1
|
Luo F, Chen T, Chen S, Bai D, Li X. Regulation of osteoclast-mediated bone resorption by lipids. Bone 2025; 193:117423. [PMID: 39933643 DOI: 10.1016/j.bone.2025.117423] [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: 10/30/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
Hyperactivation of osteoclasts has been identified as a significant etiological factor in several bone resorption-related disorders, including osteoporosis, periodontitis, arthritis, and bone metastasis of tumors. It has been demonstrated that the severity of these diseases is influenced by lipids that regulate osteoclast differentiation and activity through specific signaling pathways and cytokine levels. The regulatory mechanisms of different types of lipids on osteoclastogenesis vary across diverse disease contexts in bone resorption regulated by osteoclasts. This review presents an overview of the mechanisms underlying osteoclast formation and summarizes the pathways through which various lipids regulate osteoclastogenesis in different pathological contexts. We also discuss effective therapeutic strategies for osteolytic diseases based on modulation of lipid metabolism.
Collapse
Affiliation(s)
- Fang Luo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Tianyi Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Song Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinyi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Yoon D, Choi B, Kim JE, Kim EY, Chung SH, Min HJ, Sung Y, Chang EJ, Song JK. Autotaxin inhibition attenuates the aortic valve calcification by suppressing inflammation-driven fibro-calcific remodeling of valvular interstitial cells. BMC Med 2024; 22:122. [PMID: 38486246 PMCID: PMC10941471 DOI: 10.1186/s12916-024-03342-x] [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: 06/25/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Patients with fibro-calcific aortic valve disease (FCAVD) have lipid depositions in their aortic valve that engender a proinflammatory impetus toward fibrosis and calcification and ultimately valve leaflet stenosis. Although the lipoprotein(a)-autotaxin (ATX)-lysophosphatidic acid axis has been suggested as a potential therapeutic target to prevent the development of FCAVD, supportive evidence using ATX inhibitors is lacking. We here evaluated the therapeutic potency of an ATX inhibitor to attenuate valvular calcification in the FCAVD animal models. METHODS ATX level and activity in healthy participants and patients with FCAVD were analyzed using a bioinformatics approach using the Gene Expression Omnibus datasets, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and western blotting. To evaluate the efficacy of ATX inhibitor, interleukin-1 receptor antagonist-deficient (Il1rn-/-) mice and cholesterol-enriched diet-induced rabbits were used as the FCAVD models, and primary human valvular interstitial cells (VICs) from patients with calcification were employed. RESULTS The global gene expression profiles of the aortic valve tissue of patients with severe FCAVD demonstrated that ATX gene expression was significantly upregulated and correlated with lipid retention (r = 0.96) or fibro-calcific remodeling-related genes (r = 0.77) in comparison to age-matched non-FCAVD controls. Orally available ATX inhibitor, BBT-877, markedly ameliorated the osteogenic differentiation and further mineralization of primary human VICs in vitro. Additionally, ATX inhibition significantly attenuated fibrosis-related factors' production, with a detectable reduction of osteogenesis-related factors, in human VICs. Mechanistically, ATX inhibitor prohibited fibrotic changes in human VICs via both canonical and non-canonical TGF-β signaling, and subsequent induction of CTGF, a key factor in tissue fibrosis. In the in vivo FCAVD model system, ATX inhibitor exposure markedly reduced calcific lesion formation in interleukin-1 receptor antagonist-deficient mice (Il1rn-/-, P = 0.0210). This inhibition ameliorated the rate of change in the aortic valve area (P = 0.0287) and mean pressure gradient (P = 0.0249) in the FCAVD rabbit model. Moreover, transaortic maximal velocity (Vmax) was diminished with ATX inhibitor administration (mean Vmax = 1.082) compared to vehicle control (mean Vmax = 1.508, P = 0.0221). Importantly, ATX inhibitor administration suppressed the effects of a high-cholesterol diet and vitamin D2-driven fibrosis, in association with a reduction in macrophage infiltration and calcific deposition, in the aortic valves of this rabbit model. CONCLUSIONS ATX inhibition attenuates the development of FCAVD while protecting against fibrosis and calcification in VICs, suggesting the potential of using ATX inhibitors to treat FCAVD.
Collapse
Affiliation(s)
- Dohee Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Bongkun Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Ji-Eun Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Young Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Soo-Hyun Chung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyo-Jin Min
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yoolim Sung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Jae-Kwan Song
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
3
|
Contreras HR, López-Moncada F, Castellón EA. Cancer stem cell and mesenchymal cell cooperative actions in metastasis progression and hormone resistance in prostate cancer: Potential role of androgen and gonadotropin‑releasing hormone receptors (Review). Int J Oncol 2020; 56:1075-1082. [PMID: 32319606 DOI: 10.3892/ijo.2020.5008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the leading cause of male cancer‑associated mortality worldwide. Mortality is associated with metastasis and hormone resistance. Cellular, genetic and molecular mechanisms underlying metastatic progression and hormone resistance are poorly understood. Studies have investigated the local effects of gonadotropin‑releasing hormone (GnRH) analogs (used for androgen deprivation treatments) and the presence of the GnRH receptor (GnRH‑R) on PCa cells. Furthermore, cell subpopulations with stem‑like properties, or cancer stem cells, have been isolated and characterized using a cell culture system derived from explants of human prostate tumors. In addition, the development of preclinical orthotopic models of human PCa in a nonobese diabetic/severe combined immunodeficiency mouse model of compromised immunity has enabled the establishment of a reproducible system of metastatic progression in vivo. There is increasing evidence that metastasis is a complex process involving the cooperative actions of different cancer cell subpopulations, in which cancer stem‑like cells would be responsible for the final step of colonizing premetastatic niches. It has been hypothesized that PCa cells with stemness and mesenchymal signatures act cooperatively in metastatic progression and the inhibition of stemness genes, and that overexpression of androgen receptor (AR) and GnRH‑R decreases the rate the metastasis and sensitizes tumors to hormone therapy. The aim of the present review is to analyze the evidence regarding this cooperative process and the possible influence of stem‑like cell phenotypes, AR and GnRH‑R in metastatic progression and hormone resistance. These aspects may represent an important contribution in the understanding of the mechanisms underlying metastasis and hormone resistance in PCa, and potential routes to blocking these processes, enabling the development of novel therapies that would be particularly relevant for patients with metastatic and castration‑resistant PCa.
