinolin-4-ones as Potent Autotaxin Inhibitors for Pulmonary Fibrosis Treatment In Vivo. J Med Chem 2025; 68:7476-7498. [PMID: 40123070 DOI: 10.1021/acs.jmedchem.4c03173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Autotaxin (ATX) has been recognized as an attractive target due to its hyperactivity in hydrolyzing lysophosphatidylcholine (LPC) into lysophosphatidic acid (LPA) throughout the initiation and progression of fibrotic diseases. Herein, a hydrophilic amide linker and sp3-rich bicyclic 4,5,6,7-tetrahydro-7H-pyrazolo[3,4-c]pyridin-7-one scaffold were employed to modify the lead compound PAT-409, followed by benzene ring fusion to generate novel tricyclic 4,5-dihydro-3H-pyrrolo[2,3-c]quinolin-4-one ATX inhibitors. Among them, the pyridine-2-carboxylic derivatives 45 and 46 demonstrated subnanomolar ATX inhibition (IC50 < 1 nM), with a favorable heart safety profile (hERG > 30 μM) and minimal fibroblast toxicity. Significantly, in bleomycin-induced pulmonary fibrosis mouse models, both compounds markedly improved respiratory function and reduced fibrotic lesions. Mechanistic studies revealed that 45 suppressed the TGF-β/Smad signaling pathway, downregulating α-smooth muscle actin (α-SMA) and extracellular matrix components (ECM). Overall, the identification of 45 and 46 for pulmonary fibrosis therapy provides a featured tricyclic scaffold for further development of novel ATX inhibitors.
Collapse
Affiliation(s)
- Deyi Ma
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bing Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lingfeng Yue
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Sen Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiujian Wei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Nan Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linghe Zang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hongrui Lei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
10
|
Jamali M, Bautista Sanchez R, Agarwal P, Khanna D. Advances and future outlook in clinical trials for treating systemic sclerosis-interstitial lung disease. Expert Rev Respir Med 2025:1-13. [PMID: 40197088 DOI: 10.1080/17476348.2025.2490729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
INTRODUCTION Systemic sclerosis-associated interstitial lung disease (SSc-ILD) is a common complication of systemic sclerosis (SSc), contributing significantly to morbidity and mortality. We aim to bridge knowledge gaps, inform clinical practice, and identify future research directions in this rapidly evolving field. AREAS COVERED This review provides a comprehensive analysis of the current understanding and emerging advances in the diagnosis, risk stratification, and treatment of SSc-ILD. High-resolution computed tomography (HRCT) and pulmonary function tests (PFTs) remain cornerstones of diagnosis, but limitations in sensitivity underscore the need for biomarkers such as Chemokine (C-C motif) Ligand 18 (CCL18), Krebs von den Lungen-6 (KL-6), Interleukin-6 (IL-6), and C-reactive protein (CRP) to enhance prognostic precision and treatment personalization. Therapeutic strategies emphasize immunosuppressants alongside antifibrotic agents. Emerging combination therapies and advanced modalities, including hematopoietic stem cell transplantation and chimeric antigen receptor T-cell therapy, show promise in refractory cases. Ongoing clinical trials exploring innovative targets highlight the evolving therapeutic landscape. The review emphasizes challenges in clinical trial design, advocating for adaptive and platform trial methodologies to address disease heterogeneity and enhance treatment sensitivity. EXPERT OPINION Advances in biomarkers, composite indices, and personalized therapeutic approaches are key to overcoming existing limitations and improving outcomes for patients with SSc-ILD.
Collapse
Affiliation(s)
- Marzieh Jamali
- Division of Rheumatology, Department of Medicine, University of Michigan Scleroderma Program, Ann Arbor, MI, USA
- Department of Medicine, University of Michigan Scleroderma Program, Ann Arbor, MI, USA
| | - Rocio Bautista Sanchez
- Division of Rheumatology, Department of Medicine, University of Michigan Scleroderma Program, Ann Arbor, MI, USA
| | - Prachi Agarwal
- Department of Radiology, University of Michigan Division of Cardiothoracic Radiology, Ann Arbor, MI, USA
| | - Dinesh Khanna
- Division of Rheumatology, Department of Medicine, University of Michigan Scleroderma Program, Ann Arbor, MI, USA
- Department of Medicine, University of Michigan Scleroderma Program, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Martin RE, Satz AL, Kuratli C, Hunziker D, Mattei P, Hert J, Ullmer C, Rudolph MG, Alker AM, Hochstrasser R, Marx A, Binder M, Müller S. Optimization of a DNA encoded library derived autotaxin inhibitor hit to a potent in vivo LPA lowering quinazolinone compound with a non‑zinc binding mode. Bioorg Med Chem Lett 2025; 123:130221. [PMID: 40194669 DOI: 10.1016/j.bmcl.2025.130221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025]
Abstract
In recent years, lysophospholipase autotaxin (ATX) has emerged as an attractive target for treating a variety of human diseases, including inflammation, neurodegeneration, angiogenesis, cancer, ocular and fibrotic diseases, among others. Starting with the quinazolinone hit structure 1, which emerged from a DNA-encoded library screen, the potent, non-Zn2+ binding ATX inhibitor 31 with good overall physicochemical properties has been developed. This compound demonstrated a sustained reduction of lysophosphatidic acid (LPA) in an in vivo rat experiment, qualifying it as a proof-of-concept compound for further mechanistic studies.
Collapse
Affiliation(s)
- Rainer E Martin
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland.
| | - Alexander L Satz
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Christoph Kuratli
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Daniel Hunziker
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Patrizio Mattei
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Jérôme Hert
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Christoph Ullmer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Markus G Rudolph
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - André M Alker
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Remo Hochstrasser
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Andreas Marx
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Martin Binder
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Stephan Müller
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| |
Collapse
|
12
|
Kheyrollah M, Brandt N, Bräuer AU, Schrader S, Mertsch S. The role of lysophosphatidic acid and its receptors in corneal nerve regeneration. Ocul Surf 2025; 36:10-18. [PMID: 39709127 DOI: 10.1016/j.jtos.2024.12.006] [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: 08/07/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
The integrity of corneal nerves is critical for ocular surface health, and damages can lead to Neurotrophic Keratopathy (NK). Despite the regenerative abilities of the peripheral nerve system (PNS), corneal nerve regeneration is often incomplete, and the underlying mechanisms are poorly understood. This study aims to identify potential factors that can enhance corneal nerve regeneration for NK treatment, with a focus on Lysophosphatidic acid (LPA). Thus, the effect of LPA and its underlying pathways in nerve regeneration is investigated in detail using in vitro mouse sensory neurons. To elucidate the impact of LPA as well as to reveal the responsible receptor, several functional assays as well as siRNA-based knock-down experiments were conducted. Additionally, possible changes in underlying pathways were investigated on mRNA levels. LPA-treated neurons significantly reduced fiber growth. However, LPAR2 knockdown neurons (Lpar2-KD) following LPA treatment showed a significant increase in fiber length. Additionally, LPA-treated neurons demonstrated enhanced levels of Lpar2 mRNA. On the other hand, nerve regeneration indicators such as Ngf, Gap-43, and Cdc42, along with LPA downstream signaling components like Pi3k and Ras, were elevated in Lpar2-KD neurons. In conclusion, this study elucidates the inhibitory effects of LPA on fiber outgrowth of sensory neurons. Furthermore, LPAR2 was identified as the responsible receptor for the LPA effect. Thus, Lpar2 knockdown might be a promising therapeutic approach to enhance neuronal regeneration in patients with NK.
Collapse
Affiliation(s)
- Maryam Kheyrollah
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany; Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany
| | - Nicola Brandt
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany
| | - Anja U Bräuer
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany; Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Stefan Schrader
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany; Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Sonja Mertsch
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany.
| |
Collapse
|
13
|
Chia ZJ, Kumarapperuma H, Zhang R, Little PJ, Kamato D. Smad transcription factors as mediators of 7 transmembrane G protein-coupled receptor signalling. Acta Pharmacol Sin 2025; 46:795-804. [PMID: 39506064 PMCID: PMC11950520 DOI: 10.1038/s41401-024-01413-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024]
Abstract
The Smad transcription factors are well known for their role at the core of transforming growth factor-β (TGF-β) signalling. However, recent evidence shows that the Smad transcription factors play a vital role downstream of other classes of receptors including G protein-coupled receptors (GPCR). The versatility of Smad transcription factors originated from the two regions that can be differently activated by the TGF-β receptor superfamily or through the recruitment of intracellular kinases stimulated by other receptors classes such as GPCRs. The classic GPCR signalling cascade is further expanded to conditional adoption of the Smad transcription factor under the stimulation of Akt, demonstrating the unique involvement of the Smad transcription factor in GPCR signalling pathways in disease environments. In this review, we provide a summary of the signalling pathways of the Smad transcription factors as an important downstream mediator of GPCRs, presenting exciting opportunities for discovering new therapeutic targets for diseases.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Hirushi Kumarapperuma
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Ruizhi Zhang
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, QLD, Australia
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, QLD, Australia.
| |
Collapse
|
14
|
Patnaik R, Varghese RL, Banerjee Y. Selective Modulation of PAR-2-Driven Inflammatory Pathways by Oleocanthal: Attenuation of TNF-α and Calcium Dysregulation in Colorectal Cancer Models. Int J Mol Sci 2025; 26:2934. [PMID: 40243559 PMCID: PMC11988659 DOI: 10.3390/ijms26072934] [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: 02/07/2025] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Colorectal cancer (CRC) remains a principal contributor to oncological mortality worldwide, with chronic inflammation serving as a fundamental driver of its pathogenesis. Protease-activated receptor-2 (PAR-2), a G-protein-coupled receptor, orchestrates inflammation-driven tumorigenesis by potentiating NF-κB and Wnt/β-catenin signaling, thereby fostering epithelial-mesenchymal transition (EMT), immune evasion, and therapeutic resistance. Despite its pathological significance, targeted modulation of PAR-2 remains an underexplored avenue in CRC therapeutics. Oleocanthal (OC), a phenolic constituent of extra virgin olive oil, is recognized for its potent anti-inflammatory and anti-cancer properties; however, its regulatory influence on PAR-2 signaling in CRC is yet to be elucidated. This study interrogates the impact of OC on PAR-2-mediated inflammatory cascades using HT-29 and Caco-2 CRC cell lines subjected to lipopolysaccharide (LPS)-induced activation of PAR-2. Expression levels of PAR-2 and TNF-α were quantified through Western blotting and RT-PCR, while ELISA assessed TNF-α secretion. Intracellular calcium flux, a pivotal modulator of PAR-2-driven oncogenic inflammation, was evaluated via Fluo-4 calcium assays. LPS markedly elevated PAR-2 expression at both mRNA and protein levels in CRC cells (p < 0.01, one-way ANOVA). OC administration (20-150 μg/mL) elicited a dose-dependent suppression of PAR-2, with maximal inhibition at 100-150 μg/mL (p < 0.001, Tukey's post hoc test). Concomitant reductions in TNF-α transcription (p < 0.01) and secretion (p < 0.001) were observed, corroborating the anti-inflammatory efficacy of OC. Additionally, OC ameliorated LPS-induced calcium dysregulation, restoring intracellular calcium homeostasis in a concentration-dependent manner (p < 0.01). Crucially, OC exhibited selectivity for PAR-2, leaving PAR-1 expression unaltered (p > 0.05), underscoring its precision as a therapeutic agent. These findings position OC as a selective modulator of PAR-2-driven inflammation in CRC, disrupting the pro-tumorigenic microenvironment through attenuation of TNF-α secretion, calcium dysregulation, and oncogenic signaling pathways. This study furnishes mechanistic insights into OC's potential as a nutraceutical intervention in inflammation-associated CRC. Given the variability in OC bioavailability and content in commercial olive oil, future investigations should delineate optimal dosing strategies and in vivo efficacy to advance its translational potential in CRC therapy.
Collapse
Affiliation(s)
- Rajashree Patnaik
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates; (R.P.); (R.L.V.)
| | - Riah Lee Varghese
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates; (R.P.); (R.L.V.)
| | - Yajnavalka Banerjee
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates; (R.P.); (R.L.V.)
