1
|
Pathak V, Bertelli PM, Pedrini E, Harkin K, Peixoto E, Allen LD, Mcloughlin K, Chavda ND, Hamill KJ, Guduric-Fuchs J, Inforzato A, Bottazzi B, Stitt AW, Medina RJ. Modulation of diabetes-related retinal pathophysiology by PTX3. Proc Natl Acad Sci U S A 2024; 121:e2320034121. [PMID: 39348530 PMCID: PMC11474045 DOI: 10.1073/pnas.2320034121] [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: 11/23/2023] [Accepted: 08/09/2024] [Indexed: 10/02/2024] Open
Abstract
Diabetic retinopathy (DR) is a common complication of diabetes characterized by vascular pathology and neuroinflammation. Pentraxin 3 (PTX3) is a soluble pattern recognition molecule that functions at the crossroads between innate immunity, inflammation, and tissue remodeling. DR is known to involve inflammatory pathways, although the potential relevance of PTX3 has not been explored. We found that PTX3 protein levels increased in the retina of diabetic mice. Similarly, evaluation of a publicly available transcriptomic human dataset revealed increased PTX3 expression in DR with diabetic macular edema and proliferative retinopathy, when compared to nondiabetic retinas or diabetic retinas without complications. To further understand the role of PTX3 within DR, we employed the streptozotocin-induced diabetes model in PTX3 knockout mice (PTX3KO), which were followed up for 9 mo to evaluate hallmarks of disease progression. In diabetic PTX3KO mice, we observed decreased reactive gliosis, diminished microglia activation, and reduced vasodegeneration, when compared to diabetic PTX3 wild-type littermates. The decrease in DR-associated pathological features in PTX3KO retinas translated into preserved visual function, as evidenced by improved optokinetic response, restored b-wave amplitude in electroretinograms, and attenuated neurodegeneration. We showed that PTX3 induced an inflammatory phenotype in human retinal macroglia, characterized by GFAP upregulation and increased secretion of IL6 and PAI-1. We confirmed that PTX3 was required for TNF-α-induced reactive gliosis, as PTX3KO retinal explants did not up-regulate GFAP in response to TNF-α. This study reveals a unique role for PTX3 as an enhancer of sterile inflammation in DR, which drives pathogenesis and ultimately visual impairment.
Collapse
Affiliation(s)
- Varun Pathak
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
| | - Pietro M. Bertelli
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
| | - Edoardo Pedrini
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
| | - Kevin Harkin
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
| | - Elisa Peixoto
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
| | - Lynsey-Dawn Allen
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
| | - Kiran Mcloughlin
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
| | - Natasha D. Chavda
- Department for Eye and Vision Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, LiverpoolL7 8TX, United Kingdom
| | - Kevin J. Hamill
- Department for Eye and Vision Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, LiverpoolL7 8TX, United Kingdom
| | - Jasenka Guduric-Fuchs
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
| | - Antonio Inforzato
- Laboratory of Cellular and Humoral Innate Immunity, Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Rozzano, Milan20089, Italy
- Department of Biomedical Sciences, Humanitas University, Milan20072, Italy
| | - Barbara Bottazzi
- Laboratory of Cellular and Humoral Innate Immunity, Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Rozzano, Milan20089, Italy
| | - Alan W. Stitt
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
| | - Reinhold J. Medina
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, BelfastBT9 7BL, United Kingdom
- Department for Eye and Vision Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, LiverpoolL7 8TX, United Kingdom
| |
Collapse
|
2
|
Mkhize SA, Manilall A, Mokotedi L, Gunter S, Michel FS. Involvement of pentraxin-3 in the development of hypertension but not left ventricular hypertrophy in male spontaneously hypertensive rats. Physiol Rep 2024; 12:e70086. [PMID: 39414396 PMCID: PMC11483509 DOI: 10.14814/phy2.70086] [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: 03/21/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
Hypertension drives the development of concentric left ventricular hypertrophy (LVH). However, the relative contribution of pentraxin-3 (PTX-3), a novel marker for inflammatory cardiovascular disease, in the hypertrophic response to pressure overload has not been adequately elucidated. We investigated the role of PTX-3 in the development of LVH in spontaneously hypertensive rats (SHR), untreated and treated with either captopril (an ACE inhibitor) or hydralazine (a non-specific vasodilator). Three-month-old SHR received either 20 mg/kg/day hydralazine (SHR + H, n = 6), 40 mg/kg/day captopril (SHR + C, n = 6), or plain gelatine cubes (untreated SHR, n = 7) orally for 4 months. Wistar Kyoto rats (WKY, n = 7) were used as the normotensive controls. Blood pressure (BP) was measured using the tail-cuff method. Cardiac geometry and function were determined using M-mode echocardiography. Circulating concentrations of inflammatory markers were measured in plasma by ELISA. LV fibrosis and cardiomyocyte width were assessed by histology. Relative mRNA expression of PTX-3 was determined in the LV by RT-PCR. Untreated SHR exhibited greater systolic BP and relative wall thickness (RWT) compared to WKY. Captopril and hydralazine normalized BP but only captopril reversed RWT in SHR. Circulating PTX-3 and VCAM-1 levels were elevated in untreated SHR and reduced with captopril and hydralazine. Circulating PTX-3 was positively associated with systolic BP but lacked independent relations with indices of LVH. LV relative mRNA expression of PTX-3 was similar between the groups. PTX-3 may not be involved in the development of LVH in SHR, but plausibly reflects the localized inflammatory milieu associated with hypertension.
Collapse
Affiliation(s)
- Siluleko A. Mkhize
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Ashmeetha Manilall
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Lebogang Mokotedi
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
- Department of Physiology, School of MedicineSefako Makgatho Health Sciences UniversityPretoriaSouth Africa
| | - Sule Gunter
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
- Department of Health Sciences and MedicineBond UniversityGold CoastQueenslandAustralia
| | - Frederic S. Michel
- Integrated Molecular Physiology Research Initiative, Faculty of Health Sciences, School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
3
|
Huffer A, Mao M, Ballard K, Ozdemir T. Biomimetic Hyaluronan Binding Biomaterials to Capture the Complex Regulation of Hyaluronan in Tissue Development and Function. Biomimetics (Basel) 2024; 9:499. [PMID: 39194478 DOI: 10.3390/biomimetics9080499] [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: 06/24/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Within native ECM, Hyaluronan (HA) undergoes remarkable structural remodeling through its binding receptors and proteins called hyaladherins. Hyaladherins contain a group of tandem repeat sequences, such as LINK domains, BxB7 homologous sequences, or 20-50 amino acid long short peptide sequences that have high affinity towards side chains of HA. The HA binding sequences are critical players in HA distribution and regulation within tissues and potentially attractive therapeutic targets to regulate HA synthesis and organization. While HA is a versatile and successful biopolymer, most HA-based therapeutics have major differences from a native HA molecule, such as molecular weight discrepancies, crosslinking state, and remodeling with other HA binding proteins. Recent studies showed the promise of HA binding domains being used as therapeutic biomaterials for osteoarthritic, ocular, or cardiovascular therapeutic products. However, we propose that there is a significant potential for HA binding materials to reveal the physiological functions of HA in a more realistic setting. This review is focused on giving a comprehensive overview of the connections between HA's role in the body and the potential of HA binding material applications in therapeutics and regenerative medicine. We begin with an introduction to HA then discuss HA binding molecules and the process of HA binding. Finally, we discuss HA binding materials anf the future prospects of potential HA binding biomaterials systems in the field of biomaterials and tissue engineering.