Collapse
Affiliation(s)
- Héctor R Contreras
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Fernanda López-Moncada
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Enrique A Castellón
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
4
|
Wu XP, Yang YP, She RX, Xing ZM, Chen HW, Zhang YW. microRNA-329 reduces bone cancer pain through the LPAR1-dependent LPAR1/ERK signal transduction pathway in mice. Ther Adv Med Oncol 2019; 11:1758835919875319. [PMID: 31692673 PMCID: PMC6811758 DOI: 10.1177/1758835919875319] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Bone cancer pain (BCP) is a common symptom occurring among patients with
cancer and has a detrimental effect on their quality of life. Growing
evidence has implicated microRNA-329 (miR-329) in the progression of bone
diseases. In the present study, we aimed to elucidate the potential effects
of miR-329 on BCP in a BCP mouse model via binding to
lysophosphatidic acid receptor 1 (LPAR1) through the LPAR1/extracellular
signal-regulated kinase (ERK) signaling pathway. Methods: Initially, a BCP mouse model was established via injection
of 4 × 104 murine breast tumor (4T1 cell) cells (4 μl). The
interaction between miR-329 and LPAR1 was identified using a bioinformatics
website and dual luciferase reporter gene assay. The modeled mice were
subsequently treated with miR-329 mimic, LPAR1 shRNA, or both, in order to
examine the effect of miR-329 on the paw withdrawal threshold (PWT) and paw
withdrawal latency (PWL) of mice, the expression of LPAR1/ERK signaling
pathway-related genes. Results: The positive expression rate of LPAR1 protein and extent of ERK1/2
phosphorylation were increased in BCP mouse models. LPAR1 is a target gene
of miR-329, which can inhibit the expression of LPAR1. In response to
miR-329 overexpression and LPAR1 silencing, BCP mice showed increased PWT
and PWL, along with decreased LPAR1 expression and ratio of p-ERK/ERK. Conclusions: Altogether, the results obtained indicated that miR-329 can potentially
alleviate BCP in mice via the inhibition of LPAR1 and
blockade of the LPAR1/ERK signaling pathway, highlighting that upregulation
of miR-329 could serve as a therapeutic target for BCP treatment.
Collapse
Affiliation(s)
- Xian-Ping Wu
- Department of Anesthesiology, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, P.R. China
| | - Yan-Ping Yang
- Department of Anesthesiology, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, P.R. China
| | - Rui-Xuan She
- Department of Anesthesiology, Shunde Hospital of Guangzhou University of Chinese Medicine, Foshan, P.R. China
| | - Zu-Min Xing
- Department of Anesthesiology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, P.R. China
| | - Han-Wen Chen
- Department of Anesthesiology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, P.R. China
| | - Yi-Wen Zhang
- Department of Anesthesiology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, P.R. China
| |
Collapse
|
5
|
Wu X, Ma Y, Su N, Shen J, Zhang H, Wang H. Lysophosphatidic acid: Its role in bone cell biology and potential for use in bone regeneration. Prostaglandins Other Lipid Mediat 2019; 143:106335. [PMID: 31054330 DOI: 10.1016/j.prostaglandins.2019.106335] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 02/05/2023]
Abstract
Lysophosphatidic acid (LPA) is a simple phospholipid that exerts pleiotropic effects on numerous cell types by activating its family of cognate G protein-coupled receptors (GPCRs) and participates in many biological processes, including organismal development, wound healing, and carcinogenesis. Bone cells, such as bone marrow mesenchymal stromal (stem) cells (BMSCs), osteoblasts, osteocytes and osteoclasts play essential roles in bone homeostasis and repair. Previous studies have identified the presence of specific LPA receptors in these bone cells. In recent years, an increasing number of cellular effects of LPA, such as the induction of cell proliferation, survival, migration, differentiation and cytokine secretion, have been found in different bone cells. Moreover, some biomaterials containing LPA have shown the ability to enhance osteogenesis. This review will focus on findings associated with LPA functions in these bone cells and present current studies related to the application of LPA in bone regenerative medicine. Further understanding this information will help us develop better strategies for bone healing.