- Centre for Medical Education, School of Medicine, University of Dundee Ninewells Hospital Dundee, Dundee DD2 1SG, UK
| |
Collapse
|
15
|
Antonopoulou G, Magkrioti C, Chatzidaki I, Nastos D, Grammenoudi S, Bozonelos K, Aidinis V. Generation of New Knock-Out Mouse Strains of Lysophosphatidic Acid Receptor 1. Int J Mol Sci 2025; 26:2811. [PMID: 40141453 PMCID: PMC11942715 DOI: 10.3390/ijms26062811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
The lysophosphatidic acid receptor 1 (LPAR1) is one of the six cognate G protein-coupled receptors of the bioactive, growth factor-like phospholipid lysophosphatidic acid (LPA). LPAR1 is widely expressed in different cell types and mediates many LPA effects. LPAR1 has been implicated in several chronic inflammatory diseases, and especially pulmonary fibrosis, where it has been established as a promising therapeutic target. Herein, we present the generation of several Lpar1 mouse strains through genetic recombination. These strains include an initial versatile Lpar1 strain (tm1a) from which three other strains derive: an Lpar1 reporter knockout strain (tm1b) where LacZ has replaced exon 3 of Lpar1; a "floxed" Lpar1 strain (tm1c), where exon 3 is flanked by two loxP sites allowing conditional, cell-specific Lpar1 inactivation; and a complete KO strain of Lpar1 (tm1d), where exon 3 has been deleted. The generated strains are novel genetic tools, that can have various applications in studying LPA-LPAR1 signaling and its role in normal physiology and disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vassilis Aidinis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece; (G.A.); (C.M.); (I.C.); (D.N.); (S.G.); (K.B.)
| |
Collapse
|
16
|
Kwak BJ, Park JH, Kim OH, Lee D, Hong TH, Lee SC, Kim KH, Choi HJ, Kim SJ. A novel strategy for sorafenib-resistant hepatocellular carcinoma: autotaxin Inhibition by PF-8380. J Cancer Res Clin Oncol 2025; 151:110. [PMID: 40082280 PMCID: PMC11906571 DOI: 10.1007/s00432-025-06156-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
By inhibiting the conversion of lysophosphatidylcholine into lysophosphatidic acid, a process pivotal to tumor progression, the autotaxin (ATX) inhibitor PF-8380 offers a new anticancer therapeutic strategy, distinct from the action mechanism of sorafenib. This study explored the potential anticancer effects of the PF-8380 on hepatocellular carcinoma (HCC) cells, especially sorafenib-resistant strains. The investigation included both in vitro and in vivo experiments to evaluate the impact of PF-8380 treatment on epithelial-mesenchymal transition (EMT) and autophagy markers. An orthotopic HCC model served as the in vivo platform. PF-8380 showed a significant reduction in cell viability in both sorafenib-susceptible and resistant HCC cells. It effectively altered EMT by increasing E-cadherin and reducing Snail levels, and inhibited autophagy, as indicated by changes in LC3 and p62 markers. These effects were consistently observed in the orthotopic HCC mouse model, reinforcing PF-8380's potential as a dual inhibitor of EMT and autophagy in HCC treatment. Our research indicates that PF-8380 could provide substantial therapeutic benefits in the treatment of HCC, even in cases resistant to sorafenib, primarily by suppressing both EMT and autophagy processes.
Collapse
Affiliation(s)
- Bong Jun Kwak
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Jung Hyun Park
- Department of Surgery, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 03312, Republic of Korea
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Ok-Hee Kim
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Translational Research Team, Surginex Co., Ltd., Seoul, 06591, Republic of Korea
| | - Dosang Lee
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Tae Ho Hong
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Sang Chul Lee
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, 34943, Republic of Korea
| | - Kee-Hwan Kim
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Surgery, College of Medicine, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Gyeonggi-do, 11765, Republic of Korea
| | - Ho Joong Choi
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Say-June Kim
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
17
|
Gutiérrez-Rojas C, Córdova-Casanova A, Faundez-Contreras J, Cruz-Soca M, Gallardo FS, Bock-Pereda A, Casar JC, Barton ER, Brandan E. Dysregulated ATX-LPA and YAP/TAZ signaling in dystrophic Sgcd -/- mice with early fibrosis and inflammation. Skelet Muscle 2025; 15:6. [PMID: 40050938 PMCID: PMC11884125 DOI: 10.1186/s13395-025-00375-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 02/13/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Sarcoglycanopathies are muscle dystrophies caused by mutations in the genes encoding sarcoglycans (α, β, γ, and δ) that can destabilize the dystrophin-associated glycoprotein complex at the sarcolemma, leaving muscle fibers vulnerable to damage after contraction, followed by inflammatory and fibrotic responses and resulting in muscle weakness and atrophy. Two signaling pathways have been implicated in fibrosis and inflammation in various tissues: autotaxin/lysophosphatidic acid (ATX-LPA) and yes-associated protein 1/transcriptional co-activator with PDZ-binding motif (YAP/TAZ). LPA, synthesized by ATX, can act as a pleiotropic molecule due to its multiple receptors. Two Hippo pathway effectors, YAP/TAZ, can be dephosphorylated by LPA and translocated to the nucleus. They induce several target genes, such as CCN2/CTGF, involved in fibrosis and inflammation. However, no detailed characterization of these processes or whether these pathways change early in the development of sarcoglycanopathy has been evaluated in skeletal muscle. METHODS Using the δ-sarcoglycan knockout mouse model (Sgcd-/-), we investigated components of these pathways, inflammatory and fibrotic markers, and contractile properties of different skeletal muscles (triceps-TR, gastrocnemius-GST, diaphragm-DFG, tibialis anterior-TA, and extensor digitorum longus-EDL) at one and two months of age. RESULTS We found that Sgcd-/- mice show early dystrophic features (fiber damage/necrosis, centrally nucleated fibers, inflammatory infiltrate, and regenerated fibers) followed by later fiber size reduction in TR, GST, and DFG. These changes are concomitant with an early inflammatory and fibrotic response in these muscles. Sgcd-/- mice also have early impaired force generation in the TA and EDL, and resistance to mechanical damage in the EDL. In addition, an early dysregulation of the ATX-LPA axis and the YAP/TAZ signaling pathway in the TR, GST, and DFG was observed in these mice. CONCLUSIONS The ATX-LPA axis and the YAP/TAZ signaling pathway, which are involved in inflammation and fibrosis, are dysregulated in skeletal muscle from an early age in Sgcd-/- mice. These changes are concomitant with a fibrotic and inflammatory response in these mice. Unraveling the role of the LPA axis and YAP/TAZ in sarcoglycanopathy holds great promise for improving our understanding of disease pathogenesis and identifying novel therapeutic targets for this currently incurable group of muscle disorders.
Collapse
Affiliation(s)
- Cristian Gutiérrez-Rojas
- Escuela de Kinesiología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, 2340025, Valparaíso, Chile.
- Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330025, Santiago, Chile.
- Centro Científico y Tecnológico de Excelencia, Ciencia & Vida, 8580702, Santiago, Chile.
| | | | - Jennifer Faundez-Contreras
- Centro Científico y Tecnológico de Excelencia, Ciencia & Vida, 8580702, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, 7510602, Santiago, Chile
| | - Meilyn Cruz-Soca
- Centro Científico y Tecnológico de Excelencia, Ciencia & Vida, 8580702, Santiago, Chile
| | - Felipe S Gallardo
- Centro Científico y Tecnológico de Excelencia, Ciencia & Vida, 8580702, Santiago, Chile
| | - Alexia Bock-Pereda
- Centro Científico y Tecnológico de Excelencia, Ciencia & Vida, 8580702, Santiago, Chile
| | - Juan Carlos Casar
- Departamento de Neurología, Pontificia Universidad Católica de Chile, 7820436, Santiago, Chile
| | - Elisabeth R Barton
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Enrique Brandan
- Centro Científico y Tecnológico de Excelencia, Ciencia & Vida, 8580702, Santiago, Chile.
- Facultad de Medicina y Ciencia, Universidad San Sebastián, 7510602, Santiago, Chile.
| |
Collapse
|
18
|
Choi IY, Kim YJ, Kim SY, Lee MK, Seol GH. Rb 1 restores palmitic acid-induced reduction of Ca 2+ influx by activating PLC in EA cells and PLD in MOVAS cells. Biomed Pharmacother 2025; 184:117927. [PMID: 39970733 DOI: 10.1016/j.biopha.2025.117927] [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: 11/21/2024] [Revised: 02/07/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025] Open
Abstract
Recent interest has focused on the role of Ca2+ in regulating health problems, including cardiovascular disease and colorectal cancer. The inverse correlation between colon cancer and serum Ca2+ underlines the importance of understanding intracellular Ca2+ dynamics. Studies are also evaluating the contributions of abnormalities in Ca²⁺ homeostasis and intracellular dysfunction to the pathogenesis of metabolic syndrome as a precursor of cardiovascular disease. In this study, we investigated the changes of Ca2+ dynamics and ginsenoside Rb1 (Rb1)-induced recovery in two vascular cell lines exposed to palmitic acid (PA), the most abundant active ingredient in palm oil. The mechanism underlying the Rb1-induced recovery was examined in a store operated Ca2+ entry model by Ca2+ store depletion. PA reduced the Ca2+ influx in both EA.hy926 (EA) and MOVAS cells, and this change was restored by Rb1. In EA cells, the Rb1-induced restoration was abolished by U73122 or 2-APB. In MOVAS cells, meanwhile, the effect of Rb1 was abolished by FIPI, U73122 and U73343. Under normal conditions, Rb1 itself altered phospholipid signaling (PLC in EA cells and PLD in MOVAS cells), but did not affect Ca2+ homeostasis. These differences resulted in differences in downstream actions, as KB-R7943 and nifedipine inhibited Rb1-mediated Ca2+ influx recovery only in MOVAS cells. In conclusion, Rb1 rescues the PA-induced Ca2+ influx by appropriately activating PLC in EA cells and PLD in MOVAS cells. This demonstrates that Ca2+ dynamics are elaborately regulated via intracellular Ca2+ signaling networks, suggesting a potential strategy for maintaining vascular Ca2+ homeostasis in hyperlipidemic environments.
Collapse
Affiliation(s)
- In-Young Choi
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
| | - Yoo Jin Kim
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea; BK21 FOUR Program of Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University, Seoul, Republic of Korea
| | - So Young Kim
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
| | - Min Kyung Lee
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea
| | - Geun Hee Seol
- Department of Basic Nursing Science, College of Nursing, Korea University, Seoul, Republic of Korea; BK21 FOUR Program of Transdisciplinary Major in Learning Health Systems, Graduate School, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Birker-Robaczewska M, Boucher M, Ranieri G, Poirey S, Studer R, Freti D, Schnoebelen M, Froidevaux S, Morrison K, Wyss C, Scherer J, Lescop C, Brotschi C, Bolli MH, Kramberg M, Di Stefano S, Rey M, Iglarz M, Delahaye S, Vezzali E, Sieber P, Schäfer A, Caimi SL, Hesse C, Nayler O. The novel lysophosphatidic acid receptor 1-selective antagonist, ACT-1016-0707, has unique binding properties that translate into effective antifibrotic and anti-inflammatory activity in different models of pulmonary fibrosis. J Pharmacol Exp Ther 2025; 392:103396. [PMID: 40073729 DOI: 10.1016/j.jpet.2025.103396] [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: 06/15/2024] [Accepted: 01/30/2025] [Indexed: 03/14/2025] Open
Abstract
Pulmonary fibrosis encompasses different chronic interstitial lung diseases, and the predominant form, idiopathic pulmonary fibrosis, remains to have a poor prognosis despite 2 approved therapies. Although the exact pathobiological mechanisms are still incompletely understood, epithelial injury and aberrant wound healing responses contribute to the gradual change in lung architecture and functional impairment. Lysophosphatidic acid (LPA)-induced lysophosphatidic receptor 1 (LPA1) signaling was proposed to be a driver of lung fibrosis, and LPA1 antagonists have shown promising antifibrotic profiles in early clinical development. The novel, potent, and selective LPA1 antagonist, ACT-1016-0707, displayed insurmountable LPA1 antagonism in vitro with slow off-rate kinetics, leading to efficient inhibition of LPA1 signaling even in presence of high concentrations of LPA. This binding property translated into potent and highly efficient prevention of LPA-induced skin vascular leakage by ACT-1016-0707 in vivo, differentiating the compound from surmountable LPA1 antagonists. Furthermore, ACT-1016-0707 attenuated proinflammatory and profibrotic signaling in different lung fibrosis models in vitro and in the bleomycin-induced lung fibrosis model in vivo. Based on these data, ACT-1016-0707 shows potential as best-in-class LPA1 antagonist for treatment of fibrotic diseases. SIGNIFICANCE STATEMENT: ACT-1016-0707 is a potent, selective, and insurmountable lysophosphatidic receptor 1 (LPA1) antagonist demonstrating robust antifibrotic and anti-inflammatory activity in different lung fibrosis models in vitro and in vivo. This study is the first to demonstrate functional in vivo evidence of insurmountable LPA1 antagonist superiority by side-by-side comparison with surmountable LPA1 antagonists in highly controlled conditions, suggesting potential for ACT-1016-0707 as best-in-class LPA1 antagonist for treatment of fibrotic diseases.