Collapse
Affiliation(s)
- Amelia Huffer
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines, Rapid City, SD 57701, USA
| | - Mingyang Mao
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines, Rapid City, SD 57701, USA
| | - Katherine Ballard
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines, Rapid City, SD 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines, Rapid City, SD 57701, USA
| |
Collapse
|
4
|
Ye X, Wang Z, Lei W, Shen M, Tang J, Xu X, Yang Y, Zhang H. Pentraxin 3: A promising therapeutic target for cardiovascular diseases. Ageing Res Rev 2024; 93:102163. [PMID: 38092307 DOI: 10.1016/j.arr.2023.102163] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/23/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023]
Abstract
Cardiovascular disease (CVD) is the primary global cause of death, and inflammation is a crucial factor in the development of CVDs. The acute phase inflammatory protein pentraxin 3 (PTX3) is a biomarker reflecting the immune response. Recent research indicates that PTX3 plays a vital role in CVDs and has been investigated as a possible biomarker for CVD in clinical trials. PTX3 is implicated in the progression of CVDs through mechanisms such as exacerbating vascular endothelial dysfunction, affecting angiogenesis, and regulating inflammation and oxidative stress. This review summarized the structure and function of PTX3, focusing on its multifaceted effects on CVDs, such as atherosclerosis, myocardial infarction, and hypertension. This may help in explaining the varying PTX3 functions and usage, as well as in utilizing target organs to manage diseases. Moreover, elucidating the opposite role of PTX3 in the cardiovascular system will demonstrate the therapeutic and predictive potential in human diseases.
Collapse
Affiliation(s)
- Xingyan Ye
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, China
| | - Wangrui Lei
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China
| | - Mingzhi Shen
- Department of General Medicine, Hainan Hospital of Chinese People's Liberation Army (PLA) General Hospital, 80 Jianglin Road, Hainan, China
| | - Jiayou Tang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, China
| | - Xuezeng Xu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, China
| | - Yang Yang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China.
| | - Huan Zhang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China.
| |
Collapse
|
5
|
Moraes ACCG, da Luz RCFV, Fernandes ALM, Barbosa MXS, de Andrade LV, Armstrong ADC, de Souza CDF, do Carmo RF. Association of PTX3 gene polymorphisms and PTX3 plasma levels with leprosy susceptibility. BMC Infect Dis 2023; 23:853. [PMID: 38053036 DOI: 10.1186/s12879-023-08862-0] [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: 06/13/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Pentraxin 3 (PTX3) is a soluble pattern recognition receptor that plays a crucial role in modulating the inflammatory response and activating the complement system. Additionally, plasma PTX3 has emerged as a potential biomarker for various infectious diseases. The aim of this study was to evaluate the association of PTX3 gene polymorphisms and PTX3 plasma levels with susceptibility to leprosy and clinical characteristics. METHODS Patients with leprosy from a hyperendemic area in the Northeast Region of Brazil were included. Healthy household contacts and healthy blood donors from the same geographical area were recruited as a control group. The rs1840680 and rs2305619 polymorphisms of PTX3 were determined by real-time PCR. Plasma levels of PTX3 were determined by ELISA. RESULTS A total of 512 individuals were included. Of these, 273 were patients diagnosed with leprosy; 53 were household contacts, and 186 were healthy blood donors. No association was observed between PTX3 polymorphisms and susceptibility to leprosy or development of leprosy reaction or physical disability. On the other hand, plasma levels of PTX3 were significantly higher in patients with leprosy when compared to household contacts (p = 0.003) or blood donors (p = 0.04). It was also observed that PTX3 levels drop significantly after multidrug therapy (p < 0.0001). CONCLUSIONS Our results suggest that PTX3 may play an important role in the pathogenesis of leprosy and point to the potential use of this molecule as an infection marker.
Collapse
Affiliation(s)
- Ana Clara Cadidé Gonzaga Moraes
- Postgraduate Program in Health and Biological Sciences, Federal University of the São Francisco Valley (UNIVASF), Pernambuco, Brazil
| | | | | | - Milena Xavier Silva Barbosa
- Postgraduate Program in Biosciences, Federal University of the São Francisco Valley (UNIVASF), Pernambuco, Brazil
| | - Lorena Viana de Andrade
- Postgraduate Program in Biosciences, Federal University of the São Francisco Valley (UNIVASF), Pernambuco, Brazil
| | - Anderson da Costa Armstrong
- College of Medicine, Federal University of the São Francisco Valley (UNIVASF), Av. José de Sá Maniçoba, s/n, Centro, Petrolina, PE, Brazil
| | - Carlos Dornels Freire de Souza
- College of Medicine, Federal University of the São Francisco Valley (UNIVASF), Av. José de Sá Maniçoba, s/n, Centro, Petrolina, PE, Brazil
| | - Rodrigo Feliciano do Carmo
- Postgraduate Program in Health and Biological Sciences, Federal University of the São Francisco Valley (UNIVASF), Pernambuco, Brazil.
- Postgraduate Program in Biosciences, Federal University of the São Francisco Valley (UNIVASF), Pernambuco, Brazil.
- College of Medicine, Federal University of the São Francisco Valley (UNIVASF), Av. José de Sá Maniçoba, s/n, Centro, Petrolina, PE, Brazil.
| |
Collapse
|
6
|
Li D, Hao Z, Nan Y, Chen Y. Role of long pentraxin PTX3 in cancer. Clin Exp Med 2023; 23:4401-4411. [PMID: 37438568 DOI: 10.1007/s10238-023-01137-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Cancer has become a leading cause of death and disease burden worldwide, closely related to rapid socioeconomic development. However, the fundamental reason is the lack of comprehensive understanding of the mechanism of cancer, accurate identification of preclinical cancer, and effective treatment of the disease. Therefore, it is particularly urgent to study specific mechanisms of cancer and develop effective prediction and treatment methods. Long Pentraxin PTX3 is a soluble pattern recognition molecule produced by various cells in inflammatory sites, which plays a role as a promoter or suppressor of cancer in multiple tumors through participating in innate immune response, neovascularization, energy metabolism, invasion, and metastasis mechanisms. Based on this, this article mainly reviews the role of PTX3 in various cancers.
Collapse
Affiliation(s)
- Duo Li
- Department of Respiratory Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an 710038, China
| | - Zhaozhao Hao
- Department of Respiratory Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an 710038, China
| | - Yandong Nan
- Department of Respiratory Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an 710038, China.
| | - Yanwei Chen
- Department of Respiratory Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an 710038, China
| |
Collapse
|
7
|
Wang Y, Chen W, Ding S, Wang W, Wang C. Pentraxins in invertebrates and vertebrates: From structure, function and evolution to clinical applications. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 149:105064. [PMID: 37734429 DOI: 10.1016/j.dci.2023.105064] [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: 05/12/2023] [Revised: 09/18/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
The immune system is divided into two broad categories, consisting of innate and adaptive immunity. As recognition and effector factors of innate immunity and regulators of adaptive immune responses, lectins are considered to be important defense chemicals against microbial pathogens, cell trafficking, immune regulation, and prevention of autoimmunity. Pentraxins, important members of animal lectins, play a significant role in protecting the body from pathogen infection and regulating inflammatory reactions. They can recognize and bind to a variety of ligands, including carbohydrates, lipids, proteins, nucleic acids and their complexes, and protect the host from pathogen invasion by activating the complement cascade and Fcγ receptor pathways. Based on the primary structure of the subunit, pentraxins are divided into short and long pentraxins. The short pentraxins are comprised of C-reactive protein (CRP) and serum amyloid P (SAP), and the most important member of the long pentraxins is pentraxin 3 (PTX3). The CRP and SAP exist in both vertebrates and invertebrates, while the PTX3 may be present only in vertebrates. The major ligands and functions of CRP, SAP and PTX3 and three activation pathways involved in the complement system are summarized in this review. Their different characteristics in various animals including humans, and their evolutionary trees are analyzed. The clinical applications of CRP, SAP and PTX3 in human are reviewed. Some questions that remain to be understood are also highlighted.