Collapse
Affiliation(s)
- Xiangnan Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuanyuan Ma
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Naichuan Su
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiefei Shen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hai Zhang
- Department of Restorative Dentistry, School of Dentistry, University of Washington, Seattle, WA, 98195, USA
| | - Hang Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Tigyi GJ, Yue J, Norman DD, Szabo E, Balogh A, Balazs L, Zhao G, Lee SC. Regulation of tumor cell - Microenvironment interaction by the autotaxin-lysophosphatidic acid receptor axis. Adv Biol Regul 2018; 71:183-193. [PMID: 30243984 DOI: 10.1016/j.jbior.2018.09.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 12/12/2022]
Abstract
The lipid mediator lysophosphatidic acid (LPA) in biological fluids is primarily produced by cleavage of lysophospholipids by the lysophospholipase D enzyme Autotaxin (ATX). LPA has been identified and abundantly detected in the culture medium of various cancer cell types, tumor effusates, and ascites fluid of cancer patients. Our current understanding of the physiological role of LPA established its role in fundamental biological responses that include cell proliferation, metabolism, neuronal differentiation, angiogenesis, cell migration, hematopoiesis, inflammation, immunity, wound healing, regulation of cell excitability, and the promotion of cell survival by protecting against apoptotic death. These essential biological responses elicited by LPA are seemingly hijacked by cancer cells in many ways; transcriptional upregulation of ATX leading to increased LPA levels, enhanced expression of multiple LPA GPCR subtypes, and the downregulation of its metabolic breakdown. Recent studies have shown that overexpression of ATX and LPA GPCR can lead to malignant transformation, enhanced proliferation of cancer stem cells, increased invasion and metastasis, reprogramming of the tumor microenvironment and the metastatic niche, and development of resistance to chemo-, immuno-, and radiation-therapy of cancer. The fundamental role of LPA in cancer progression and the therapeutic inhibition of the ATX-LPA axis, although highly appealing, remains unexploited as drug development to these targets has not reached into the clinic yet. The purpose of this brief review is to highlight some unique signaling mechanisms engaged by the ATX-LPA axis and emphasize the therapeutic potential that lies in blocking the molecular targets of the LPA system.
Collapse
Affiliation(s)
- Gabor J Tigyi
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA; Institute of Clinical Experimental Research, Semmelweis University, POB 2, H-1428, Budapest, Hungary.
| | - Junming Yue
- Department of Pathology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Derek D Norman
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Erzsebet Szabo
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Andrea Balogh
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA; Institute of Clinical Experimental Research, Semmelweis University, POB 2, H-1428, Budapest, Hungary
| | - Louisa Balazs
- Department of Pathology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Guannan Zhao
- Department of Pathology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Sue Chin Lee
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| |
Collapse
|
7
|
Ninou I, Magkrioti C, Aidinis V. Autotaxin in Pathophysiology and Pulmonary Fibrosis. Front Med (Lausanne) 2018; 5:180. [PMID: 29951481 PMCID: PMC6008954 DOI: 10.3389/fmed.2018.00180] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/25/2018] [Indexed: 12/17/2022] Open
Abstract
Lysophospholipid signaling is emerging as a druggable regulator of pathophysiological responses, and especially fibrosis, exemplified by the relative ongoing clinical trials in idiopathic pulmonary fibrosis (IPF) patients. In this review, we focus on ectonucleotide pyrophosphatase-phosphodiesterase 2 (ENPP2), or as more widely known Autotaxin (ATX), a secreted lysophospholipase D (lysoPLD) largely responsible for extracellular lysophosphatidic acid (LPA) production. In turn, LPA is a bioactive phospholipid autacoid, forming locally upon increased ATX levels and acting also locally through its receptors, likely guided by ATX's structural conformation and cell surface associations. Increased ATX activity levels have been detected in many inflammatory and fibroproliferative conditions, while genetic and pharmacologic studies have confirmed a pleiotropic participation of ATX/LPA in different processes and disorders. In pulmonary fibrosis, ATX levels rise in the broncheoalveolar fluid (BALF) and stimulate LPA production. LPA engagement of its receptors activate multiple G-protein mediated signal transduction pathways leading to different responses from pulmonary cells including the production of pro-inflammatory signals from stressed epithelial cells, the modulation of endothelial physiology, the activation of TGF signaling and the stimulation of fibroblast accumulation. Genetic or pharmacologic targeting of the ATX/LPA axis attenuated disease development in animal models, thus providing the proof of principle for therapeutic interventions.
Collapse
Affiliation(s)
- Ioanna Ninou
- Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Athens, Greece
| | - Christiana Magkrioti
- Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Athens, Greece
| | - Vassilis Aidinis
- Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Athens, Greece
| |
Collapse
|
8
|
Lysophosphatidic Acid Analogue rather than Lysophosphatidic Acid Promoted the Bone Formation In Vivo. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7537630. [PMID: 30003106 PMCID: PMC5996417 DOI: 10.1155/2018/7537630] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/20/2018] [Indexed: 12/29/2022]
Abstract
Lysophosphatidic acid (LPA), a bioactive lipid molecule, has recently emerged as physiological and pathophysiological regulator in skeletal biology. Here we evaluate the effects of LPA on bone formation in vivo in murine femoral critical defect model. Primary femoral osteoblasts were isolated and treated with osteogenic induction conditional media supplemented with 20 μM LPA or LPA analogue. Mineralized nodules were visualized by Alizarin Red S staining. Forty-five C57BL/6 mice underwent unilateral osteotomy. The femoral osteotomy gap was filled with porous scaffolds of degradable chitosan/beta-tricalcium phosphate containing PBS, LPA, or LPA analogue. 2, 5, and 10 weeks after surgery, mice were sacrificed and femurs were harvested and prepared for Micro-Computed Tomography (Micro-CT) and histological analysis. Alizarin Red S staining showed that LPA and LPA analogue significantly enhanced the mineral deposition in osteoblasts. Micro-CT 3D reconstruction images and HE staining revealed that significantly more newly formed bone in osteotomy was treated with LPA analogue when compared to control and LPA group, which was verified by histological analysis and biomechanical characterization testing. In summary, our study demonstrated that although LPA promotes mineralized matrix formation in vitro, the locally administrated LPA was not effective in promoting bone formation in vivo. And bone formation was enhanced by LPA analogue, administrated locally in vivo. LPA analogue was a potent stimulating factor for bone formation in vivo due to its excellent stability.