Collapse
Affiliation(s)
| | | | | | | | - Rolf Studer
- Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Diego Freti
- Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | | | | | | | - Conrad Wyss
- Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | | | | | | | | | | | | | - Markus Rey
- Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Marc Iglarz
- Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | | | | | | | - Anny Schäfer
- Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | | | - Christina Hesse
- Fraunhofer Institute for Toxicology and Experimental Medicine, Department of Preclinical Pharmacology and In-Vitro Toxicology, Member of German Center for Lung Research (DZL), Hannover, Germany
| | | |
Collapse
|
20
|
Jose A, Pakkiriswami S, Mercer A, Paudel Y, Yi E, Fernando J, Pulinilkunnil T, Kienesberger PC. Effect of cardiomyocyte-specific lipid phosphate phosphatase 3 overexpression on high-fat diet-induced cardiometabolic dysfunction in mice. Am J Physiol Heart Circ Physiol 2025; 328:H333-H347. [PMID: 39805037 DOI: 10.1152/ajpheart.00518.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Lipid phosphate phosphatase 3 (LPP3) is a membrane-bound enzyme that hydrolyzes lipid phosphates including the bioactive lipid, lysophosphatidic acid (LPA). Elevated circulating LPA production and cellular LPA signaling are implicated in obesity-induced metabolic and cardiac dysfunction. Deletion of LPP3 in the cardiomyocyte increases circulating LPA levels and causes heart failure and mitochondrial dysfunction in mice. To examine the influence of LPP3 modulation in the cardiomyocyte on obesity-induced cardiomyopathy, we generated mice with cardiomyocyte-specific LPP3 overexpression (LPP3OE mice) driven by the α myosin heavy chain promoter. Female and male control (LPP3FL) and LPP3OE mice were fed low-fat diet (LFD) or high-fat diet (HFD) for up to 22-23 wk, followed by the analysis of glucose homeostasis, cardiac function, plasma LPA levels, and mitochondrial respiration in cardiac myofibers. On LFD, both female and male LPP3OE mice had markedly reduced plasma LPA levels and increased pyruvate-linked respiration when compared with LPP3FL mice while body weight and global insulin sensitivity were similar between genotypes. Following HFD feeding, female LPP3OE mice were protected from increased plasma LPA levels, excess adiposity, systemic insulin resistance, and systolic and diastolic cardiac dysfunction compared with LPP3FL mice. Female LPP3OE mice also maintained elevated cardiac pyruvate-linked mitochondrial respiration following HFD feeding while mitochondrial respiration was similar between genotypes in HFD-fed male mice. This study suggests that cardiomyocyte-specific LPP3 upregulation protects particularly female mice from HFD-induced metabolic dysfunction and cardiomyopathy.NEW & NOTEWORTHY Lipid phosphate phosphatase 3 (LPP3) hydrolyzes bioactive lipids including lysophosphatidic acid (LPA), elevated levels of which are implicated in obesity-induced metabolic and cardiac dysfunction. We show that cardiac-specific overexpression of LPP3 lowers plasma LPA levels, blunts LPA signaling in cardiomyocytes, and increases pyruvate-linked mitochondrial respiration in the heart at baseline in both male and female mice. In female mice, LPP3 overexpression also protects from high-fat diet-induced obesity, insulin resistance, and cardiac dysfunction.
Collapse
Affiliation(s)
- Anu Jose
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Shanmugasundaram Pakkiriswami
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Angella Mercer
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Yadab Paudel
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Esther Yi
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Jeffy Fernando
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| | - Petra C Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
| |
Collapse
|
21
|
Pang X, Gu L, Han QY, Xing JQ, Zhao M, Huang SY, Yi JX, Pan J, Hong H, Xue W, Zhou XQ, Su ZH, Zhang XR, Sun LM, Jiang SZ, Luo D, Chen L, Wang ZJ, Yu Y, Xia T, Zhang XM, Li AL, Zhou T, Cai H, Li T. RGS22 maintains the physiological function of ependymal cells to prevent hydrocephalus. SCIENCE CHINA. LIFE SCIENCES 2025; 68:441-453. [PMID: 39400871 DOI: 10.1007/s11427-024-2720-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/30/2024] [Indexed: 10/15/2024]
Abstract
Ependymal cells line the wall of cerebral ventricles and ensure the unidirectional cerebrospinal fluid (CSF) flow by beating their motile cilia coordinately. The ependymal denudation or ciliary dysfunction causes hydrocephalus. Here, we report that the deficiency of regulator of G-protein signaling 22 (RGS22) results in severe congenital hydrocephalus in both mice and rats. Interestingly, RGS22 is specifically expressed in ependymal cells within the brain. Using conditional knock-out mice, we further demonstrate that the deletion of Rgs22 exclusively in nervous system is sufficient to induce hydrocephalus. Mechanistically, we show that Rgs22 deficiency leads to the ependymal denudation and impaired ciliogenesis. This phenomenon can be attributed to the excessive activation of lysophosphatidic acid receptor (LPAR) signaling under Rgs22-/- condition, as the LPAR blockade effectively alleviates hydrocephalus in Rgs22-/- rats. Therefore, our findings unveil a previously unrecognized role of RGS22 in the central nervous system, and present RGS22 as a potential diagnostic and therapeutic target for hydrocephalus.
Collapse
Affiliation(s)
- Xue Pang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Lin Gu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Qiu-Ying Han
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Jia-Qing Xing
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Ming Zhao
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Shao-Yi Huang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Jun-Xi Yi
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Jie Pan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Hao Hong
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Wen Xue
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Xue-Qing Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Zhi-Hui Su
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Xin-Ran Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Li-Ming Sun
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Shao-Zhen Jiang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Dan Luo
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ling Chen
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Zheng-Jie Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Yu Yu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Tian Xia
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Xue-Min Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ai-Ling Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Hong Cai
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China.
| | - Tao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China.
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
22
|
Kim KS, Lee R, Park I, Hwang SH, Kim Y, Jang JW, Kim HS, Choi SM, Kim SJ, Cho HJ, Cho IH, Kim JH, Kim DG, Nah SY. Gintonin Binds to Reduced LPA4 Receptor Subtype in Human Cortical Neurons in Alzheimer's Disease Brains. Biomolecules 2025; 15:179. [PMID: 40001482 PMCID: PMC11853258 DOI: 10.3390/biom15020179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Ginseng, a traditional herbal medicine with a long history of use, is known to support human health, particularly by influencing brain function. Recent studies have identified gintonin, a lysophosphatidic acid (LPA) receptor ligand derived from ginseng, as a key bioactive. However, the specific LPA receptor subtypes targeted by gintonin in the human brain to exert its anti-Alzheimer's (AD) effects remain unclear. This study aimed to elucidate the LPA receptor subtype targeted by gintonin in the human cortex. Using a fluorescent gintonin conjugate, we investigated receptor binding in cortical samples from healthy individuals (n = 4) and AD patients (n = 4). Our results demonstrated that fluorescent gintonin selectively binds to human cortical neurons rather than glial cells and that gintonin-binding sites are co-localized with the LPA4 receptor subtype. Furthermore, the expression of LPA4 receptors was significantly reduced in the cortical neurons of AD patients. These results suggest that the LPA4 receptor may serve as a novel histopathological marker for AD and represent a promising therapeutic target for gintonin-based prevention and treatment strategies.
Collapse
Affiliation(s)
- Kyu-Sung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea;
- Neuroimmunology Laboratory, Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Rami Lee
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Inyeong Park
- Neuroimmunology Laboratory, Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Sung-Hee Hwang
- Department of Pharmaceutical Engineering, College of Health Sciences, Sangji University, Wonju 26339, Republic of Korea;
| | - Yeshin Kim
- Department of Neurology, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea; (Y.K.), (J.-W.J.)
| | - Jae-Won Jang
- Department of Neurology, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea; (Y.K.), (J.-W.J.)
| | - Hyung-Seok Kim
- Department of Neurosurgery, Chonnam National University Medical School, Research Institute of Medical Sciences, Gwangju 61469, Republic of Korea;
| | - Seong-Min Choi
- Department of Neurology, Chonnam National University Medical School, Jebong-ro, Gwangju 61469, Republic of Korea;
| | - Sang Jin Kim
- Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan 47392, Republic of Korea;
- Dementia and Neurodegenerative Disease Research Center, Inje University, Busan 47392, Republic of Korea
| | - Hwa Jin Cho
- Busan & Gyeongnam Reference Laboratory, Department of Pathology, Seegene Medical Foundation, Busan 48792, Republic of Korea;
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan-City 54596, Republic of Korea;
| | - Do-Geun Kim
- Neuroimmunology Laboratory, Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
23
|
Ma D, Tan Z, Li S, Zhao B, Yue L, Wei X, Xu S, Jiang N, Lei H, Zhai X. Discovery of Novel 4,5,6,7-Tetrahydro-7 H-pyrazolo[3,4- c]pyridin-7-one Derivatives as Orally Efficacious ATX Allosteric Inhibitors for the Treatment of Pulmonary Fibrosis. J Med Chem 2025; 68:792-818. [PMID: 39720950 DOI: 10.1021/acs.jmedchem.4c02719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Pulmonary fibrosis (PF) is a progressive, fatal lung disease lacking effective treatments. Autotaxin (ATX) plays a crucial role in exacerbating inflammation and fibrosis, making it a promising target for fibrosis therapies. Herein, starting from PAT-409 (Cudetaxestat), a series of novel ATX inhibitors bearing 1H-indole-3-carboxamide, 4,5,6,7-tetrahydro-7H-pyrazolo[3,4-c]pyridin-7-one, or 4,5,6,7-tetrahydro-1H-pyrazolo[4,3-c]pyridine cores were designed based on the structure of ATX hydrophobic tunnel. The optimal 31 and 35 inhibited ATX with IC50 values of 2.8 and 0.7 nM, respectively. In a bleomycin-induced mouse PF model, both compounds significantly reduced fibrosis by regulating the TGF-β/Smad signaling pathway and downregulating collagen deposition. Furthermore, 35 exhibited both negligibly low hERG channel inhibition (IC50 > 30 μM) and remarkable microsomal stability. Notably, 35 was characterized by favorable pharmacokinetic properties (F = 69.5%) and excellent safety in vivo. Overall, 35 turned out to be a well-characterized potent and safe ATX inhibitor warranting further investigation for the treatment of PF.