Collapse
Affiliation(s)
- Yuying Wang
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China
| | - Wei Chen
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China; Yantai Productivity Promotion Center, Yantai, 264003, People's Republic of China
| | - Shuo Ding
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China
| | - Wenjun Wang
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China
| | - Changliu Wang
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China.
| |
Collapse
|
8
|
Li Y, Zhang S, Liu J, Zhang Y, Zhang N, Cheng Q, Zhang H, Wu X. The pentraxin family in autoimmune disease. Clin Chim Acta 2023; 551:117592. [PMID: 37832905 DOI: 10.1016/j.cca.2023.117592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
The pentraxins represent a family of multifunctional proteins composed of long and short pentamers. The latter includes serum amyloid P component (SAP) and C-reactive protein (CRP) whereas the former includes neuronal PTX1 and PTX2 (NPTX1 and NPTX2, respectively), PTX3 and PTX4. These serve as a bridge between adaptive immunity and innate immunity and a link between inflammation and immunity. Similarities and differences between long and short pentamers are examined and their roles in autoimmune disease are discussed. Increased CRP and PTX3 could indicate the activity of rheumatoid arthritis, systemic lupus erythematosus or other autoimmune diseases. Mechanistically, CRP and PTX3 may predict target organ injury, regulate bone metabolic immunity and maintain homeostasis as well as participate in vascular endothelial remodeling. Interestingly, PTX3 is pleiotropic, being involved in inflammation and tissue repair. Given the therapeutic potential of PTX3 and CRP, targeting these factors to exert a beneficial effect is the focus of research efforts. Unfortunately, studies on NPTX1, NPTX2, PTX4 and SAP are scarce and more research is clearly needed to elaborate their potential roles in autoimmune disease.
Collapse
Affiliation(s)
- Yongzhen Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Shouzan Zhang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, PR China
| | - Jingqi Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yudi Zhang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, PR China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China.
| | - Xiaochuan Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
9
|
Massimino AM, Colella FE, Bottazzi B, Inforzato A. Structural insights into the biological functions of the long pentraxin PTX3. Front Immunol 2023; 14:1274634. [PMID: 37885881 PMCID: PMC10598717 DOI: 10.3389/fimmu.2023.1274634] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Soluble pattern recognition molecules (PRMs) are a heterogenous group of proteins that recognize pathogen- and danger-associated molecular patterns (PAMPs and DAMPs, respectively), and cooperate with cell-borne receptors in the orchestration of innate and adaptive immune responses to pathogenic insults and tissue damage. Amongst soluble PRMs, pentraxins are a family of highly conserved proteins with distinctive structural features. Originally identified in the early 1990s as an early inflammatory gene, PTX3 is the prototype of long pentraxins. Unlike the short pentraxin C reactive protein (CRP), whose expression is mostly confined to the liver, PTX3 is made by several immune and non-immune cells at sites of infection and inflammation, where it intercepts fundamental aspects of infection immunity, inflammation, and tissue remodeling. Of note, PTX3 cross talks to components of the complement system to control cancer-related inflammation and disposal of pathogens. Also, it is an essential component of inflammatory extracellular matrices (ECMs) through crosslinking of hyaluronic acid and turn-over of provisional fibrin networks that assemble at sites of tissue injury. This functional diversity is mediated by unique structural characteristics whose fine details have been unveiled only recently. Here, we revisit the structure/function relationships of this long pentraxin in light of the most recent advances in its structural biology, with a focus on the interplay with complement and the emerging roles as a component of the ECM. Differences to and similarities with the short pentraxins are highlighted and discussed.
Collapse
Affiliation(s)
| | | | - Barbara Bottazzi
- Laboratory of Cellular and Humoral Innate Immunity, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Antonio Inforzato
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Laboratory of Cellular and Humoral Innate Immunity, IRCCS Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
10
|
Panchalingam S, Kasivelu G, Jayaraman M, Kumar R, Kalimuthu S, Jeyaraman J. Differential gene expression analysis combined with molecular dynamics simulation study to elucidate the novel potential biomarker involved in pulmonary TB. Microb Pathog 2023; 182:106266. [PMID: 37482113 DOI: 10.1016/j.micpath.2023.106266] [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: 05/31/2023] [Revised: 06/21/2023] [Accepted: 07/21/2023] [Indexed: 07/25/2023]
Abstract
Tuberculosis (TB) is a lethal multisystem disease that attacks the lungs' first line of defense. A substantial threat to public health and a primary cause of death is pulmonary TB. This study aimed to identify and investigate the probable differentially expressed genes (DEGs) primarily involved in Pulmonary TB. Accordingly, three independent gene expression data sets, numbered GSE139825, GSE139871, and GSE54992, were utilized for this purpose. The identified DEGs were used for bioinformatics-based analysis, including physical gene interaction, Gene Ontology (GO), network analysis and pathway studies using the Kyoto Encyclopedia of Genes and Genomes pathway (KEGG). The computational analysis predicted that TNFAIP6 is the significant DEG in the gene expression profiling of TB datasets. According to gene ontology analysis, TNFAIP6 is also essential in injury and inflammation. Further, TNFA1P6 is strongly linked to arsenic poisoning, evident from the results of NetworkAnalyst, a comprehensive and interactive platform for gene expression profiling via network visual analytics. As a result, the TNFAIP6 gene was ultimately chosen as a candidate DEG and subsequently employed for in silico structural characterization studies. The tertiary structure of TNFAIP6 was modelled using the ROBETTA server, followed by validation with SAVES and ProSA webserver. Additionally, structural dynamic studies, including molecular dynamics simulation (MDS) and essential dynamics analysis, including principal component (PC) based free energy landscape (FEL) analysis, was used for checking the stability of TNFAIP6 models. The dynamics result established the structural rigidity of modelled TNFAIP6 through RMSD, RMSF and RoG results. The FEL analysis revealed the restricted conformational flexibility of TNFAIP6 by displaying a single minimum energy basin in the contour plot. The comprehensive computational analysis established that TNFAIP6 could serve as a viable biomarker to assess the severity of pulmonary TB.
Collapse
Affiliation(s)
- Santhiya Panchalingam
- Centre for Ocean Research, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, 600 119, Tamil Nadu, India
| | - Govindaraju Kasivelu
- Centre for Ocean Research, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, 600 119, Tamil Nadu, India.
| | - Manikandan Jayaraman
- Structural Biology and Biocomputing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, 630 004, Tamil Nadu, India
| | - Rajalakshmi Kumar
- Mahatma Gandhi Medical Advanced Research Institute, Sri Balaji Vidyapeeth (Deemed to Be University), Pillayarkuppam, Puducherry, 607 402, India
| | | | - Jeyakanthan Jeyaraman
- Structural Biology and Biocomputing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, 630 004, Tamil Nadu, India.