Collapse
|
9
|
Understanding the Progression of Bone Metastases to Identify Novel Therapeutic Targets. Int J Mol Sci 2018; 19:ijms19010148. [PMID: 29300334 PMCID: PMC5796097 DOI: 10.3390/ijms19010148] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/22/2017] [Accepted: 01/02/2018] [Indexed: 12/15/2022] Open
Abstract
Bone is one of the most preferential target site for cancer metastases, particularly for prostate, breast, kidney, lung and thyroid primary tumours. Indeed, numerous chemical signals and growth factors produced by the bone microenvironment constitute factors promoting cancer cell invasion and aggression. After reviewing the different theories proposed to provide mechanism for metastatic progression, we report on the gene expression profile of bone-seeking cancer cells. We also discuss the cross-talk between the bone microenvironment and invading cells, which impacts on the tumour actions on surrounding bone tissue. Lastly, we detail therapies for bone metastases. Due to poor prognosis for patients, the strategies mainly aim at reducing the impact of skeletal-related events on patients' quality of life. However, recent advances have led to a better understanding of molecular mechanisms underlying bone metastases progression, and therefore of novel therapeutic targets.
Collapse
|
10
|
Suppression of NADPH Oxidase Activity May Slow the Expansion of Osteolytic Bone Metastases. Healthcare (Basel) 2016; 4:healthcare4030060. [PMID: 27571113 PMCID: PMC5041061 DOI: 10.3390/healthcare4030060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/11/2016] [Accepted: 08/22/2016] [Indexed: 12/28/2022] Open
Abstract
Lysophosphatidic acid (LPA), generated in the microenvironment of cancer cells, can drive the proliferation, invasion, and migration of cancer cells by activating G protein-coupled LPA receptors. Moreover, in cancer cells that have metastasized to bone, LPA signaling can promote osteolysis by inducing cancer cell production of cytokines, such as IL-6 and IL-8, which can stimulate osteoblasts to secrete RANKL, a key promoter of osteoclastogenesis. Indeed, in cancers prone to metastasize to bone, LPA appears to be a major driver of the expansion of osteolytic bone metastases. Activation of NADPH oxidase has been shown to play a mediating role in the signaling pathways by which LPA, as well as RANKL, promote osteolysis. In addition, there is reason to suspect that Nox4 activation is a mediator of the feed-forward mechanism whereby release of TGF-beta from bone matrix by osteolysis promotes expression of PTHrP in cancer cells, and thereby induces further osteolysis. Hence, measures which can down-regulate NADPH oxidase activity may have potential for slowing the expansion of osteolytic bone metastases in cancer patients. Phycocyanin and high-dose statins may have utility in this regard, and could be contemplated as complements to bisphosphonates or denosumab for the prevention and control of osteolytic lesions. Ingestion of omega-3-rich flaxseed or fish oil may also have potential for controlling osteolysis in cancer patients.
Collapse
|
11
|
Wu JX, Yuan XM, Wang Q, Wei W, Xu MY. Rho/ROCK acts downstream of lysophosphatidic acid receptor 1 in modulating P2X3 receptor-mediated bone cancer pain in rats. Mol Pain 2016; 12:12/0/1744806916644929. [PMID: 27094551 PMCID: PMC4956381 DOI: 10.1177/1744806916644929] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/20/2016] [Indexed: 12/27/2022] Open
Abstract
Background Lysophosphatidic acid receptor 1 and Rho/ROCK signaling is implicated in bone cancer pain development. However, it remains unknown whether the two signaling pathways function together in P2X3 receptor-mediated bone cancer pain. Results In this study, using a rat model of bone cancer, we examined the expression of P2X3 and lysophosphatidic acid receptor 1 in rat dorsal root ganglion neurons and further dissected whether lysophosphatidic acid receptor 1 and Rho/ROCK-mediated pathways interacted in modulating rat pain behavior. Bone cancer was established by inoculating Walker 256 cells into the left tibia of female Wistar rats. We observed a gradual and yet significant decline in mean paw withdrawal threshold in rats with bone cancer, but not in control rats. Our immunohistochemical staining revealed that the number of P2X3- and lysophosphatidic acid receptor 1-positive dorsal root ganglion neurons was significantly greater in rats with bone cancer than control rats. Lysophosphatidic acid receptor 1 blockade with VPC32183 significantly attenuated decline in mean paw withdrawal threshold. Flinching behavior test further showed that lysophosphatidic acid receptor 1 inhibition with VPC32183 transiently but significantly attenuated α,β-meATP-induced increase in paw lift time per minute. Rho inhibition by intrathecal BoTXC3 caused a rapid reversal in decline in mean paw withdrawal threshold of rats with bone cancer. Flinching behavior test showed that BoTXC3 transiently and significantly attenuated α,β-meATP-induced increase in paw lift time per minute. Similar findings were observed with ROCK inhibition by intrathecal Y27632. Furthermore, VPC32183 and BoTXC3 effectively aborted the appearance of lysophosphatidic acid-induced calcium influx peak. Conclusions Lysophosphatidic acid and its receptor LPAR1, acting through the Rho-ROCK pathway, regulate P2X3 receptor in the development of both mechanical and spontaneous pain in bone cancer.