Collapse
Affiliation(s)
- Deyi Ma
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zehui Tan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Sen Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bing Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lingfeng Yue
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiujian Wei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Sha Xu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Nan Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hongrui Lei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
24
|
Lin TY, Huang TY, Chiu HC, Chung YY, Lin WC, Lin HY, Lee SY. 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucoside-stimulated dental pulp stem cells-derived exosomes for wound healing and bone regeneration. J Dent Sci 2025; 20:154-163. [PMID: 39873051 PMCID: PMC11762248 DOI: 10.1016/j.jds.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/18/2024] [Indexed: 01/30/2025] Open
Abstract
Background/purpose -2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucoside (THSG) is a bioactive component in the Chinese herb Polygonum multiflorum, recognized for its anti-inflammatory and lipid-lowering properties. Human dental pulp stem cells (hDPSCs) have excellent capabilities in tooth regeneration, wound healing, and neural repair. The exosomes (Exo) released by hDPSCs contain bioactive molecules that influence cell proliferation, differentiation, and immune responses. Therefore, we aimed to unveil the potential of THSG-Exo and evaluate its regenerative capabilities through the in vitro experiment and rat bone defect model. Materials and methods The effects of hDPSC-derived exosomes, with or without THSG treatment, on repair and bone regeneration were evaluated through in vitro and in vivo studies. Finally, we conducted a proteomic analysis to meticulously compare the compositional contents of the two types of exosomes. Results In vitro data showed that 10 and 100 μM THSG-Exo enhanced cell proliferation and osteogenic differentiation, reducing wound size to 40 % of its original size. In our maxillary bone defect rat model, THSG-Exo significantly increased bone volume, trabecular thickness, and bone density in the bone defect area. In addition, proteomic analysis of THSG-Exo revealed diverse proteins linked to bone differentiation and tissue repair, including bone morphogenetic protein-1 (BMP-1) and tumor necrosis factor (TNF)-α-stimulated gene 6 (TNFAIP6). Our searches in functional databases revealed that THSG-Exo is involved in numerous biological pathways. Conclusion THSG-Exo enhanced cell proliferation, wound healing, and osteogenesis in vitro, while also expediting tissue repair and bone regeneration in vivo. The protein diversity of THSG-Exo contributes significant value in both basic and regenerative medicine.
Collapse
Affiliation(s)
- Tzu-Yu Lin
- Department of Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tung-Yung Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| | - Hsien-Chung Chiu
- Department of Periodontology, School of Dentistry, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Yao-Yu Chung
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chun Lin
- Department of Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei, Taiwan
- Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yun Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Yang Lee
- Department of Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
25
|
Zhang X, Lau HCH, Ha S, Liu C, Liang C, Lee HW, Ng QWY, Zhao Y, Ji F, Zhou Y, Pan Y, Song Y, Zhang Y, Lo JCY, Cheung AHK, Wu J, Li X, Xu H, Wong CC, Wong VWS, Yu J. Intestinal TM6SF2 protects against metabolic dysfunction-associated steatohepatitis through the gut-liver axis. Nat Metab 2025; 7:102-119. [PMID: 39779889 PMCID: PMC11774752 DOI: 10.1038/s42255-024-01177-7] [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: 09/18/2023] [Accepted: 11/14/2024] [Indexed: 01/11/2025]
Abstract
Transmembrane-6 superfamily member 2 (TM6SF2) regulates hepatic fat metabolism and is associated with metabolic dysfunction-associated steatohepatitis (MASH). TM6SF2 genetic variants are associated with steatotic liver disease. The pathogenesis of MASH involves genetic factors and gut microbiota alteration, yet the role of host-microbe interactions in MASH development remains unclear. Here, we discover that mice with intestinal epithelial cell-specific knockout of Tm6sf2 (Tm6sf2ΔIEC) develop MASH, accompanied by impaired intestinal barrier and microbial dysbiosis. Transplanting stools from Tm6sf2ΔIEC mice induces steatohepatitis in germ-free recipient mice, whereas MASH is alleviated in Tm6sf2ΔIEC mice co-housed with wild-type mice. Mechanistically, Tm6sf2-deficient intestinal cells secrete more free fatty acids by interacting with fatty acid-binding protein 5 to induce intestinal barrier dysfunction, enrichment of pathobionts, and elevation of lysophosphatidic acid (LPA) levels. LPA is translocated from the gut to the liver, contributing to lipid accumulation and inflammation. Pharmacological inhibition of the LPA receptor suppresses MASH in both Tm6sf2ΔIEC and wild-type mice. Hence, modulating microbiota or blocking the LPA receptor is a potential therapeutic strategy in TM6SF2 deficiency-induced MASH.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Suki Ha
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chuanfa Liu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Cong Liang
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hye Won Lee
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Queena Wing-Yin Ng
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yi Zhao
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fenfen Ji
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yunfei Zhou
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yasi Pan
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yang Song
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Yating Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jennie Ching Yin Lo
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alvin Ho Kwan Cheung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, China
| | - Xiaoxing Li
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongzhi Xu
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Chi Chun Wong
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jun Yu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
26
|
Dietrich J, Kang A, Tielemans B, Verleden SE, Khalil H, Länger F, Bruners P, Mentzer SJ, Welte T, Dreher M, Jonigk DD, Ackermann M. The role of vascularity and the fibrovascular interface in interstitial lung diseases. Eur Respir Rev 2025; 34:240080. [PMID: 39909504 PMCID: PMC11795288 DOI: 10.1183/16000617.0080-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/24/2024] [Indexed: 02/07/2025] Open
Abstract
Interstitial lung disease (ILD) is a clinical term that refers to a diverse group of non-neoplastic lung diseases. This group includes idiopathic and secondary pulmonary entities that are often associated with progressive pulmonary fibrosis. Currently, therapeutic approaches based on specific structural targeting of pulmonary fibrosis are limited to nintedanib and pirfenidone, which can only slow down disease progression leading to a lower mortality rate. Lung transplantation is currently the only available curative treatment, but it is associated with high perioperative mortality. The pulmonary vasculature plays a central role in physiological lung function, and vascular remodelling is considered a hallmark of the initiation and progression of pulmonary fibrosis. Different patterns of pulmonary fibrosis commonly exhibit detectable pathological features such as morphomolecular changes, including intussusceptive and sprouting angiogenesis, vascular morphometry, broncho-systemic anastomoses, and aberrant angiogenesis-related gene expression patterns. Dynamic cellular interactions within the fibrovascular interface, such as endothelial activation and endothelial-mesenchymal transition, are also observed. This review aims to summarise the current clinical, radiological and pathological diagnostic algorithm for different ILDs, including usual interstitial pneumonia/idiopathic pulmonary fibrosis, non-specific interstitial pneumonia, alveolar fibroelastosis/pleuroparenchymal fibroelastosis, hypersensitivity pneumonitis, systemic sclerosis-related ILD and coronavirus disease 2019 injury. It emphasises an interdisciplinary clinicopathological perspective. Additionally, the review covers current therapeutic strategies and knowledge about associated vascular abnormalities.
Collapse
Affiliation(s)
- Jana Dietrich
- Institute of Pathology, University Clinics Aachen, RWTH University of Aachen, Aachen, Germany
- J. Dietrich and A. Kang share first authorship
| | - Alice Kang
- Department of Pneumology and Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany
- J. Dietrich and A. Kang share first authorship
| | - Birger Tielemans
- Institute of Pathology, University Clinics Aachen, RWTH University of Aachen, Aachen, Germany
| | - Stijn E Verleden
- Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), University of Antwerp, Edegem, Belgium
- Department of Respiratory Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Hassan Khalil
- Laboratory of Adaptive and Regenerative Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Thoracic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Florian Länger
- Institute of Pathology, University Clinics Aachen, RWTH University of Aachen, Aachen, Germany
| | - Philipp Bruners
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Steven J Mentzer
- Laboratory of Adaptive and Regenerative Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Thoracic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tobias Welte
- Department of Respiratory Medicine and Infectious Disease, Hannover Medical School, Hannover, Germany
| | - Michael Dreher
- Department of Pneumology and Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Danny D Jonigk
- Institute of Pathology, University Clinics Aachen, RWTH University of Aachen, Aachen, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- D.D. Jonigk and M. Ackermann share senior authorship
| | - Maximilian Ackermann
- Institute of Pathology, University Clinics Aachen, RWTH University of Aachen, Aachen, Germany
- Institute of Pathology and Molecular Pathology, Helios University Clinic Wuppertal, University of Witten/Herdecke, Wuppertal, Germany
- Institute of Anatomy, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany
- D.D. Jonigk and M. Ackermann share senior authorship
| |
Collapse
|
27
|
Piotrowska-Tomala KK, Szóstek-Mioduchowska A, Jonczyk AW, Drzewiecka EM, Wrobel MH, Hojo T, Ferreira-Dias G, Skarzynski DJ. The effect of lysophosphatidic acid on myometrial contractility and the mRNA transcription of its receptors in the myometrium at different stages of endometrosis in mares. BMC Vet Res 2024; 20:571. [PMID: 39696406 DOI: 10.1186/s12917-024-04384-2] [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: 08/28/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Endometrosis (chronic degenerative endometritis) results in morphological changes in the equine endometrium and impairs its secretory function. However, the effect of this condition on the myometrium remains unclear. Lysophosphatidic acid (LPA) may affect female reproductive function and embryo transport by influencing uterine contractility through its receptors (LPARs). The objective of this study was to determine myometrial LPAR1-6 mRNA transcription, and the effects of LPA on myometrial contractions in mares with endometrosis during the mid-luteal and follicular phases of the estrous cycle. RESULTS A reduction in myometrial LPAR1 mRNA transcription was observed in mares with endometrosis during the mid-luteal phase, in comparison to those with category I endometria (P < 0.05). While, upregulation of myometrial LPAR3 or LPAR6 mRNA transcription was observed in mares with category III or IIB endometria; respectively (P < 0.05). An increase in myometrial LPAR1, LPAR3 and LPAR5 mRNA transcription was observed during the follicular phase in mares with category IIA endometrium in comparison to their expression in category I endometrium (P < 0.05). During endometrosis progression LPA reduced the force of myometrial contractions in both phases of the estrous cycle (P < 0.05). However, in mares with category IIA endometrium during the follicular phase, LPA was found to increase the force of contraction of myometrial strips in comparison to mares with category I endometrium (P < 0.01). CONCLUSION In the course of endometrosis in mares, a disruption in the myometrial mRNA transcription of LPARs has been observed. This is the first study to examine the impact of LPA on myometrial contractility at diffrent stage of endometrosis. However, it is essential to consider that multiple factors may contribute to this process. Alternations in contractile activity and changes in myometrial LPARs mRNA transcription may indicate impaired LPA-signaling mechanisms in equine myometrium during endometrosis.
Collapse
Affiliation(s)
| | - Anna Szóstek-Mioduchowska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748, Olsztyn, Poland
| | | | - Ewa Monika Drzewiecka
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748, Olsztyn, Poland
| | - Michał Hubert Wrobel
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748, Olsztyn, Poland
| | - Takuo Hojo
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748, Olsztyn, Poland
- Kyushu Okinawa Agricultural Research Center, NARO, 2421, Suya, Koshi, Kumamoto, 861-1192, Japan
| | - Graca Ferreira-Dias
- Faculty of Veterinary Medicine, C.I.I.S.A, University of Lisbon, Lisbon, Portugal
- AL4AnimalS-Associate Laboratory for Animal and Veterinary Sciences, Lisbon, Portugal
| | - Dariusz Jan Skarzynski
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748, Olsztyn, Poland.
| |
Collapse
|
28
|
Bin Dayel AF, Alrasheed NM, Alonazi AS, Alamin MA, Al-Mutairi NM, Alateeq RA. Renoprotective effect of liraglutide on diabetic nephropathy by modulation of Krüppel-like transcription factor 5 expression in rats. J Pharm Pharmacol 2024; 76:1563-1571. [PMID: 39403839 DOI: 10.1093/jpp/rgae127] [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: 05/17/2024] [Accepted: 09/17/2024] [Indexed: 03/22/2025]
Abstract
OBJECTIVES Diabetic nephropathy (DN) is a serious consequence of diabetes that can develop through the lysophosphatidic acid axis. The purpose of this study was to determine whether the antidiabetic drug liraglutide can slow the development of diabetic kidney damage by altering the lysophosphatidic acid axis via KLF5. METHODS Wistar albino rats were divided into nondiabetic and diabetic rats (resulting from an intraperitoneal streptozotocin dose of 30 mg/kg and a high-fat diet). These rats were further divided into four groups: nondiabetic control, liraglutide-treated nondiabetic, diabetic control, and liraglutide-treated diabetic. The nondiabetic and diabetic control groups received normal saline for 42 days, while the liraglutide-treated nondiabetic and diabetic groups received normal saline for 21 days, followed by a subcutaneous dose of liraglutide (200 μg/kg/day) for 21 days. Subsequently, serum levels of DN biomarkers were evaluated, and kidney tissues were histologically examined. The protein expression of PCNA, autotaxin, and KLF5 was detected. KEY FINDINGS Liraglutide treatment in diabetic rats decreased DN biomarkers, histological abnormalities in kidney tissues, and the protein expression of PCNA, autotaxin, and KLF5. CONCLUSION Liraglutide can slow the progression of DN by modulating KLF5-related lysophosphatidic acid axis. Thus, liraglutide may be an effective treatment for preventing or mitigating diabetes-related kidney damage.