| |
Collapse
|
11
|
Zhang H, Wang R, Wang Z, Wu W, Zhang N, Zhang L, Hu J, Luo P, Zhang J, Liu Z, Feng S, Peng Y, Liu Z, Cheng Q. Molecular insight into pentraxin-3: Update advances in innate immunity, inflammation, tissue remodeling, diseases, and drug role. Biomed Pharmacother 2022; 156:113783. [PMID: 36240615 DOI: 10.1016/j.biopha.2022.113783] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
Abstract
Pentraxin-3 (PTX3) is the prototype of the long pentraxin subfamily, an acute-phase protein consisting of a C-terminal pentraxin domain and a unique N-terminal domain. PTX3 was initially isolated from human umbilical vein endothelial cells and human FS-4 fibroblasts. It was subsequently found to be also produced by synoviocytes, chondrocytes, osteoblasts, smooth muscle cells, myeloid dendritic cells, epithelial cells, and tumor cells. Various modulatory factors, such as miRNAs, cytokines, drugs, and hypoxic conditions, could regulate the expression level of PTX3. PTX3 is essential in regulating innate immunity, inflammation, angiogenesis, and tissue remodeling. Besides, PTX3 may play dual (pro-tumor and anti-tumor) roles in oncogenesis. PTX3 is involved in the occurrence and development of many non-cancerous diseases, including COVID-19, and might be a potential biomarker indicating the prognosis, activity,and severity of diseases. In this review, we summarize and discuss the potential roles of PTX3 in the oncogenesis and pathogenesis of non-cancerous diseases and potential targeted therapies based on PTX3.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, China; Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, China
| | - Ruixuan Wang
- Department of Oncology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, China
| | - Nan Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, China; One-third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Longbo Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, China; Department of Neurosurgery, and Department of Cellular & Molecular Physiology,Yale University School of Medicine, USA; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, China
| | - Jason Hu
- Department of Neonatology, Yale University School of Medicine, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, China
| | - Songshan Feng
- Department of Neurosurgery, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, China
| | - Yun Peng
- Department of Geriatrics, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, China
| | - Zhengzheng Liu
- Department of Oncology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, China.
| |
Collapse
|
12
|
PTX3 structure determination using a hybrid cryoelectron microscopy and AlphaFold approach offers insights into ligand binding and complement activation. Proc Natl Acad Sci U S A 2022; 119:e2208144119. [PMID: 35939690 PMCID: PMC9388099 DOI: 10.1073/pnas.2208144119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pattern recognition molecules (PRMs) form an important part of innate immunity, where they facilitate the response to infections and damage by triggering processes such as inflammation. The pentraxin family of soluble PRMs comprises long and short pentraxins, with the former containing unique N-terminal regions unrelated to other proteins or each other. No complete high-resolution structural information exists about long pentraxins, unlike the short pentraxins, where there is an abundance of both X-ray and cryoelectron microscopy (cryo-EM)-derived structures. This study presents a high-resolution structure of the prototypical long pentraxin, PTX3. Cryo-EM yielded a 2.5-Å map of the C-terminal pentraxin domains that revealed a radically different quaternary structure compared to other pentraxins, comprising a glycosylated D4 symmetrical octameric complex stabilized by an extensive disulfide network. The cryo-EM map indicated α-helices that extended N terminal of the pentraxin domains that were not fully resolved. AlphaFold was used to predict the remaining N-terminal structure of the octameric PTX3 complex, revealing two long tetrameric coiled coils with two hinge regions, which was validated using classification of cryo-EM two-dimensional averages. The resulting hybrid cryo-EM/AlphaFold structure allowed mapping of ligand binding sites, such as C1q and fibroblast growth factor-2, as well as rationalization of previous biochemical data. Given the relevance of PTX3 in conditions ranging from COVID-19 prognosis, cancer progression, and female infertility, this structure could be used to inform the understanding and rational design of therapies for these disorders and processes.
Collapse
|
13
|
Spinelli FM, Rosales P, Pluda S, Vitale DL, Icardi A, Guarise C, Reszegi A, Kovalszky I, García M, Sevic I, Galesso D, Alaniz L. The effects of sulfated hyaluronan in breast, lung and colorectal carcinoma and monocytes/macrophages cells: Its role in angiogenesis and tumor progression. IUBMB Life 2022; 74:927-942. [PMID: 35218610 DOI: 10.1002/iub.2604] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 11/11/2022]
Abstract
Hyaluronan (HA) is a component of the extracellular matrix (ECM) it is the main non-sulfated glycosaminoglycan able to modulate cell behavior in the healthy and tumor context. Sulfated hyaluronan (sHA) is a biomaterial derived from chemical modifications of HA, since this molecule is not naturally sulfated. The HA sulfation modifies several properties of the native molecule, acquiring antitumor properties in different cancers. In this study, we evaluated the action of sHA of ~30-60 kDa with different degrees of sulfation (0.7 sHA1 and 2.5 sHA3) on tumor cells of a breast, lung, and colorectal cancer model and its action on other cells of the tumor microenvironment, such as endothelial and monocytes/macrophage cells. Our data showed that in breast and lung tumor cells, sHA3 is able to modulate cell viability, cytotoxicity, and proliferation, but no effects were observed on colorectal cancer cells. In 3D cultures of breast and lung cancer cells, sHA3 diminished the size of the tumorsphere and modulated total HA levels. In these tumor models, treatment of monocytes/macrophages with sHA3 showed a downregulation of the expression of angiogenic factors. We also observed a decrease in endothelial cell migration and modulation of the hyaluronan-binding protein TSG-6. In the breast in vivo xenograft model, monocytes/macrophages preincubated with sHA1 or sHA3 decreased tumor vasculature, TSG-6 and HA levels. Besides, in silico analysis showed an association of TSG-6, HAS2, and IL-8 with biological processes implicated in the progression of the tumor. Taken together, our data indicate that sHA in a breast and lung tumor context is able to induce an antiangiogenic action on tumor cells as well as in monocytes/macrophages (Mo/MØ) by modulation of endothelial migration, angiogenic factors, and vessel formation.
Collapse
Affiliation(s)
- Fiorella M Spinelli
- Laboratorio de Microambiente Tumoral, CIBA, UNNOBA/CIT NOBA (UNNOBA-UNSADA-CONICET), Jorge Newbery 261, Junín, Argentina
| | - Paolo Rosales
- Laboratorio de Microambiente Tumoral, CIBA, UNNOBA/CIT NOBA (UNNOBA-UNSADA-CONICET), Jorge Newbery 261, Junín, Argentina
| | | | - Daiana L Vitale
- Laboratorio de Microambiente Tumoral, CIBA, UNNOBA/CIT NOBA (UNNOBA-UNSADA-CONICET), Jorge Newbery 261, Junín, Argentina
| | - Antonella Icardi
- Laboratorio de Microambiente Tumoral, CIBA, UNNOBA/CIT NOBA (UNNOBA-UNSADA-CONICET), Jorge Newbery 261, Junín, Argentina
| | | | - Andrea Reszegi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ilona Kovalszky
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Mariana García
- Laboratorio de Terapia Génica, IIMT - CONICET, Universidad Austral, Derqui-Pilar, Argentina
| | - Ina Sevic
- Laboratorio de Microambiente Tumoral, CIBA, UNNOBA/CIT NOBA (UNNOBA-UNSADA-CONICET), Jorge Newbery 261, Junín, Argentina
| | | | - Laura Alaniz
- Laboratorio de Microambiente Tumoral, CIBA, UNNOBA/CIT NOBA (UNNOBA-UNSADA-CONICET), Jorge Newbery 261, Junín, Argentina
| |
Collapse
|
14
|
Hsiao Y, Chi J, Li C, Chen L, Chen Y, Liang H, Lo Y, Hong J, Chuu C, Hung L, Du J, Chang W, Wang J. Disruption of the pentraxin 3/CD44 interaction as an efficient therapy for triple-negative breast cancers. Clin Transl Med 2022; 12:e724. [PMID: 35090088 PMCID: PMC8797470 DOI: 10.1002/ctm2.724] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/29/2022] Open
Abstract
Due to the heterogeneity and high frequency of genome mutations in cancer cells, targeting vital protumour factors found in stromal cells in the tumour microenvironment may represent an ideal strategy in cancer therapy. However, the regulation and mechanisms of potential targetable therapeutic candidates need to be investigated. An in vivo study demonstrated that loss of pentraxin 3 (PTX3) in stromal cells significantly decreased the metastasis and growth of cancer cells. Clinically, our results indicate that stromal PTX3 expression correlates with adverse prognostic features and is associated with worse survival outcomes in triple-negative breast cancer (TNBC). We also found that transforming growth factor beta 1 (TGF-β1) induces PTX3 expression by activating the transcription factor CCAAT/enhancer binding protein delta (CEBPD) in stromal fibroblasts. Following PTX3 stimulation, CD44, a PTX3 receptor, activates the downstream ERK1/2, AKT and NF-κB pathways to specifically contribute to the metastasis/invasion and stemness of TNBC MDA-MB-231 cells. Two types of PTX3 inhibitors were developed to disrupt the PTX3/CD44 interaction and they showed a significant effect on attenuating growth and restricting the metastasis/invasion of MDA-MB-231 cells, suggesting that targeting the PTX3/CD44 interaction could be a new strategy for future TNBC therapies.