Collapse
Affiliation(s)
- Jing-Xiang Wu
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, PR China
| | - Xiao-Min Yuan
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, PR China
| | - Qiong Wang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, PR China
| | - Wang Wei
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, PR China
| | - Mei-Ying Xu
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, PR China
| |
Collapse
|
12
|
YU ZILI, LI DIANQI, HUANG XIANGYU, XING XIN, YU RUQING, LI ZHI, LI ZUBING. Lysophosphatidic acid upregulates connective tissue growth factor expression in osteoblasts through the GPCR/PKC and PKA pathways. Int J Mol Med 2016; 37:468-74. [DOI: 10.3892/ijmm.2016.2450] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 12/29/2015] [Indexed: 11/05/2022] Open
|
13
|
Benesch MGK, Tang X, Venkatraman G, Bekele RT, Brindley DN. Recent advances in targeting the autotaxin-lysophosphatidate-lipid phosphate phosphatase axis in vivo. J Biomed Res 2015; 30:272-84. [PMID: 27533936 PMCID: PMC4946318 DOI: 10.7555/jbr.30.20150058] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 05/12/2015] [Accepted: 05/20/2015] [Indexed: 12/21/2022] Open
Abstract
Extracellular lysophosphatidate (LPA) is a potent bioactive lipid that signals through six G-protein-coupled receptors. This signaling is required for embryogenesis, tissue repair and remodeling processes. LPA is produced from circulating lysophosphatidylcholine by autotaxin (ATX), and is degraded outside cells by a family of three enzymes called the lipid phosphate phosphatases (LPPs). In many pathological conditions, particularly in cancers, LPA concentrations are increased due to high ATX expression and low LPP activity. In cancers, LPA signaling drives tumor growth, angiogenesis, metastasis, resistance to chemotherapy and decreased efficacy of radiotherapy. Hence, targeting the ATX-LPA-LPP axis is an attractive strategy for introducing novel adjuvant therapeutic options. In this review, we will summarize current progress in targeting the ATX-LPA-LPP axis with inhibitors of autotaxin activity, LPA receptor antagonists, LPA monoclonal antibodies, and increasing low LPP expression. Some of these agents are already in clinical trials and have applications beyond cancer, including chronic inflammatory diseases.
Collapse
Affiliation(s)
- Matthew G K Benesch
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada
| | - Xiaoyun Tang
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada
| | - Ganesh Venkatraman
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada
| | - Raie T Bekele
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada
| | - David N Brindley
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, T6G 2S2, Canada.
| |
Collapse
|
14
|
Abstract
A small library of truncated/lipid-conjugated neuromedin U (NmU) analogs was synthesized and tested in vitro using an intracellular calcium signaling assay. The selected, most active analogs were then tested in vivo, and showed potent anorexigenic effects in a diet-induced obese (DIO) mouse model. The most promising compound, NM4-C16 was effective in a once-weekly-dose regimen. Collectively, our findings suggest that short, lipidated analogs of NmU are suitable leads for the development of novel anti-obesity therapeutics.
Collapse
|
15
|
Barbayianni E, Kaffe E, Aidinis V, Kokotos G. Autotaxin, a secreted lysophospholipase D, as a promising therapeutic target in chronic inflammation and cancer. Prog Lipid Res 2015; 58:76-96. [DOI: 10.1016/j.plipres.2015.02.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 01/20/2015] [Accepted: 02/12/2015] [Indexed: 02/07/2023]
|
16
|
Abstract
Lysophosphatidic acid (LPA) and its receptors, LPA1-6, are integral parts of signaling pathways involved in cellular proliferation, migration and survival. These signaling pathways are of therapeutic interest for the treatment of multiple types of cancer and to reduce cancer metastasis and side effects. Validated therapeutic potential of key receptors, as well as recent structure-activity relationships yielding compounds with low nanomolar potencies are exciting recent advances in the field. Some compounds have proven efficacious in vivo against tumor proliferation and metastasis, bone cancer pain and the pulmonary fibrosis that can result as a side effect of pulmonary cancer radiation treatment. However, recent studies have identified that LPA contributes through multiple pathways to the development of chemotherapeutic resistance suggesting new applications for LPA antagonists in cancer treatment. This review summarizes the roles of LPA signaling in cancer pathophysiology and recent progress in the design and evaluation of LPA agonists and antagonists.