Collapse
Affiliation(s)
- Anfal F Bin Dayel
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nouf M Alrasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Asma S Alonazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Maha A Alamin
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nawal M Al-Mutairi
- PharmD Program, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Raghad A Alateeq
- PharmD Program, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
29
|
Jose A, Fernando JJ, Kienesberger PC. Lysophosphatidic acid metabolism and signaling in heart disease. Can J Physiol Pharmacol 2024; 102:685-696. [PMID: 38968609 DOI: 10.1139/cjpp-2024-0077] [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] [Indexed: 07/07/2024]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid that is mainly produced by the secreted lysophospholipase D, autotaxin (ATX), and signals through at least six G protein-coupled receptors (LPA1-6). Extracellular LPA is degraded through lipid phosphate phosphatases (LPP1, LPP2, and LPP3) at the plasmamembrane, terminating LPA receptor signaling. The ATX-LPA-LPP3 pathway is critically involved in a wide range of physiological processes, including cell survival, migration, proliferation, angiogenesis, and organismal development. Similarly, dysregulation of this pathway has been linked to many pathological processes, including cardiovascular disease. This review summarizes and interprets current literature examining the regulation and role of the ATX-LPA-LPP3 axis in heart disease. Specifically, the contribution of altered LPA metabolism via ATX and LPP3 and resulting changes to LPA receptor signaling in obesity cardiomyopathy, cardiac mitochondrial dysfunction, myocardial infarction/ischemia-reperfusion injury, hypertrophic cardiomyopathy, and aortic valve stenosis is discussed.
Collapse
Affiliation(s)
- Anu Jose
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Jeffy J Fernando
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Petra C Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| |
Collapse
|
30
|
Akasaka H, Sano FK, Shihoya W, Nureki O. Structural mechanisms of potent lysophosphatidic acid receptor 1 activation by nonlipid basic agonists. Commun Biol 2024; 7:1444. [PMID: 39506093 PMCID: PMC11541586 DOI: 10.1038/s42003-024-07152-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: 12/12/2023] [Accepted: 10/25/2024] [Indexed: 11/08/2024] Open
Abstract
Lysophosphatidic acid receptor 1 (LPA1) is one of the G protein-coupled receptors activated by the lipid mediator, lysophosphatidic acid (LPA). LPA1 is associated with a variety of diseases, and LPA1 agonists have potential therapeutic value for treating obesity and depression. Although potent nonlipid LPA1 agonists have recently been identified, the mechanisms of nonlipid molecule-mediated LPA1 activation remain unclear. Here, we report a cryo-electron microscopy structure of the human LPA1-Gi complex bound to a nonlipid basic agonist, CpY, which has 30-fold higher agonistic activity as compared with LPA. Structural comparisons of LPA1 with other lipid GPCRs revealed that the negative charge in the characteristic binding pocket of LPA1 allows the selective recognition of CpY, which lacks a polar head. In addition, our structure show that the ethyl group of CpY directly pushes W2716.48 to fix the active conformation. Endogenous LPA lacks these chemical features, which thus represent the crucial elements of nonlipid agonists that potently activate LPA1. This study provides detailed mechanistic insights into the ligand recognition and activation of LPA1 by nonlipid agonists, expanding the scope for drug development targeting the LPA receptors.
Collapse
Affiliation(s)
- Hiroaki Akasaka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Fumiya K Sano
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Wataru Shihoya
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| | - Osamu Nureki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
31
|
Ghosh R, Herberg S. The role of YAP/TAZ mechanosignaling in trabecular meshwork and Schlemm's canal cell dysfunction. Vision Res 2024; 224:108477. [PMID: 39208753 PMCID: PMC11470804 DOI: 10.1016/j.visres.2024.108477] [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: 06/01/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
This focused review highlights the importance of yes-associated protein (YAP)/transcriptional coactivator with PDZ binding motif (TAZ) mechanosignaling in human trabecular meshwork and Schlemm's canal cells in response to glaucoma-associated extracellular matrix stiffening and cyclic mechanical stretch, as well as biochemical pathway modulators (with signaling crosstalk) including transforming growth factor beta 2, glucocorticoids, Wnt, lysophosphatidic acid, vascular endothelial growth factor, and oxidative stress. We provide a comprehensive overview of relevant literature from the last decade, highlight intriguing research avenues with translational potential, and close with an outlook on future directions.
Collapse
Affiliation(s)
- Rajanya Ghosh
- Department of Ophthalmology and Visual Sciences, Center for Vision Research, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, Center for Vision Research, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA.
| |
Collapse
|
32
|
Zhang J, Hu J, Li Y, Zhou X, Ke Y, Chen Y. Serum Autotaxin Level Positively Associates with Metabolic-Associated Fatty Liver Disease and Hyperuricemia in Postmenopausal Women. Dig Dis 2024; 43:54-62. [PMID: 39442506 DOI: 10.1159/000542061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Autotaxin (ATX) is an adipokine known to affect energy metabolism and lipid homeostasis. We aimed to evaluate serum ATX levels in metabolic-associated fatty liver disease (MAFLD) and other metabolic disorders in postmenopausal women. METHODS Postmenopausal women who received an annual health examination were included. The metabolic and demographic characteristics of the subjects were collected, including age, gender, weight, height, blood pressure, and biochemical parameters. Serum ATX level was determined by ELISA. RESULTS This cross-sectional includes 20 postmenopausal women and 20 age-paired healthy controls. MAFLD patients showed significant metabolic disturbance presented with increased body mass index (BMI), blood pressure (p < 0.001) and decreased high-density lipoprotein cholesterol (p < 0.05), as well as liver injury companied by elevated ALT (p < 0.05). Serum ATX levels were statistically higher in MAFLD (253.1 ± 52.1 vs. 202.2 ± 53.2 ng/mL; p < 0.01) and positively correlated with ALT (p < 0.001), γ-glutamyltransferase and BMI (p < 0.01), SBP and TG (p < 0.05). Higher ATX group demonstrated worsen metabolic states with greater proportion of MAFLD, higher BMI (p < 0.01), and ALT (p < 0.05). Logistic regression analysis revealed that serum ATX levels would positively independently predicted MAFLD (OR 1.049, 95% CI: 1.001-1.098, p < 0.05) with AUC of 0.763. Serum level of ATX is significantly elevated in hyperuricemia group (257.3 ± 60.9 vs. 214.5 ± 49.4 ng/mL; p < 0.05) and positively correlated with uric acid level (p < 0.01). Serum ATX would also act as diagnosing parameter of hyperuricemia with AUC of 0.706. CONCLUSIONS Among postmenopausal women, serum ATX level is significantly elevated in MAFLD and related to multiple metabolic characteristics, especially hyperuricemia, which would thus serve as a potential noninvasive biomarker as well as a therapeutic target.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiahui Hu
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Li
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xuefeng Zhou
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yini Ke
- Department of Rheumatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Chen
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
33
|
Zeng Y, Mo G, Wang X, Yang Y, Dong Y, Zhong R, Tian N. Investigating the relationship between blood metabolites and diabetic retinopathy using two-sample mendelian randomization and in vivo validation. Sci Rep 2024; 14:22947. [PMID: 39362968 PMCID: PMC11450153 DOI: 10.1038/s41598-024-73337-4] [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: 01/01/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024] Open
Abstract
We addressed fundamental questions about the influence of metabolites on the development of Diabetic retinopathy (DR), and explored the related pathological mechanism. Genome-wide association study (GWAS) database data for metabolites and DR were used to perform Mendelian randomization (MR) studies. The inverse variance weighting (IVW) was chosen as the primary analysis method. Sensitivity analysis was conducted using MR-PRESSO, leave-one-out and Cochran's Q test. Confounding factors were eliminated to ensure robustness. We also conducted metabolic pathway analysis. In vivo experimental validation was conducted using Sprague Dawley rats. The serum metabolites of the DR group rats and normal group rats were examined to evaluate the MR results. The screen identified eighteen metabolites associated with DR risk, twelve of which were known components. Seven metabolites were positively correlated with DR risk, while five could reduce it. Eight metabolites associated with proliferative DR (PDR) risk were identified, four of which are known components. Three of these were positively associated with PDR risk and one metabolite reduced PDR risk. Additionally, two possible metabolic pathways involved in the biological mechanism of DR were identified. The ELISA results showed that the serum levels of isoleucine and 4-HPA were significantly increased in DR rats, while the level of inosine was decreased. This study offers novel insights into the biological mechanisms underlying DR. Metabolites that are causally linked to DR may serve as promising biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Yihuan Zeng
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510504, Guangdong Province, China
| | - Guangmeng Mo
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510504, Guangdong Province, China
| | - Xiaoyv Wang
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510504, Guangdong Province, China
| | - Yan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510504, Guangdong Province, China
| | - Yan Dong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Ruiying Zhong
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510504, Guangdong Province, China
| | - Ni Tian
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Baiyun District, Guangzhou, 510504, Guangdong Province, China.
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510504, Guangdong Province, China.
| |
Collapse
|
34
|
Gao Y, Yang Z, Bajpai AK, Wang W, Zhang L, Xia Z. Resveratrol enhances the antiliver cancer effect of cisplatin by targeting the cell membrane protein PLA2. Front Oncol 2024; 14:1453164. [PMID: 39381045 PMCID: PMC11458693 DOI: 10.3389/fonc.2024.1453164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/30/2024] [Indexed: 10/10/2024] Open
Abstract
Background In this study, we aimed to explore the mechanism by which resveratrol promotes cisplatin-induced death of HepG2 cells and to provide a potential strategy for resveratrol in the treatment of cancer. Methods HepG2 cells were exposed to a range of drug concentrations for 24 h: resveratrol (2.5 μg/mL [10.95 μM], 5 μg/mL [21.91 μM], 10 μg/mL [43.81 μM], 20 μg/mL [87.62 μM], 40 μg/mL [175.25 μM], and 80 μg/mL [350.50 μM]), cisplatin (0.625 μg/mL [2.08 μM], 1.25 μg/mL [4.17 μM], 2.5 μg/mL [8.33 μM], 4.5 μg/mL [15.00 μM], and 10 μg/mL [33.33 μM]), 24 μg/mL (105.15 μM) resveratrol + 9 μg/mL (30.00 μM) cisplatin, and 12 μg/mL (52.57 μM) resveratrol + 4.5 μg/mL (15.00 μM) cisplatin. The interaction of two drugs was evaluated by coefficient of drug interaction (CDI), which was based on the Pharmacological Additivity model. The MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was used to detect the effect of different concentrations of drugs on cell viability, while transcriptome sequencing was used to identify pathways associated with higher gene enrichment. Synchrotron radiation FTIR microspectroscopy experiments and data analysis were conducted to obtain detailed spectral information. The second-derivative spectra were calculated using the Savitzky-Golay algorithm. Single-cell infrared spectral absorption matrices were constructed to analyze the spectral characteristics of individual cells. The Euclidean distance between cells was calculated to assess their spectral similarity. The cell-to-cell Euclidean distance was computed to evaluate the spatial relationships between cells. The target protein of resveratrol was verified by performing a Western blot analysis. Results After 24 h of treatment with resveratrol, HepG2 cell growth was inhibited in a dose-dependent manner. Resveratrol promotes cisplatin-induced HepG2 cell death through membrane-related pathways. It also significantly changes the membrane components of HepG2 cells. Additionally, resveratrol changes the morphology of the HepG2 cell membrane by decreasing the expression of PLA2G2. Conclusion Resveratrol changes the morphology of the HepG2 cell membrane by decreasing the expression of PLA2G2 and promotes cisplatin-induced HepG2 cell death. The combination of cisplatin and resveratrol can play a synergistic therapeutic effect on HepG2 cells.