Collapse
Affiliation(s)
- Yu‐Wei Hsiao
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Jhih‐Ying Chi
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Chien‐Feng Li
- Department of PathologyChi‐Mei Medical CenterTainanTaiwan R. O. C.
| | - Lei‐Yi Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Yi‐Ting Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Hsin‐Yin Liang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Yu‐Chih Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Jhen‐Yi Hong
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Chin‐Pin Chuu
- Institute of Cellular and System MedicineNational Health Research InstitutesMiaoli CountyTaiwan R. O. C.
| | - Liang‐Yi Hung
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Jyun‐Yi Du
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Wen‐Chang Chang
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan R. O. C.
| | - Ju‐Ming Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan R. O. C.
- International Research Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan R. O. C.
- Department of Physiology, College of MedicineNational Cheng Kung UniversityTainanTaiwan R. O. C.
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan R. O. C.
| |
Collapse
|
15
|
Wesley UV, Sutton I, Clark PA, Cunningham K, Larrain C, Kuo JS, Dempsey RJ. Enhanced expression of pentraxin-3 in glioblastoma cells correlates with increased invasion and IL8-VEGF signaling axis. Brain Res 2021; 1776:147752. [PMID: 34906547 DOI: 10.1016/j.brainres.2021.147752] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/13/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023]
Abstract
Glioblastoma (GB) is highly invasive and resistant to multimodal treatment partly due to distorted vasculature and exacerbated inflammation. The aggressiveness of brain tumors may be attributed to the dysregulated release of angiogenic and inflammatory factors. The glycoprotein pentraxin-3 (PTX3) is correlated with the severity of some cancers. However, the mechanism responsible for the invasive oncogenic role of PTX3 in GB malignancy remains unclear. In this study, we examined the role of PTX3 in GB growth, angiogenesis, and invasion using in vitro and in vivo GB models, proteomic profiling, molecular and biochemical approaches. Under in vitro conditions, PTX3 over-expression in U87 cells correlated with cell cycle progression, increased migratory potential, and proliferation under hypoxic conditions. Conditioned media containing PTX3 enhanced the angiogenic potential of endothelial cells. While silencing of PTX3 by siRNA decreased the proliferation, migration, and angiogenic potential of U87 cells in vitro. Importantly, PTX3 over-expression increased tumor growth, angiogenesis, and invasion in an orthotopic mouse model. Higher levels of PTX3 in these tumors were associated with the upregulation of inflammatory and angiogenic markers including interleukin-8 (IL-8) and vascular endothelial growth factor (VEGF), but decreased levels of thrombospondin-1, an anti-angiogenic factor. Mechanistically, exogenous production of PTX3 triggered an IKK/NFκB signaling pathway that enhances the expression of the motility genes AHGEF7 and Rac1. Taken together, PTX3 expression is dysregulated in GB. PTX3 may augment invasion through enhanced angiogenesis in the GB microenvironment through the IL8-VEGF axis. Thus, PTX3 may represent a potential therapeutic target to mitigate the aggressive behavior of gliomas.
Collapse
Affiliation(s)
- Umadevi V Wesley
- Department of Neurosurgery, University of Wisconsin, Madison, WI 53792, United States.
| | - Ian Sutton
- Department of Neurosurgery, University of Wisconsin, Madison, WI 53792, United States
| | - Paul A Clark
- Department of Neurosurgery, University of Wisconsin, Madison, WI 53792, United States; Department of Human Oncology, University of Wisconsin, Madison, WI 53792, United States
| | - Katelin Cunningham
- Department of Neurosurgery, University of Wisconsin, Madison, WI 53792, United States
| | - Carolina Larrain
- Department of Neurosurgery, University of Wisconsin, Madison, WI 53792, United States
| | - John S Kuo
- Department of Neurosurgery, University of Wisconsin, Madison, WI 53792, United States; Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, United States; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, TAIWAN
| | - Robert J Dempsey
- Department of Neurosurgery, University of Wisconsin, Madison, WI 53792, United States.
| |
Collapse
|
16
|
Koussih L, Atoui S, Tliba O, Gounni AS. New Insights on the Role of pentraxin-3 in Allergic Asthma. FRONTIERS IN ALLERGY 2021; 2:678023. [PMID: 35387000 PMCID: PMC8974764 DOI: 10.3389/falgy.2021.678023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022] Open
Abstract
Pentraxins are soluble pattern recognition receptors that play a major role in regulating innate immune responses. Through their interaction with complement components, Fcγ receptors, and different microbial moieties, Pentraxins cause an amplification of the inflammatory response. Pentraxin-3 is of particular interest since it was identified as a biomarker for several immune-pathological diseases. In allergic asthma, pentraxin-3 is produced by immune and structural cells and is up-regulated by pro-asthmatic cytokines such as TNFα and IL-1β. Strikingly, some recent experimental evidence demonstrated a protective role of pentraxin-3 in chronic airway inflammatory diseases such as allergic asthma. Indeed, reduced pentraxin-3 levels have been associated with neutrophilic inflammation, Th17 immune response, insensitivity to standard therapeutics and a severe form of the disease. In this review, we will summarize the current knowledge of the role of pentraxin-3 in innate immune response and discuss the protective role of pentraxin-3 in allergic asthma.
Collapse
Affiliation(s)
- Latifa Koussih
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Department des Sciences Experimentales, Universite de Saint-Boniface, Winnipeg, MB, Canada
| | - Samira Atoui
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Omar Tliba
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| | - Abdelilah S. Gounni
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- *Correspondence: Abdelilah S. Gounni
| |
Collapse
|
17
|
Greggi C, Cariati I, Onorato F, Iundusi R, Scimeca M, Tarantino U. PTX3 Effects on Osteogenic Differentiation in Osteoporosis: An In Vitro Study. Int J Mol Sci 2021; 22:5944. [PMID: 34073015 PMCID: PMC8198053 DOI: 10.3390/ijms22115944] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022] Open
Abstract
Pentraxin 3 (PTX3) is a glycoprotein belonging to the humoral arm of innate immunity that participates in the body's defence mechanisms against infectious diseases. It has recently been defined as a multifunctional protein, given its involvement in numerous physiological and pathological processes, as well as in the pathogenesis of age-related diseases such as osteoporosis. Based on this evidence, the aim of our study was to investigate the possible role of PTX3 in both the osteoblastic differentiation and calcification process: to this end, primary osteoblast cultures from control and osteoporotic patients were incubated with human recombinant PTX3 (hrPTX3) for 72 h. Standard osteinduction treatment, consisting of β-glycerophosphate, dexamethasone and ascorbic acid, was used as control. Our results showed that treatment with hrPTX3, as well as with the osteogenic cocktail, induced cell differentiation towards the osteoblastic lineage. We also observed that the treatment not only promoted an increase in cell proliferation, but also the formation of calcification-like structures, especially in primary cultures from osteoporotic patients. In conclusion, the results reported here suggest the involvement of PTX3 in osteogenic differentiation, highlighting its osteoinductive capacity, like the standard osteoinduction treatment. Therefore, this study opens new and exciting perspectives about the possible role of PTX3 as biomarker and therapeutic agent for osteoporosis.