Collapse
|
17
|
Navab M, Chattopadhyay A, Hough G, Meriwether D, Fogelman SI, Wagner AC, Grijalva V, Su F, Anantharamaiah GM, Hwang LH, Faull KF, Reddy ST, Fogelman AM. Source and role of intestinally derived lysophosphatidic acid in dyslipidemia and atherosclerosis. J Lipid Res 2015; 56:871-87. [PMID: 25646365 DOI: 10.1194/jlr.m056614] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We previously reported that i) a Western diet increased levels of unsaturated lysophosphatidic acid (LPA) in small intestine and plasma of LDL receptor null (LDLR(-/-)) mice, and ii) supplementing standard mouse chow with unsaturated (but not saturated) LPA produced dyslipidemia and inflammation. Here we report that supplementing chow with unsaturated (but not saturated) LPA resulted in aortic atherosclerosis, which was ameliorated by adding transgenic 6F tomatoes. Supplementing chow with lysophosphatidylcholine (LysoPC) 18:1 (but not LysoPC 18:0) resulted in dyslipidemia similar to that seen on adding LPA 18:1 to chow. PF8380 (a specific inhibitor of autotaxin) significantly ameliorated the LysoPC 18:1-induced dyslipidemia. Supplementing chow with LysoPC 18:1 dramatically increased the levels of unsaturated LPA species in small intestine, liver, and plasma, and the increase was significantly ameliorated by PF8380 indicating that the conversion of LysoPC 18:1 to LPA 18:1 was autotaxin dependent. Adding LysoPC 18:0 to chow increased levels of LPA 18:0 in small intestine, liver, and plasma but was not altered by PF8380 indicating that conversion of LysoPC 18:0 to LPA 18:0 was autotaxin independent. We conclude that i) intestinally derived unsaturated (but not saturated) LPA can cause atherosclerosis in LDLR(-/-) mice, and ii) autotaxin mediates the conversion of unsaturated (but not saturated) LysoPC to LPA.
Collapse
Affiliation(s)
- Mohamad Navab
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Arnab Chattopadhyay
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Greg Hough
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - David Meriwether
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Spencer I Fogelman
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Alan C Wagner
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Victor Grijalva
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Feng Su
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - G M Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Lin H Hwang
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Kym F Faull
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Srinivasa T Reddy
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| |
Collapse
|
18
|
Wang L, Sibrian-Vazquez M, Escobedo JO, Wang J, Moore RG, Strongin RM. Spiroguanidine rhodamines as fluorogenic probes for lysophosphatidic acid. Chem Commun (Camb) 2015; 51:1697-700. [PMID: 25516957 PMCID: PMC4320994 DOI: 10.1039/c4cc08818b] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Direct determination of total lysophosphatidic acid (LPA) was accomplished using newly developed spiroguanidines derived from rhodamine B as universal fluorogenic probes. Optimum conditions for the quantitative analysis of total LPA were investigated. The linear range for the determination of total LPA is up to 5 μM with a limit of detection of 0.512 μM.
Collapse
Affiliation(s)
- Lei Wang
- Department of Chemistry, Portland State University, Portland, OR 97201, USA. Tel: +1-503-725-9724
| | - Martha Sibrian-Vazquez
- Department of Chemistry, Portland State University, Portland, OR 97201, USA. Tel: +1-503-725-9724
| | - Jorge O. Escobedo
- Department of Chemistry, Portland State University, Portland, OR 97201, USA. Tel: +1-503-725-9724
| | - Jialu Wang
- Department of Chemistry, Portland State University, Portland, OR 97201, USA. Tel: +1-503-725-9724
| | - Richard G. Moore
- Women and Infants Hospital, Brown University, 101 Dudley Street, Providence, RI 02905, USA; Tel: +1-401-453-7520
| | - Robert M. Strongin
- Department of Chemistry, Portland State University, Portland, OR 97201, USA. Tel: +1-503-725-9724
| |
Collapse
|
19
|
Leblanc R, Peyruchaud O. New insights into the autotaxin/LPA axis in cancer development and metastasis. Exp Cell Res 2014; 333:183-189. [PMID: 25460336 DOI: 10.1016/j.yexcr.2014.11.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 11/13/2014] [Indexed: 12/13/2022]
Abstract
Lysophosphatidic acid (LPA) is a simple lipid with a single fatty acyl chain linked to a glycerophosphate backbone. Despite the simplicity of its structure but owing to its interactions with a series of at least six G protein-coupled receptors (LPA1-6), LPA exerts pleiotropic bioactivities including stimulation of proliferation, migration and survival of many cell types. Autotaxin (ATX) is a unique enzyme with a lysophospholipase D (lysoPLD) activity that is responsible for the levels of LPA in the blood circulation. Both LPA receptor family members and ATX/LysoPLD are aberrantly expressed in many human cancers. This review will present the more striking as well as novel experimental evidences using cell lines, cancer mouse models and transgenic animals identifying the roles for ATX and LPA receptors in cancer progression, tumor cell invasion and metastasis.
Collapse
Affiliation(s)
- Raphaël Leblanc
- INSERM, UMR1033, UCB Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
| | - Olivier Peyruchaud
- INSERM, UMR1033, UCB Lyon 1, Faculté de Médecine Lyon Est, Lyon, France.
| |
Collapse
|
20
|
Abstract
Unlike other blood cells, platelets are small anucleate structures derived from marrow megakaryocytes. Thought for almost a century to possess solely hemostatic potentials, platelets, however, play a much wider role in tissue regeneration and repair and interact intimately with tumor cells. On one hand, tumor cells induce platelet aggregation (TCIPA), known to act as the trigger of cancer-associated thrombosis. On the other hand, platelets recruited to the tumor microenvironment interact, directly, with tumor cells, favoring their proliferation, and, indirectly, through the release of a wide palette of growth factors, including angiogenic and mitogenic proteins. In addition, the role of platelets is not solely confined to the primary tumor site. Indeed, they escort tumor cells, helping their intravasation, vascular migration, arrest, and extravasation to the tissues to form distant metastasis. As expected, nonspecific or specific inhibition of platelets and their content represents an attractive novel approach in the fight against cancer. This review illustrates the role played by platelets at primary tumor sites and in the various stages of the metastatic process.