Collapse
Affiliation(s)
- Yu Gao
- Department of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zhanyi Yang
- Department of Pharmacy, Binzhou Medical University, Yantai, China
| | - Akhilesh Kumar Bajpai
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Wenben Wang
- Department of Pharmacy, Binzhou Medical University, Yantai, China
| | - Liyuan Zhang
- Department of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zhenhong Xia
- Department of Pharmacy, Binzhou Medical University, Yantai, China
- Key Laboratory of Ion Beam Bioengineering, Hefei Institute of Physical Sciences, Chinese Academy of Sciences, Hefei, China
| |
Collapse
|
35
|
Chen W, Wu J, Yang C, Li S, Liu Z, An Y, Wang X, Cao J, Xu J, Duan Y, Yuan X, Zhang X, Zhou Y, Ip JPK, Fu AKY, Ip NY, Yao Z, Liu K. Lipin1 depletion coordinates neuronal signaling pathways to promote motor and sensory axon regeneration after spinal cord injury. Proc Natl Acad Sci U S A 2024; 121:e2404395121. [PMID: 39292743 PMCID: PMC11441493 DOI: 10.1073/pnas.2404395121] [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: 08/05/2024] [Indexed: 09/20/2024] Open
Abstract
Adult central nervous system (CNS) neurons down-regulate growth programs after injury, leading to persistent regeneration failure. Coordinated lipids metabolism is required to synthesize membrane components during axon regeneration. However, lipids also function as cell signaling molecules. Whether lipid signaling contributes to axon regeneration remains unclear. In this study, we showed that lipin1 orchestrates mechanistic target of rapamycin (mTOR) and STAT3 signaling pathways to determine axon regeneration. We established an mTOR-lipin1-phosphatidic acid/lysophosphatidic acid-mTOR loop that acts as a positive feedback inhibitory signaling, contributing to the persistent suppression of CNS axon regeneration following injury. In addition, lipin1 knockdown (KD) enhances corticospinal tract (CST) sprouting after unilateral pyramidotomy and promotes CST regeneration following complete spinal cord injury (SCI). Furthermore, lipin1 KD enhances sensory axon regeneration after SCI. Overall, our research reveals that lipin1 functions as a central regulator to coordinate mTOR and STAT3 signaling pathways in the CNS neurons and highlights the potential of lipin1 as a promising therapeutic target for promoting the regeneration of motor and sensory axons after SCI.
Collapse
Affiliation(s)
- Weitao Chen
- Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen518036, China
| | - Junqiang Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Chao Yang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Hong Kong University of Science and Technology Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong518057, China
| | - Suying Li
- State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food, Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen518057, China
- Shenzhen Key Laboratory of Food Biological Safety Control, Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen518057, China
| | - Zhewei Liu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yongyan An
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Xuejie Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Jiaming Cao
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jiahui Xu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Hong Kong University of Science and Technology Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong518057, China
| | - Yangyang Duan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Hong Kong University of Science and Technology Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong518057, China
| | - Xue Yuan
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Xin Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yiren Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jacque Pak Kan Ip
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Amy K. Y. Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Hong Kong University of Science and Technology Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong518057, China
| | - Nancy Y. Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Hong Kong University of Science and Technology Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong518057, China
| | - Zhongping Yao
- State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food, Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen518057, China
- Shenzhen Key Laboratory of Food Biological Safety Control, Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen518057, China
| | - Kai Liu
- Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen518036, China
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Hong Kong University of Science and Technology Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong518057, China
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
36
|
Roy S, Chakrabarti M, Mondal T, Das TK, Sarkar T, Datta S, Kundu M, Banerjee M, Kulkarni OP. Effect of an Autotaxin Inhibitor, 2-(4-Chlorophenyl)-7-methyl-8-pentylimidazo[1,2- a] Pyrimidin-5(8 H)-one (CBT-295), on Bile Duct Ligation-Induced Chronic Liver Disease and Associated Hepatic Encephalopathy in Rats. ACS Pharmacol Transl Sci 2024; 7:2662-2676. [PMID: 39296254 PMCID: PMC11406694 DOI: 10.1021/acsptsci.4c00066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/21/2024]
Abstract
The role of autotaxin (ATX)-lysophosphatidic acid (LPA) is yet to be explored in the context of liver cirrhosis and associated encephalopathy. Our objective of this study was to evaluate the role of an ATX inhibitor in biliary cirrhosis and associated hepatic encephalopathy in rats. The preliminary investigation revealed significant impairment in liver function, which eventually led to the development of hepatic encephalopathy. Interestingly, LPA levels were significantly increased in the plasma, liver, and brain of rats following bile duct ligation. Subsequently, we tested the efficacy of an ATX inhibitor, CBT-295, in bile duct-induced biliary cirrhosis and neuropsychiatric symptoms associated with hepatic encephalopathy. CBT-295 showed good oral bioavailability and favorable pharmacokinetic properties. CBT-295 exhibited a significant reduction in inflammatory cytokines like TGF-β, TNF-α, and IL-6 levels, also reduced bile duct proliferation marker CK-19, and lowered liver fibrosis, as evident from reduced collagen deposition. The reversal of liver fibrosis with CBT-295 led to a reduction in blood and brain ammonia levels. Furthermore, CBT-295 also reduced neuroinflammation induced by ammonia, which is characterized by a significant reduction in brain cytokine levels. It improved neuropsychiatric symptoms such as locomotor activities, cognitive impairment, and clinical grading scores associated with hepatic encephalopathy. The improvement in hepatic encephalopathy observed with the ATX inhibitor could be the result of its hepatoprotective action and its ability to attenuate neuroinflammation. Therefore, inhibition of ATX-LPA signaling can be a multifactorial approach for the treatment of chronic liver diseases.
Collapse
Affiliation(s)
- Subhasis Roy
- TCG Lifesciences Private Ltd., Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Monali Chakrabarti
- TCG Lifesciences Private Ltd., Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Trisha Mondal
- TCG Lifesciences Private Ltd., Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Tapas Kumar Das
- TCG Lifesciences Private Ltd., Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Tonmoy Sarkar
- TCG Lifesciences Private Ltd., Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Sebak Datta
- TCG Lifesciences Private Ltd., Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Mrinalkanti Kundu
- TCG Lifesciences Private Ltd., Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Manish Banerjee
- TCG Lifesciences Private Ltd., Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Onkar Prakash Kulkarni
- Metabolic Disorders and Neuroscience Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani-Hyderabad Campus, Hyderabad 500078, India
| |
Collapse
|
37
|
Promny T, Scherrer I, Kadam S, Schmid R, Jost T, Distel LV, Arkudas A, Horch RE, Kengelbach-Weigand A. The Effect of Ionizing Irradiation on the Autotaxin-Lysophasphatidic Acid Axis and Interleukin-6/8 Secretion in Different Breast Cancer Cell Lines. J Pers Med 2024; 14:968. [PMID: 39338222 PMCID: PMC11433306 DOI: 10.3390/jpm14090968] [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: 07/30/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The Autotaxin (ATX)-lysophosphatidic acid (LPA) axis is involved in decreasing radiation sensitivity of breast tumor cells. This study aims to further elucidate the effect of irradiation on the ATX-LPA axis and cytokine secretion in different breast cancer cell lines to identify suitable breast cancer subtypes for targeted therapies. METHODS Different breast cancer cell lines (MCF-7 (luminal A), BT-474 (luminal B), SKBR-3 (HER2-positive), MDA-MB-231 and MDA-MB-468 (triple-negative)) and the breast epithelial cell line MCF-10A were irradiated. The influence of irradiation on LPA receptor (LPAR) expression, ATX expression, and Interleukin (IL)-6 and IL-8 secretion was analyzed. Further, the effect of IL-6 and IL-8 on ATX expression of adipose-derived stem cells (ADSC) was investigated. RESULTS Irradiation increased ATX and LPAR2 expression in MDA-MB-231 cells. Additionally, IL-6 secretion was enhanced in MDA-MB-231, and IL-8 secretion in MDA-MB-231 and MDA-MB-468. Stimulation of ADSC with IL-6 and IL-8 increased ATX expression in ADSC. CONCLUSIONS Targeting ATX or its downstream signaling pathways might enhance the sensitivity of triple-negative breast cancer cells to radiation. Further exploration of the interplay between irradiation, the ATX-LPA axis, and inflammatory cytokines may elucidate novel pathways for overcoming radioresistance and improving individual treatment outcomes.
Collapse
Affiliation(s)
- Theresa Promny
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Isabell Scherrer
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Sheetal Kadam
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Rafael Schmid
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Tina Jost
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Luitpold V Distel
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Annika Kengelbach-Weigand
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
38
|
Kanno T, Miyako K, Endo Y. Lipid metabolism: a central modulator of RORγt-mediated Th17 cell differentiation. Int Immunol 2024; 36:487-496. [PMID: 38824406 DOI: 10.1093/intimm/dxae031] [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: 04/07/2024] [Accepted: 05/22/2024] [Indexed: 06/03/2024] Open
Abstract
Among the T helper cell subsets, Th17 cells contribute to the development of various inflammatory and autoimmune diseases, including psoriasis, rheumatoid arthritis, inflammatory bowel disease, steroid-resistant asthma, and multiple sclerosis. Retinoid-related orphan receptor gamma t (RORγt), a nuclear hormone receptor, serves as a master transcription factor for Th17 cell differentiation. Recent findings have shown that modulating the metabolic pathway is critical for Th17 cell differentiation, particularly through the engagement of de novo lipid biosynthesis. Suppression of lipid biosynthesis, either through the pharmacological inhibition or gene deletion of related enzymes in CD4+ T cells, results in significant impairment of Th17 cell differentiation. Mechanistic studies indicate that metabolic fluxes through both the fatty acid and cholesterol biosynthetic pathways have a pivotal role in the regulation of RORγt activity through the generation of endogenous RORγt lipid ligands. This review discusses recent discoveries highlighting the importance of lipid metabolism in Th17 cell differentiation and function, as well as exploring specific molecular pathways involved in RORγt activation through cellular lipid metabolism. We further elaborate on a pioneering therapeutic approach to improve inflammatory and autoimmune disorders via the inhibition of RORγt.
Collapse
Affiliation(s)
- Toshio Kanno
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, Kisarazu, Chiba 292-0818, Japan
| | - Keisuke Miyako
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, Kisarazu, Chiba 292-0818, Japan
| | - Yusuke Endo
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, Kisarazu, Chiba 292-0818, Japan
| |
Collapse
|
39
|
Yun CC, Han Y, McConnell B. Lysophosphatidic Acid Signaling in the Gastrointestinal System. Cell Mol Gastroenterol Hepatol 2024; 18:101398. [PMID: 39233124 PMCID: PMC11532463 DOI: 10.1016/j.jcmgh.2024.101398] [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: 03/12/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/06/2024]
Abstract
The intestinal epithelium undergoes continuous homeostatic renewal to conduct the digestion and absorption of nutrients. At the same time, the intestinal epithelial barrier separates the host from the intestinal lumen, preventing systemic infection from enteric pathogens. To maintain homeostasis and epithelial functionality, stem cells, which reside in the base of intestinal crypts, generate progenitor cells that ultimately differentiate to produce an array of secretory and absorptive cells. Intestinal regeneration is regulated by niche signaling pathways, specifically, Wnt, bone morphogenetic protein, Notch, and epidermal growth factor. In addition, growth factors and other peptides have emerged as potential modulators of intestinal repair and inflammation through their roles in cellular proliferation, differentiation, migration, and survival. Lysophosphatidic acid (LPA) is such a factor that modulates the proliferation, survival, and migration of epithelial cells while also regulating trafficking of immune cells, both of which are important for tissue homeostasis. Perturbation of LPA signaling, however, has been shown to promote cancer and inflammation. This review focuses on the recent advances in LPA-mediated signaling that contribute to physiological and pathophysiological regulation of the gastrointestinal system.