Collapse
Affiliation(s)
- Chiara Greggi
- Ph.D. in Medical-Surgical Biotechnologies and Translational Medicine, “Tor Vergata” University of Rome, via Montpellier 1, 00133 Rome, Italy; (C.G.); (I.C.)
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, via Montpellier 1, 00133 Rome, Italy
| | - Ida Cariati
- Ph.D. in Medical-Surgical Biotechnologies and Translational Medicine, “Tor Vergata” University of Rome, via Montpellier 1, 00133 Rome, Italy; (C.G.); (I.C.)
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, via Montpellier 1, 00133 Rome, Italy
| | - Federica Onorato
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, viale Oxford 81, 00133 Rome, Italy; (F.O.); (R.I.)
| | - Riccardo Iundusi
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, viale Oxford 81, 00133 Rome, Italy; (F.O.); (R.I.)
| | - Manuel Scimeca
- Department of Biomedicine and Prevention, “Tor Vergata” University of Rome, via Montpellier 1, 00133 Rome, Italy;
| | - Umberto Tarantino
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, via Montpellier 1, 00133 Rome, Italy
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, viale Oxford 81, 00133 Rome, Italy; (F.O.); (R.I.)
| |
Collapse
|
18
|
Liu Y, Li P. Circulating pentraxin-3 and preeclampsia: a meta-analysis of 17 case-control studies. J Matern Fetal Neonatal Med 2019; 34:3669-3677. [PMID: 31744359 DOI: 10.1080/14767058.2019.1689560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Change of circulating pentraxin-3 (PTX-3), a novel marker of inflammation, has been observed in women with preeclampsia (PE). However, results of previous studies were inconsistent. We performed a meta-analysis to evaluate the difference of circulating PTX-3 between women with PE and normal pregnancies.Methods: Case-control studies comparing circulating PTX-3 level between women with PE and normal pregnancies were identified via search of PubMed and Embase databases according to a predefined search strategy and inclusion criteria by two independent authors. Meta-analysis was performed with a random-effect model to incorporate heterogeneity.Results: Seventeen studies including 814 women with PE and 949 women with normal pregnancy were included. Results showed that women with PE had significantly higher circulating PTX-3 at diagnosis as compared to women with normal pregnancy (standardized mean difference [SMD]: = 1.74, 95% CI: 1.20-2.29, p < .001; I2 = 94%). The results were consistent regardless of study characteristics including study location, maternal age, sample size, early or late onset of PE, blood sample for PTX-3 measurement, or NOS quality scores. Moreover, higher circulating PTX-3 was also observed before the diagnosis of PE (SMD = 0.65, 95% CI: 0.02-1.29, p = .04; I2 = 87%).Conclusion: Women with PE have higher circulating PTX-3 than women with normal pregnancy. The elevated PTX-3 could be observed before the clinical onset of PE. Future studies are needed to determine whether PTX-3 is an active molecular in the pathogenesis of PE.
Collapse
Affiliation(s)
- Yun Liu
- Department of Obstetrics, Nankai Hospital, Tianjin, China
| | - Ping Li
- Department of Obstetrics, Nankai Hospital, Tianjin, China
| |
Collapse
|
19
|
Wang S, Kim J, Lee C, Oh D, Han J, Kim TJ, Kim SW, Seo YS, Oh SH, Jung Y. Tumor necrosis factor-inducible gene 6 reprograms hepatic stellate cells into stem-like cells, which ameliorates liver damage in mouse. Biomaterials 2019; 219:119375. [DOI: 10.1016/j.biomaterials.2019.119375] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/19/2019] [Accepted: 07/20/2019] [Indexed: 12/12/2022]
|
20
|
Doni A, Stravalaci M, Inforzato A, Magrini E, Mantovani A, Garlanda C, Bottazzi B. The Long Pentraxin PTX3 as a Link Between Innate Immunity, Tissue Remodeling, and Cancer. Front Immunol 2019; 10:712. [PMID: 31019517 PMCID: PMC6459138 DOI: 10.3389/fimmu.2019.00712] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/15/2019] [Indexed: 12/20/2022] Open
Abstract
The innate immune system comprises a cellular and a humoral arm. Humoral pattern recognition molecules include complement components, collectins, ficolins, and pentraxins. These molecules are involved in innate immune responses by recognizing microbial moieties and damaged tissues, activating complement, exerting opsonic activity and facilitating phagocytosis, and regulating inflammation. The long pentraxin PTX3 is a prototypic humoral pattern recognition molecule that, in addition to providing defense against infectious agents, plays several functions in tissue repair and regulation of cancer-related inflammation. Characterization of the PTX3 molecular structure and biochemical properties, and insights into its interactome and multiple roles in tissue damage and remodeling support the view that microbial and matrix recognition are evolutionarily conserved functions of humoral innate immunity molecules.
Collapse
Affiliation(s)
- Andrea Doni
- Humanitas Clinical and Research Institute-IRCCS, Milan, Italy
| | - Matteo Stravalaci
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Antonio Inforzato
- Humanitas Clinical and Research Institute-IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Elena Magrini
- Humanitas Clinical and Research Institute-IRCCS, Milan, Italy
| | - Alberto Mantovani
- Humanitas Clinical and Research Institute-IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Cecilia Garlanda
- Humanitas Clinical and Research Institute-IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | |
Collapse
|
21
|
Kim Y, Park JS, Park HJ, Kim MK, Kim YI, Bae SK, Kim HJ, Jeong CH, Bae MK. Pentraxin 3 Modulates the Inflammatory Response in Human Dental Pulp Cells. J Endod 2019; 44:1826-1831. [PMID: 30477668 DOI: 10.1016/j.joen.2018.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/17/2018] [Accepted: 08/12/2018] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Pentraxin 3 (PTX3) has been suggested as a novel inflammatory biomarker in inflammation-associated diseases. The aim of this study was to examine the role of PTX3 in the inflammatory response of human dental pulp cells (HDPCs). METHODS HDPCs were treated with tumor necrosis factor alpha (TNF-α), and total RNA and protein were extracted. PTX3 messenger RNA and protein expression levels were analyzed using reverse transcription polymerase chain reaction and Western blotting, respectively. For PTX3 knockdown, HDPCs were transfected with a small interfering RNA against human PTX3. Macrophage chemotaxis after PTX3 silencing in HDPCs was assessed by transwell migration assays. RESULTS TNF-α increased PTX3 messenger RNA and protein levels in HDPCs. TNF-α-induced PTX3 expression was mediated by extracellular signal-regulated kinase 1/2 and nuclear factor kappa B. PTX3 knockdown decreased the expression levels of interleukin 6, interleukin 8, and monocyte chemoattractant protein 1 after stimulation with TNF-α in HDPCs. Moreover, PTX3 silencing in HDPCs significantly decreased the chemotactic migration of macrophages. CONCLUSIONS Our findings indicate PTX3 plays a critical role in the regulation of pulp inflammatory processes and reveal its underlying molecular mechanism.