Collapse
Affiliation(s)
- Hadi A Goubran
- Saskatoon Cancer Centre and Division of Oncology, Department of Medicine, College of Medicine, University of Saskatchewan, SK, Canada.
| | - Julie Stakiw
- Saskatoon Cancer Centre and Division of Oncology, Department of Medicine, College of Medicine, University of Saskatchewan, SK, Canada
| | | | - Thierry Burnouf
- Institute of Biomedical Materials and Tissue Engineering, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
21
|
Autotaxin in the crosshairs: taking aim at cancer and other inflammatory conditions. FEBS Lett 2014; 588:2712-27. [PMID: 24560789 DOI: 10.1016/j.febslet.2014.02.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/11/2014] [Accepted: 02/12/2014] [Indexed: 02/07/2023]
Abstract
Autotaxin is a secreted enzyme that produces most of the extracellular lysophosphatidate from lysophosphatidylcholine, the most abundant phospholipid in blood plasma. Lysophosphatidate mediates many physiological and pathological processes by signaling through at least six G-protein coupled receptors to promote cell survival, proliferation and migration. The autotaxin/lysophosphatidate signaling axis is involved in wound healing and tissue remodeling, and it drives many chronic inflammatory conditions from fibrosis to colitis, asthma and cancer. In cancer, lysophosphatidate signaling promotes resistance to chemotherapy and radiotherapy, and increases both angiogenesis and metastasis. Research into autotaxin inhibitors is accelerating, both as primary and adjuvant therapy. Historically, autotaxin inhibitors had poor bioavailability profiles and thus had limited efficacy in vivo. This situation is now changing, especially since the recent crystal structure of autotaxin is now enabling rational inhibitor design. In this review, we will summarize current knowledge on autotaxin-mediated disease processes including cancer, and discuss recent advancements in the development of autotaxin-targeting strategies. We will also provide new insights into autotaxin as an inflammatory mediator in the tumor microenvironment that promotes cancer progression and therapy resistance.
Collapse
|
22
|
Perrakis A, Moolenaar WH. Autotaxin: structure-function and signaling. J Lipid Res 2014; 55:1010-8. [PMID: 24548887 DOI: 10.1194/jlr.r046391] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Indexed: 12/13/2022] Open
Abstract
Autotaxin (ATX), or ecto-nucleotide pyrophosphatase/phosphodiesterase-2, is a secreted lysophospholipase D (lysoPLD) that hydrolyzes extracellular lysophospholipids into the lipid mediator lysophosphatidic acid (LPA), a ligand for specific G protein-coupled receptors. ATX-LPA signaling is essential for development and has been implicated in a great diversity of (patho)physiological processes, ranging from lymphocyte homing to tumor progression. Structural and functional studies have revealed what makes ATX a unique lysoPLD, and how secreted ATX binds to its target cells. The ATX catalytic domain shows a characteristic bimetallic active site followed by a shallow binding groove that can accommodate nucleotides as well as the glycerol moiety of lysophospholipids, and by a deep lipid-binding pocket. In addition, the catalytic domain has an open tunnel of unknown function adjacent to the active site. Here, we discuss our current understanding of ATX structure-function relationships and signaling mechanisms, and how ATX isoforms use distinct mechanisms to target LPA production to the plasma membrane, notably binding to integrins and heparan sulfate proteoglycans. We also briefly discuss the development of drug-like inhibitors of ATX.
Collapse
Affiliation(s)
- Anastassis Perrakis
- Divisions of Biochemistry, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Wouter H Moolenaar
- Cell Biology, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
23
|
David M, Machuca-Gayet I, Kikuta J, Ottewell P, Mima F, Leblanc R, Bonnelye E, Ribeiro J, Holen I, Vales RL, Jurdic P, Chun J, Clézardin P, Ishii M, Peyruchaud O. Lysophosphatidic acid receptor type 1 (LPA1) plays a functional role in osteoclast differentiation and bone resorption activity. J Biol Chem 2014; 289:6551-6564. [PMID: 24429286 DOI: 10.1074/jbc.m113.533232] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a natural bioactive lipid that acts through six different G protein-coupled receptors (LPA1-6) with pleiotropic activities on multiple cell types. We have previously demonstrated that LPA is necessary for successful in vitro osteoclastogenesis of bone marrow cells. Bone cells controlling bone remodeling (i.e. osteoblasts, osteoclasts, and osteocytes) express LPA1, but delineating the role of this receptor in bone remodeling is still pending. Despite Lpar1(-/-) mice displaying a low bone mass phenotype, we demonstrated that bone marrow cell-induced osteoclastogenesis was reduced in Lpar1(-/-) mice but not in Lpar2(-/-) and Lpar3(-/-) animals. Expression of LPA1 was up-regulated during osteoclastogenesis, and LPA1 antagonists (Ki16425, Debio0719, and VPC12249) inhibited osteoclast differentiation. Blocking LPA1 activity with Ki16425 inhibited expression of nuclear factor of activated T-cell cytoplasmic 1 (NFATc1) and dendritic cell-specific transmembrane protein and interfered with the fusion but not the proliferation of osteoclast precursors. Similar to wild type osteoclasts treated with Ki16425, mature Lpar1(-/-) osteoclasts had reduced podosome belt and sealing zone resulting in reduced mineralized matrix resorption. Additionally, LPA1 expression markedly increased in the bone of ovariectomized mice, which was blocked by bisphosphonate treatment. Conversely, systemic treatment with Debio0719 prevented ovariectomy-induced cancellous bone loss. Moreover, intravital multiphoton microscopy revealed that Debio0719 reduced the retention of CX3CR1-EGFP(+) osteoclast precursors in bone by increasing their mobility in the bone marrow cavity. Overall, our results demonstrate that LPA1 is essential for in vitro and in vivo osteoclast activities. Therefore, LPA1 emerges as a new target for the treatment of diseases associated with excess bone loss.