Collapse
Affiliation(s)
- C Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia.
| | - Yiran Han
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Beth McConnell
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
40
|
Li X, Koyama Y, Taura K, Nishio T, Yoh T, Nishino H, Uemoto Y, Kimura Y, Nakamura D, Nam NH, Sato M, Seo S, Iwaisako K, Hatano E. High expression of autotaxin is associated with poor recurrence-free survival in cholangiocarcinoma. Hepatol Res 2024; 54:817-826. [PMID: 38430513 DOI: 10.1111/hepr.14031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 03/04/2024]
Abstract
BACKGROUND AND AIM Autotaxin (ATX) is an extracellular lysophospholipase D that catalyzes the hydrolysis of lysophosphatidylcholine into lysophosphatidic acid (LPA). Recent accumulating evidence indicates the biological roles of ATX in malignant tumors. However, the expression and clinical implications of ATX in human cholangiocarcinoma (CCA) remain elusive. METHODS In this study, the expression of ATX in 97 human CCA tissues was evaluated by immunohistochemistry. Serum ATX levels were determined in CCA patients (n = 26) and healthy subjects (n = 8). Autotaxin expression in cell types within the tumor microenvironment was characterized by immunofluorescence staining. RESULTS High ATX expression in CCA tissue was significantly associated with a higher frequency of lymph node metastasis (p = 0.050). High ATX expression was correlated with shorter overall survival (p = 0.032) and recurrence-free survival (RFS) (p = 0.001) than low ATX expression. In multivariate Cox analysis, high ATX expression (p = 0.019) was an independent factor for shorter RFS. Compared with low ATX expression, high ATX expression was significantly associated with higher Ki-67-positive cell counts (p < 0.001). Serum ATX levels were significantly higher in male CCA patients than in healthy male subjects (p = 0.030). In the tumor microenvironment of CCA, ATX protein was predominantly expressed in tumor cells, cancer-associated fibroblasts, plasma cells, and biliary epithelial cells. CONCLUSIONS Our study highlights the clinical evidence and independent prognostic value of ATX in human CCA.
Collapse
Affiliation(s)
- Xuefeng Li
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukinori Koyama
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kojiro Taura
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Gastroenterological Surgery and Oncology, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Takahiro Nishio
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoaki Yoh
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroto Nishino
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Uemoto
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Kimura
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daichi Nakamura
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nguyen Hai Nam
- Department of Liver Tumor, Cancer Center, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Motohiko Sato
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoru Seo
- Department of Surgery, Kochi Medical School, Kochi, Japan
| | - Keiko Iwaisako
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Etsuro Hatano
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
41
|
Polyzou A, Fuchs J, Kroon C, Kotoula A, Delis F, Turko P, Antoniou K, Eickholt B, Leondaritis G. Cell type-specific and subcellular expression of phospholipid phosphatase-related proteins to modulate lyso-phosphatidic acid synaptic signaling in the developing and adult CNS. J Neurochem 2024; 168:3050-3062. [PMID: 38994820 DOI: 10.1111/jnc.16169] [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: 12/24/2023] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024]
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid that participates in critical processes in neural development and adult brain function and is implicated in various pathophysiological conditions. Along with its six well-characterized receptors, atypical regulators of LPA signaling have also been suggested, including phospholipid phosphatase-related proteins (PLPPRs). PLPPRs have been mostly studied in the developing brain where they control LPA-dependent axon guidance, cortical network hyperexcitability, and glutamatergic neurotransmission. PLPPR4 and PLPPR3 represent two closely related proteins reported to localize predominantly in dendrites and axons, respectively, and differ in their developmental expression patterns. Herein, we have revised the expression patterns of PLPPRs in the cerebellum, dorsal and ventral hippocampus, prefrontal cortex (PFC), nucleus accumbens, and striatum during development and in the adult using quantitative PCR. Expression patterns of Plppr2,4 and 5 were consistent with previous studies, whereas Plppr3 and Plppr1 exhibited a unique expression profile in nucleus accumbens (NAc) and striatum in later developmental and adult stages, which we verified at the protein level for PLPPR3. To investigate neuron type-specific expression at the single cell level, we developed a bioinformatic tool to analyze recent single-cell RNA-sequencing data in the cerebral cortex and hippocampus of adult mice. Our analysis revealed a widespread but also selective adult neuron-type expression with higher expression levels of Plppr3, Plppr1, and Plppr5 in GABAergic and Plppr4 and Plppr2 in glutamatergic neurons. PLPPR4 has been identified as a post-synaptic modulator of LPA levels in glutamatergic synapses operating via an uptake mechanism, to control LPA-dependent cortical network hyperexcitability. Using subcellular fractionation experiments, we found that both PLPPR4 and PLPPR3 are co-expressed in adult synaptosomal membranes. Furthermore, flow cytometry experiments in HEK293 cells showed comparable LPA uptake by PLPPR4 and PLPPR3, whereas PLPRR3, but not PLPPR4, induced also uptake of monoacylglycerol, the dephosphorylation product of LPA. We propose that synaptic LPA may be subject to both pre-synaptic and post-synaptic mechanisms of regulation by PLPPRs in addition to LPARs in developing and adult synapses.
Collapse
Affiliation(s)
- Alexandra Polyzou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Joachim Fuchs
- Institute of Molecular Biology and Biochemistry, Charité -Universitätsmedizin-Berlin, Berlin, Germany
| | - Cristina Kroon
- Institute of Molecular Biology and Biochemistry, Charité -Universitätsmedizin-Berlin, Berlin, Germany
| | - Androniki Kotoula
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Foteini Delis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Paul Turko
- Institut für Integrative Neuroanatomie, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Katerina Antoniou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Institute of Biosciences, University Research Center Ioannina, University of Ioannina, Ioannina, Greece
| | - Britta Eickholt
- Institute of Molecular Biology and Biochemistry, Charité -Universitätsmedizin-Berlin, Berlin, Germany
| | - George Leondaritis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Institute of Biosciences, University Research Center Ioannina, University of Ioannina, Ioannina, Greece
| |
Collapse
|
42
|
Birgbauer E. Lysophospholipid receptors in neurodegeneration and neuroprotection. EXPLORATION OF NEUROPROTECTIVE THERAPY 2024; 4:349-365. [PMID: 39247084 PMCID: PMC11379401 DOI: 10.37349/ent.2024.00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024]
Abstract
The central nervous system (CNS) is one of the most complex physiological systems, and treatment of CNS disorders represents an area of major medical need. One critical aspect of the CNS is its lack of regeneration, such that damage is often permanent. The damage often leads to neurodegeneration, and so strategies for neuroprotection could lead to major medical advances. The G protein-coupled receptor (GPCR) family is one of the major receptor classes, and they have been successfully targeted clinically. One class of GPCRs is those activated by bioactive lysophospholipids as ligands, especially sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA). Research has been increasingly demonstrating the important roles that S1P and LPA, and their receptors, play in physiology and disease. In this review, I describe the role of S1P and LPA receptors in neurodegeneration and potential roles in neuroprotection. Much of our understanding of the role of S1P receptors has been through pharmacological tools. One such tool, fingolimod (also known as FTY720), which is a S1P receptor agonist but a functional antagonist in the immune system, is clinically efficacious in multiple sclerosis by producing a lymphopenia to reduce autoimmune attacks; however, there is evidence that fingolimod is also neuroprotective. Furthermore, fingolimod is neuroprotective in many other neuropathologies, including stroke, Parkinson's disease, Huntington's disease, Rett syndrome, Alzheimer's disease, and others that are discussed here. LPA receptors also appear to be involved, being upregulated in a variety of neuropathologies. Antagonists or mutations of LPA receptors, especially LPA1, are neuroprotective in a variety of conditions, including cortical development, traumatic brain injury, spinal cord injury, stroke and others discussed here. Finally, LPA receptors may interact with other receptors, including a functional interaction with plasticity related genes.
Collapse
Affiliation(s)
- Eric Birgbauer
- Department of Biology, Winthrop University, Rock Hill, SC 29733, USA
| |
Collapse
|
43
|
Liu W, Mousa AAK, Hopkins AM, Wu YF, Thu KL, Campbell M, Lees SJ, Ramachandran R, Hou J. Lysophosphatidic Acid Receptor 1 (LPA 1) Antagonists as Potential Migrastatics for Triple Negative Breast Cancer. ChemMedChem 2024; 19:e202400013. [PMID: 38648251 DOI: 10.1002/cmdc.202400013] [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: 01/05/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Metastasis is responsible for about 90 % of cancer deaths. Anti-metastatic drugs, termed as migrastatics, offer a distinctive therapeutic approach to address cancer migration and invasion. However, therapeutic exploitation of metastasis-specific targets remains limited, and the effective prevention and suppression of metastatic cancer continue to be elusive. Lysophosphatidic acid receptor 1 (LPA1) is activated by an endogenous lipid molecule LPA, leading to a diverse array of cellular activities. Previous studies have shown that the LPA/LPA1 axis supports the progression of metastasis for many types of cancer. In this study, we report the synthesis and biological evaluation of fluorine-containing triazole derivatives as potent LPA1 antagonists, offering potential as migrastatic drugs for triple negative breast cancer (TNBC). In particular, compound 12 f, the most potent and highly selective in this series with an IC50 value of 16.0 nM in the cAMP assay and 18.4 nM in the calcium mobilization assay, inhibited cell survival, migration, and invasion in the TNBC cell line. Interestingly, the compound did not induce apoptosis in TNBC cells and demonstrated no cytotoxic effects. These results highlight the potential of LPA1 as a migrastatic target. Consequently, the LPA1 antagonists developed in this study hold promise as potential migrastatic candidates for TNBC.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Chemistry, Lakehead University, 955 Oliver Rd, Thunder Bay, ON, P7B 5E1, Canada
- Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON, P7B 6 V4, Canada
| | - Amr A K Mousa
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Austin M Hopkins
- Department of Chemistry, Lakehead University, 955 Oliver Rd, Thunder Bay, ON, P7B 5E1, Canada
| | - Yin Fang Wu
- Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
| | - Kelsie L Thu
- Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Michael Campbell
- Department of Chemistry, Lakehead University, 955 Oliver Rd, Thunder Bay, ON, P7B 5E1, Canada
- Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON, P7B 6 V4, Canada
| | - Simon J Lees
- Northern Ontario School of Medicine University, Thunder Bay, Ontario, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Jinqiang Hou
- Department of Chemistry, Lakehead University, 955 Oliver Rd, Thunder Bay, ON, P7B 5E1, Canada
- Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON, P7B 6 V4, Canada
| |
Collapse
|
44
|
Takai M, Mori S, Honoki K, Tsujiuchi T. Roles of lysophosphatidic acid (LPA) receptor-mediated signaling in cancer cell biology. J Bioenerg Biomembr 2024; 56:475-482. [PMID: 38886303 DOI: 10.1007/s10863-024-10028-9] [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: 09/30/2023] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Lysophosphatidic acid (LPA) is a simple lipid which is endogenously synthesized from lysophosphatidylcholine (LPC) by autotaxin (ATX). LPA mediates a variety of cellular responses through the binding of G protein-coupled LPA receptors (LPA1 to LPA6). It is considered that LPA receptor-mediated signaling plays an important role in the pathogenesis of human malignancy. Genetic alterations and epigenetic changes of LPA receptors have been detected in some cancer cells as well as LPA per se. Moreover, LPA receptors contribute to the promotion of tumor progression, including cell proliferation, invasion, metastasis, tumorigenicity, and angiogenesis. In recent studies, the activation of LPA receptor-mediated signaling regulates chemoresistance and radiosensitivity in cancer cells. This review provides an updated overview on the roles of LPA receptor-mediated signaling in the regulation of cancer cell functions and its potential utility as a molecular target for novel therapies in clinical cancer approaches.