Collapse
Affiliation(s)
- Yeon Kim
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Jin-Sung Park
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Hyun-Joo Park
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Mi-Kyoung Kim
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Yong-Il Kim
- Department of Orthodontics, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Soo-Kyung Bae
- Department of Dental Pharmacology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Hyung Joon Kim
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu, South Korea
| | - Moon-Kyoung Bae
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, South Korea.
| |
Collapse
|
22
|
Barros RG, Lima PF, Soares ACS, Sanches L, Price CA, Buratini J. Fibroblast growth factor 2 regulates cumulus differentiation under the control of the oocyte. J Assist Reprod Genet 2019; 36:905-913. [PMID: 30887159 DOI: 10.1007/s10815-019-01436-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/07/2019] [Indexed: 12/30/2022] Open
Abstract
PURPOSE We first assessed regulation of FGF2 expression in cumulus cells by FSH and oocyte-secreted factors during in vitro maturation (IVM). Then, we tested the hypothesis that FGF2 regulates meiotic progression, cumulus expansion, and apoptosis in cumulus-oocyte complexes (COC) undergoing IVM. METHODS In vitro maturation of bovine COC was utilized as a model to assess regulation of FGF2 expression by FSH and oocyte-secreted factors (via microsurgical removal of the oocyte), as well as effects of graded doses of FGF2 on meiotic progression, degree of cumulus expansion, dissociation of cumulus cells, and cumulus cells apoptosis. Expression of genes regulating functional endpoints altered by FGF2 treatment was assessed in cumulus cells by real-time PCR. Cultures were replicated 4-5 times and effects of treatments were tested by ANOVA. RESULTS FGF2 mRNA expression was increased by FSH and oocyte-secreted factors during IVM. Addition of FGF2 to the IVM medium advanced meiosis resumption, decreased the ease with which cumulus cells were dissociated, and inhibited cumulus cells apoptosis. Decreased cumulus dissociation was accompanied by decreased expression of TNFAIP6. CONCLUSIONS This is the first study showing that FGF2 expression is regulated by the oocyte in cumulus cells. Moreover, we report novel effects of FGF2 on cumulus cell survival and extracellular matrix (ECM) quality during IVM that may favor acquisition of developmental competence and suggest physiological roles during the final steps of COC differentiation.
Collapse
Affiliation(s)
- Rodrigo G Barros
- Departamento de Fisiologia, Instituto de Biociências, Universidade Estadual Paulista, Rubião Junior, Botucatu, São Paulo, 18618-970, Brazil.
| | - Paula F Lima
- Departamento de Fisiologia, Instituto de Biociências, Universidade Estadual Paulista, Rubião Junior, Botucatu, São Paulo, 18618-970, Brazil
| | - Ana Caroline S Soares
- Departamento de Fisiologia, Instituto de Biociências, Universidade Estadual Paulista, Rubião Junior, Botucatu, São Paulo, 18618-970, Brazil
| | - Lorena Sanches
- Departamento de Fisiologia, Instituto de Biociências, Universidade Estadual Paulista, Rubião Junior, Botucatu, São Paulo, 18618-970, Brazil
| | - Christopher A Price
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, J2S 7C6, Canada
| | - José Buratini
- Departamento de Fisiologia, Instituto de Biociências, Universidade Estadual Paulista, Rubião Junior, Botucatu, São Paulo, 18618-970, Brazil
| |
Collapse
|
23
|
Bonacina F, Moregola A, Porte R, Baragetti A, Bonavita E, Salatin A, Grigore L, Pellegatta F, Molgora M, Sironi M, Barbati E, Mantovani A, Bottazzi B, Catapano AL, Garlanda C, Norata GD. Pentraxin 3 deficiency protects from the metabolic inflammation associated to diet-induced obesity. Cardiovasc Res 2019; 115:1861-1872. [DOI: 10.1093/cvr/cvz068] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/23/2019] [Accepted: 03/11/2019] [Indexed: 01/18/2023] Open
Abstract
Abstract
Aims
Low-grade chronic inflammation characterizes obesity and metabolic syndrome. Here, we aim at investigating the impact of the acute-phase protein long pentraxin 3 (PTX3) on the immune-inflammatory response occurring during diet-induced obesity.
Methods and results
PTX3 deficiency in mice fed a high-fat diet for 20 weeks protects from weight gain and adipose tissue deposition in visceral and subcutaneous depots. This effect is not related to changes in glucose homeostasis and lipid metabolism but is associated with an improved immune cell phenotype in the adipose tissue of Ptx3 deficient animals, which is characterized by M2-macrophages polarization and increased angiogenesis. These findings are recapitulated in humans where carriers of a PTX3 haplotype (PTX3 h2/h2 haplotype), resulting in lower PTX3 plasma levels, presented with a reduced prevalence of obesity and decreased abdominal adiposity compared with non-carriers.
Conclusion
Our results support a critical role for PTX3 in the onset of obesity by promoting inflammation and limiting adipose tissue vascularization and delineate PTX3 targeting as a valuable strategy for the treatment of adipose tissue-associated inflammatory response.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, Milan, Italy
| | - Annalisa Moregola
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, Milan, Italy
| | - Rémi Porte
- IRCC Humanitas Clinical and Research Center, Rozzano, Italy
| | - Andrea Baragetti
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, Milan, Italy
- Centro SISA per lo Studio dell’Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, Italy
| | - Eduardo Bonavita
- Cancer Inflammation and Immunity Group, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Alice Salatin
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, Milan, Italy
| | - Liliana Grigore
- Centro SISA per lo Studio dell’Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, Italy
- IRCSS Multimedica, Milan, Italy
| | - Fabio Pellegatta
- Centro SISA per lo Studio dell’Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, Italy
- IRCSS Multimedica, Milan, Italy
| | | | - Marina Sironi
- IRCC Humanitas Clinical and Research Center, Rozzano, Italy
| | - Elisa Barbati
- IRCC Humanitas Clinical and Research Center, Rozzano, Italy
| | - Alberto Mantovani
- IRCC Humanitas Clinical and Research Center, Rozzano, Italy
- Humanitas University Rozzano, Italy
| | | | - Alberico Luigi Catapano
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, Milan, Italy
- IRCSS Multimedica, Milan, Italy
| | - Cecilia Garlanda
- IRCC Humanitas Clinical and Research Center, Rozzano, Italy
- Humanitas University Rozzano, Italy
| | - Giuseppe Danilo Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, Milan, Italy
- Centro SISA per lo Studio dell’Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, Italy
| |
Collapse
|
24
|
Abstract
Pentraxin 3 (PTX3) is involved in vascular inflammation and endothelial dysfunction through various mechanisms. Until now, most studies confirmed an important link between PTX3 and endothelial dysfunction and identified several pathogenetic pathways. PTX3 modulates inflammatory cells, thus stimulating vascular inflammation. Within endothelial cells, it decreases nitric oxide (NO) synthesis, inhibits cell proliferation and alters their functions. PTX3 blocks the effect of fibroblast growth factor 2 (FGF2) by making a molecular complex with these molecules inactivating them. However, there are substances like the tumor necrosis factor-inducible gene 6 protein (TSG-6) that block the PTX3-FGF2 interaction. Interacting with P-selectin, it promotes vascular inflammatory response and endothelial dysfunction. PTX3 also increases the matrix metalloproteinases synthesis directly or by blocking NO synthesis. From a clinical point of view, PTX3 positively correlates with arterial hypertension, flow mediated dilation and, with intima media thickness. Therefore, the involvement of PTX3 in the pathogenesis and evaluation of endothelial dysfunction is clear, and it may become a biomarker in this direction, but further studies are needed to determine its reliability in this direction. Last but not least, PTX3 could become an effective therapeutic target for preventing this dysfunction, but further research needs to be conducted.