Collapse
Affiliation(s)
- Marion David
- INSERM, UMR1033, UCB Lyon 1, Faculté de Médecine Lyon Est, 69732 Lyon, France
| | - Irma Machuca-Gayet
- CNRS, UMR5242, ENS, Équipe Biologie Cellulaire et Physiopathologie Osseuse, Institut de Génomique Fonctionnelle de Lyon, UCB Lyon 1, 69007 Lyon, France
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, 565-0871 Osaka, Japan; CREST, Japan Science and Technology Agency, 102-0076 Tokyo, Japan
| | - Penelope Ottewell
- Academic Unit of Clinical Oncology, University of Sheffield Medical School, Beech Hill Road, S10 2RX Sheffield, United Kingdom
| | - Fuka Mima
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, 565-0871 Osaka, Japan; CREST, Japan Science and Technology Agency, 102-0076 Tokyo, Japan
| | - Raphael Leblanc
- INSERM, UMR1033, UCB Lyon 1, Faculté de Médecine Lyon Est, 69732 Lyon, France
| | - Edith Bonnelye
- INSERM, UMR1033, UCB Lyon 1, Faculté de Médecine Lyon Est, 69732 Lyon, France
| | - Johnny Ribeiro
- INSERM, UMR1033, UCB Lyon 1, Faculté de Médecine Lyon Est, 69732 Lyon, France
| | - Ingunn Holen
- Academic Unit of Clinical Oncology, University of Sheffield Medical School, Beech Hill Road, S10 2RX Sheffield, United Kingdom
| | - Rùben Lopez Vales
- Grup de Neuroplasticitat i Regeneració, Unitat de Fisiologia Mèdica, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Pierre Jurdic
- CNRS, UMR5242, ENS, Équipe Biologie Cellulaire et Physiopathologie Osseuse, Institut de Génomique Fonctionnelle de Lyon, UCB Lyon 1, 69007 Lyon, France
| | - Jerold Chun
- Department of Molecular Biology, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037
| | - Philippe Clézardin
- INSERM, UMR1033, UCB Lyon 1, Faculté de Médecine Lyon Est, 69732 Lyon, France
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, 565-0871 Osaka, Japan; CREST, Japan Science and Technology Agency, 102-0076 Tokyo, Japan
| | - Olivier Peyruchaud
- INSERM, UMR1033, UCB Lyon 1, Faculté de Médecine Lyon Est, 69732 Lyon, France.
| |
Collapse
|
24
|
Phospholipases of mineralization competent cells and matrix vesicles: roles in physiological and pathological mineralizations. Int J Mol Sci 2013; 14:5036-129. [PMID: 23455471 PMCID: PMC3634480 DOI: 10.3390/ijms14035036] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/24/2013] [Accepted: 01/25/2013] [Indexed: 02/08/2023] Open
Abstract
The present review aims to systematically and critically analyze the current knowledge on phospholipases and their role in physiological and pathological mineralization undertaken by mineralization competent cells. Cellular lipid metabolism plays an important role in biological mineralization. The physiological mechanisms of mineralization are likely to take place in tissues other than in bones and teeth under specific pathological conditions. For instance, vascular calcification in arteries of patients with renal failure, diabetes mellitus or atherosclerosis recapitulates the mechanisms of bone formation. Osteoporosis—a bone resorbing disease—and rheumatoid arthritis originating from the inflammation in the synovium are also affected by cellular lipid metabolism. The focus is on the lipid metabolism due to the effects of dietary lipids on bone health. These and other phenomena indicate that phospholipases may participate in bone remodelling as evidenced by their expression in smooth muscle cells, in bone forming osteoblasts, chondrocytes and in bone resorbing osteoclasts. Among various enzymes involved, phospholipases A1 or A2, phospholipase C, phospholipase D, autotaxin and sphingomyelinase are engaged in membrane lipid remodelling during early stages of mineralization and cell maturation in mineralization-competent cells. Numerous experimental evidences suggested that phospholipases exert their action at various stages of mineralization by affecting intracellular signaling and cell differentiation. The lipid metabolites—such as arachidonic acid, lysophospholipids, and sphingosine-1-phosphate are involved in cell signaling and inflammation reactions. Phospholipases are also important members of the cellular machinery engaged in matrix vesicle (MV) biogenesis and exocytosis. They may favour mineral formation inside MVs, may catalyse MV membrane breakdown necessary for the release of mineral deposits into extracellular matrix (ECM), or participate in hydrolysis of ECM. The biological functions of phospholipases are discussed from the perspective of animal and cellular knockout models, as well as disease implications, development of potent inhibitors and therapeutic interventions.
Collapse
|
25
|
Lancaster S, Mansell JP. The role of lysophosphatidic acid on human osteoblast formation, maturation and the implications for bone health and disease. ACTA ACUST UNITED AC 2013. [DOI: 10.2217/clp.12.86] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|