Collapse
Affiliation(s)
- Miwa Takai
- Division of Molecular Oncology, Department of Life Science, Faculty of Science and Engineering, Kindai University, 3-4- 1, Kowakae, Higashiosaka, 577-8502, Osaka, Japan
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8521, Nara, Japan
| | - Kanya Honoki
- Department of Orthopedic Oncology & Reconstructive Medicine, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8521, Nara, Japan
| | - Toshifumi Tsujiuchi
- Division of Molecular Oncology, Department of Life Science, Faculty of Science and Engineering, Kindai University, 3-4- 1, Kowakae, Higashiosaka, 577-8502, Osaka, Japan.
| |
Collapse
|
45
|
Brandt N, Köper F, Hausmann J, Bräuer AU. Spotlight on plasticity-related genes: Current insights in health and disease. Pharmacol Ther 2024; 260:108687. [PMID: 38969308 DOI: 10.1016/j.pharmthera.2024.108687] [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: 02/08/2024] [Revised: 06/07/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
The development of the central nervous system is highly complex, involving numerous developmental processes that must take place with high spatial and temporal precision. This requires a series of complex and well-coordinated molecular processes that are tighly controlled and regulated by, for example, a variety of proteins and lipids. Deregulations in these processes, including genetic mutations, can lead to the most severe maldevelopments. The present review provides an overview of the protein family Plasticity-related genes (PRG1-5), including their role during neuronal differentiation, their molecular interactions, and their participation in various diseases. As these proteins can modulate the function of bioactive lipids, they are able to influence various cellular processes. Furthermore, they are dynamically regulated during development, thus playing an important role in the development and function of synapses. First studies, conducted not only in mouse experiments but also in humans, revealed that mutations or dysregulations of these proteins lead to changes in lipid metabolism, resulting in severe neurological deficits. In recent years, as more and more studies have shown their involvement in a broad range of diseases, the complexity and broad spectrum of known and as yet unknown interactions between PRGs, lipids, and proteins make them a promising and interesting group of potential novel therapeutic targets.
Collapse
Affiliation(s)
- Nicola Brandt
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Franziska Köper
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Jens Hausmann
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Anja U Bräuer
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany; Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
| |
Collapse
|
46
|
Laface C, Ricci AD, Vallarelli S, Ostuni C, Rizzo A, Ambrogio F, Centonze M, Schirizzi A, De Leonardis G, D’Alessandro R, Lotesoriere C, Giannelli G. Autotaxin-Lysophosphatidate Axis: Promoter of Cancer Development and Possible Therapeutic Implications. Int J Mol Sci 2024; 25:7737. [PMID: 39062979 PMCID: PMC11277072 DOI: 10.3390/ijms25147737] [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: 05/30/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Autotaxin (ATX) is a member of the ectonucleotide pyrophosphate/phosphodiesterase (ENPP) family; it is encoded by the ENPP2 gene. ATX is a secreted glycoprotein and catalyzes the hydrolysis of lysophosphatidylcholine to lysophosphatidic acid (LPA). LPA is responsible for the transduction of various signal pathways through the interaction with at least six G protein-coupled receptors, LPA Receptors 1 to 6 (LPAR1-6). The ATX-LPA axis is involved in various physiological and pathological processes, such as angiogenesis, embryonic development, inflammation, fibrosis, and obesity. However, significant research also reported its connection to carcinogenesis, immune escape, metastasis, tumor microenvironment, cancer stem cells, and therapeutic resistance. Moreover, several studies suggested ATX and LPA as relevant biomarkers and/or therapeutic targets. In this review of the literature, we aimed to deepen knowledge about the role of the ATX-LPA axis as a promoter of cancer development, progression and invasion, and therapeutic resistance. Finally, we explored its potential application as a prognostic/predictive biomarker and therapeutic target for tumor treatment.
Collapse
Affiliation(s)
- Carmelo Laface
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Simona Vallarelli
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Carmela Ostuni
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Francesca Ambrogio
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Matteo Centonze
- Personalized Medicine Laboratory, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, “IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.)
| | - Giampiero De Leonardis
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, “IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.)
| | - Rosalba D’Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, “IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.)
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| |
Collapse
|
47
|
Song Y, Ali FN, Ye Z, Zarzoso J, Rogowski J, Sun Y, Xin Y. Pharmacokinetics of Fipaxalparant, a Small-Molecule Selective Negative Allosteric Modulator of Lysophosphatidic Acid Receptor 1, and the Effect of Food in Healthy Volunteers. Clin Pharmacol Drug Dev 2024; 13:819-827. [PMID: 38757472 DOI: 10.1002/cpdd.1417] [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: 12/04/2023] [Accepted: 04/16/2024] [Indexed: 05/18/2024]
Abstract
Dysregulated lysophosphatidic acid receptor 1 (LPAR1) signaling is implicated in fibrotic diseases, including systemic sclerosis (SSc) and idiopathic pulmonary fibrosis (IPF). Fipaxalparant (HZN-825) is a small molecule acting as a negative allosteric modulator of LPAR1 and is in phase 2 clinical evaluations for treating diffuse cutaneous SSc and IPF. This open-label, phase 1 study examined the pharmacokinetics (PKs), food effect, and safety of fipaxalparant in healthy volunteers. Dose proportionality was evaluated for fipaxalparant single doses of 150, 300, and 450 mg under fasted conditions. Food effect was tested with a 450-mg single dose under fasted conditions or with a high-fat meal. Multiple-dose PKs for twice-daily dosing of either 300 or 450 mg with low- or high-fat meals was also assessed. Fipaxalparant was safe and well tolerated in healthy volunteers (n = 36) under all conditions. Fipaxalparant exposure increased in a less than dose-proportional manner from 150 to 450 mg. At 450 mg, a high-fat meal increased the maximum observed concentration and area under the curve by approximately 1.9- and 2.1-fold, respectively. These results, combined with prior preclinical and phase 2a data, informed dose selection of fipaxalparant 300 mg once and twice daily with a meal for phase 2b studies.
Collapse
Affiliation(s)
- Yang Song
- Amgen, Inc. (formerly Horizon Therapeutics plc), South San Francisco, CA, USA
| | - Farah N Ali
- Amgen, Inc. (formerly Horizon Therapeutics plc), Deerfield, IL, USA
| | - Zhan Ye
- Amgen, Inc. (formerly Horizon Therapeutics plc), Deerfield, IL, USA
| | - Jennifer Zarzoso
- Amgen, Inc. (formerly Horizon Therapeutics plc), South San Francisco, CA, USA
| | - John Rogowski
- Amgen, Inc. (formerly Horizon Therapeutics plc), Deerfield, IL, USA
| | - Yajing Sun
- Amgen, Inc. (formerly Horizon Therapeutics plc), South San Francisco, CA, USA
| | - Yan Xin
- Amgen, Inc. (formerly Horizon Therapeutics plc), South San Francisco, CA, USA
| |
Collapse
|
48
|
Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin 2024; 45:1337-1348. [PMID: 38351317 PMCID: PMC11192764 DOI: 10.1038/s41401-024-01235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/28/2024] [Indexed: 02/19/2024]
Abstract
Transforming growth factor-β (TGF-β) signaling is initiated by activation of transmembrane TGF-β receptors (TGFBR), which deploys Smad2/3 transcription factors to control cellular responses. Failure or dysregulation in the TGF-β signaling pathways leads to pathological conditions. TGF-β signaling is regulated at different levels along the pathways and begins with the liberation of TGF-β ligand from its latent form. The mechanisms of TGFBR activation display selectivity to cell types, agonists, and TGF-β isoforms, enabling precise control of TGF-β signals. In addition, the cell surface compartments used to release active TGF-β are surprisingly vibrant, using thrombospondins, integrins, matrix metalloproteinases and reactive oxygen species. The scope of TGFBR activation is further unfolded with the discovery of TGFBR activation initiated by other signaling pathways. The unique combination of mechanisms works in series to trigger TGFBR activation, which can be explored as therapeutic targets. This comprehensive review provides valuable insights into the diverse mechanisms underpinning TGFBR activation, shedding light on potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia
| | - Ying-Nan Cao
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia.
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia.
| |
Collapse
|
49
|
Watanabe M, Tsugeno Y, Sato T, Higashide M, Nishikiori N, Umetsu A, Ogawa T, Furuhashi M, Ohguro H. Lysophosphatidic Acid Modulates TGF-β2-Induced Biological Phenotype in Human Conjunctival Fibroblasts. Life (Basel) 2024; 14:770. [PMID: 38929752 PMCID: PMC11204428 DOI: 10.3390/life14060770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Although lysophosphatidic acid (LPA) is known to have multiple pathophysiological roles, its contributions to ocular tissues, especially conjunctival fibrogenesis, remain to be elucidated. METHODS To study this issue, the effects of LPA on transforming growth factor-β2 (TGF-β2)-induced fibrogenesis of two-dimensional (2D) and three-dimensional (3D) cultures of human conjunctival fibroblasts (HconF) were examined by the following analyses: (1) planar proliferation determined by transepithelial electrical resistance (TEER) and fluorescein isothiocyanate (FITC)-dextran permeability measurements, (2) real-time metabolic analyses, (3) measurements of the size and stiffness of 3D spheroids, and (4) mRNA expression of extracellular matrix (ECM) molecules and their modulators. RESULTS LPA had no effect on TGF-β2-induced increase in the planar proliferation of HconF cells. LPA induced a more quiescent metabolic state in 2D HconF cells, but this metabolic suppression by LPA was partially blunted in the presence of TGF-β2. In contrast, LPA caused a substantial decrease in the hardness of 3D HconF spheroids independently of TGF-β2. In agreement with these different LPA-induced effects between 2D and 3D cultured HconF cells, mRNA expressions of ECM and their modulators were differently modulated. CONCLUSION The findings that LPA induced the inhibition of both TGF-β2-related and -unrelated subepithelial proliferation of HconF cells may be clinically applicable.
Collapse
Affiliation(s)
- Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (N.N.); (A.U.)
| | - Yuri Tsugeno
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (N.N.); (A.U.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Megumi Higashide
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (N.N.); (A.U.)
| | - Nami Nishikiori
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (N.N.); (A.U.)
| | - Araya Umetsu
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (N.N.); (A.U.)
| | - Toshifumi Ogawa
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.); (M.F.)
| | - Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (N.N.); (A.U.)
| |
Collapse
|
50
|
Solís KH, Romero-Ávila MT, Rincón-Heredia R, García-Sáinz JA. Lysophosphatidic Acid Receptor 3 (LPA3): Signaling and Phosphorylation Sites. Int J Mol Sci 2024; 25:6491. [PMID: 38928196 PMCID: PMC11203643 DOI: 10.3390/ijms25126491] [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: 04/05/2024] [Revised: 06/03/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
LPA3 receptors were expressed in TREx HEK 293 cells, and their signaling and phosphorylation were studied. The agonist, lysophosphatidic acid (LPA), increased intracellular calcium and ERK phosphorylation through pertussis toxin-insensitive processes. Phorbol myristate acetate, but not LPA, desensitizes LPA3-mediated calcium signaling, the agonists, and the phorbol ester-induced LPA3 internalization. Pitstop 2 (clathrin heavy chain inhibitor) markedly reduced LPA-induced receptor internalization; in contrast, phorbol ester-induced internalization was only delayed. LPA induced rapid β-arrestin-LPA3 receptor association. The agonist and the phorbol ester-induced marked LPA3 receptor phosphorylation, and phosphorylation sites were detected using mass spectrometry. Phosphorylated residues were detected in the intracellular loop 3 (S221, T224, S225, and S229) and in the carboxyl terminus (S321, S325, S331, T333, S335, Y337, and S343). Interestingly, phosphorylation sites are within sequences predicted to constitute β-arrestin binding sites. These data provide insight into LPA3 receptor signaling and regulation.
Collapse
Affiliation(s)
- K. Helivier Solís
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico; (K.H.S.); (M.T.R.-Á.)
| | - M. Teresa Romero-Ávila
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico; (K.H.S.); (M.T.R.-Á.)
| | - Ruth Rincón-Heredia
- Unidad de Imagenología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico;
| | - J. Adolfo García-Sáinz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico; (K.H.S.); (M.T.R.-Á.)
| |
Collapse
|