Collapse
|
25
|
Fossati G, Pozzi D, Canzi A, Mirabella F, Valentino S, Morini R, Ghirardini E, Filipello F, Moretti M, Gotti C, Annis DS, Mosher DF, Garlanda C, Bottazzi B, Taraboletti G, Mantovani A, Matteoli M, Menna E. Pentraxin 3 regulates synaptic function by inducing AMPA receptor clustering via ECM remodeling and β1-integrin. EMBO J 2018; 38:embj.201899529. [PMID: 30396995 PMCID: PMC6315291 DOI: 10.15252/embj.201899529] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/12/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022] Open
Abstract
Control of synapse number and function in the developing central nervous system is critical to the formation of neural circuits. Astrocytes play a key role in this process by releasing factors that promote the formation of excitatory synapses. Astrocyte‐secreted thrombospondins (TSPs) induce the formation of structural synapses, which however remain post‐synaptically silent, suggesting that completion of early synaptogenesis may require a two‐step mechanism. Here, we show that the humoral innate immune molecule Pentraxin 3 (PTX3) is expressed in the developing rodent brain. PTX3 plays a key role in promoting functionally‐active CNS synapses, by increasing the surface levels and synaptic clustering of AMPA glutamate receptors. This process involves tumor necrosis factor‐induced protein 6 (TSG6), remodeling of the perineuronal network, and a β1‐integrin/ERK pathway. Furthermore, PTX3 activity is regulated by TSP1, which directly interacts with the N‐terminal region of PTX3. These data unveil a fundamental role of PTX3 in promoting the first wave of synaptogenesis, and show that interplay of TSP1 and PTX3 sets the proper balance between synaptic growth and synapse function in the developing brain.
Collapse
Affiliation(s)
- Giuliana Fossati
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy
| | - Davide Pozzi
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy.,Department of Biomedical Sciences Humanitas University, Milan, Italy
| | - Alice Canzi
- Department of Biomedical Sciences Humanitas University, Milan, Italy
| | - Filippo Mirabella
- Department of Biomedical Sciences Humanitas University, Milan, Italy
| | - Sonia Valentino
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy
| | - Raffaella Morini
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy
| | - Elsa Ghirardini
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy.,Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, University of Milano, Milano, Italy
| | - Fabia Filipello
- Department of Biomedical Sciences Humanitas University, Milan, Italy
| | - Milena Moretti
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, University of Milano, Milano, Italy
| | | | - Douglas S Annis
- Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, WI, USA
| | - Deane F Mosher
- Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, WI, USA
| | - Cecilia Garlanda
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy.,Department of Biomedical Sciences Humanitas University, Milan, Italy
| | - Barbara Bottazzi
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy.,Department of Biomedical Sciences Humanitas University, Milan, Italy
| | - Giulia Taraboletti
- Tumor Angiogenesis Unit, Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Alberto Mantovani
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy.,Department of Biomedical Sciences Humanitas University, Milan, Italy
| | - Michela Matteoli
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy .,Institute of Neuroscience - CNR, Milano, Italy
| | - Elisabetta Menna
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milano, Italy .,Institute of Neuroscience - CNR, Milano, Italy
| |
Collapse
|
26
|
Garlanda C, Bottazzi B, Magrini E, Inforzato A, Mantovani A. PTX3, a Humoral Pattern Recognition Molecule, in Innate Immunity, Tissue Repair, and Cancer. Physiol Rev 2018; 98:623-639. [PMID: 29412047 DOI: 10.1152/physrev.00016.2017] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Innate immunity includes a cellular and a humoral arm. PTX3 is a fluid-phase pattern recognition molecule conserved in evolution which acts as a key component of humoral innate immunity in infections of fungal, bacterial, and viral origin. PTX3 binds conserved microbial structures and self-components under conditions of inflammation and activates effector functions (complement, phagocytosis). Moreover, it has a complex regulatory role in inflammation, such as ischemia/reperfusion injury and cancer-related inflammation, as well as in extracellular matrix organization and remodeling, with profound implications in physiology and pathology. Finally, PTX3 acts as an extrinsic oncosuppressor gene by taming tumor-promoting inflammation in murine and selected human tumors. Thus evidence suggests that PTX3 is a key homeostatic component at the crossroad of innate immunity, inflammation, tissue repair, and cancer. Dissecting the complexity of PTX3 pathophysiology and human genetics paves the way to diagnostic and therapeutic exploitation.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Barbara Bottazzi
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Elena Magrini
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Antonio Inforzato
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
27
|
Presta M, Foglio E, Churruca Schuind A, Ronca R. Long Pentraxin-3 Modulates the Angiogenic Activity of Fibroblast Growth Factor-2. Front Immunol 2018; 9:2327. [PMID: 30349543 PMCID: PMC6187966 DOI: 10.3389/fimmu.2018.02327] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/19/2018] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis, the process of new blood vessel formation from pre-existing ones, plays a key role in various physiological and pathological conditions. Alteration of the angiogenic balance, consequent to the deranged production of angiogenic growth factors and/or natural angiogenic inhibitors, is responsible for angiogenesis-dependent diseases, including cancer. Fibroblast growth factor-2 (FGF2) represents the prototypic member of the FGF family, able to induce a complex “angiogenic phenotype” in endothelial cells in vitro and a potent neovascular response in vivo as the consequence of a tight cross talk between pro-inflammatory and angiogenic signals. The soluble pattern recognition receptor long pentraxin-3 (PTX3) is a member of the pentraxin family produced locally in response to inflammatory stimuli. Besides binding features related to its role in innate immunity, PTX3 interacts with FGF2 and other members of the FGF family via its N-terminal extension, thus inhibiting FGF-mediated angiogenic responses in vitro and in vivo. Accordingly, PTX3 inhibits the growth and vascularization of FGF-dependent tumors and FGF2-mediated smooth muscle cell proliferation and artery restenosis. Recently, the characterization of the molecular bases of FGF2/PTX3 interaction has allowed the identification of NSC12, the first low molecular weight pan-FGF trap able to inhibit FGF-dependent tumor growth and neovascularization. The aim of this review is to provide an overview of the impact of PTX3 and PTX3-derived molecules on the angiogenic, inflammatory, and tumorigenic activity of FGF2 and their potential implications for the development of more efficacious anti-FGF therapeutic agents to be used in those clinical settings in which FGFs play a pathogenic role.
Collapse
Affiliation(s)
- Marco Presta
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Eleonora Foglio
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Ander Churruca Schuind
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
28
|
Erreni M, Manfredi AA, Garlanda C, Mantovani A, Rovere-Querini P. The long pentraxin PTX3: A prototypical sensor of tissue injury and a regulator of homeostasis. Immunol Rev 2018; 280:112-125. [PMID: 29027216 DOI: 10.1111/imr.12570] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tissue damage frequently occurs. The immune system senses it and enforces homeostatic responses that lead to regeneration and repair. The synthesis of acute phase molecules is emerging as a crucial event in this program. The prototypic long pentraxin PTX3 orchestrates the recruitment of leukocytes, stabilizes the provisional matrix in order to facilitate leukocyte and stem progenitor cells trafficking, promotes swift and safe clearance of dying cells and of autoantigens, limiting autoimmunity and protecting the vasculature. These non-redundant actions of PTX3 are necessary for the resolution of inflammation. Recent studies have highlighted the mechanisms by which PTX3 adapts the functions of innate immune cells, orchestrates tissue repair and contributes to select the appropriate acquired immune response in various tissues. Conversely, PTX3 continues to be produced in diseases where the inflammatory response does not resolve. It is therefore a valuable biomarker for more precise and personalized stratification of patients, often independently predicting clinical evolution and outcome. There is strong promise for novel therapies based on understanding the mechanisms with which PTX3 plays its homeostatic role, especially in regulating leukocyte migration and the resolution of inflammatory processes.
Collapse
Affiliation(s)
- Marco Erreni
- IRCCS Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy
| | - Angelo A Manfredi
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy
| | - Alberto Mantovani
- IRCCS Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy
| | - Patrizia Rovere-Querini
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
29
|
Anderson CE, Hamm LL, Batuman G, Kumbala DR, Chen CS, Kallu SG, Siriki R, Gadde S, Kleinpeter MA, Krane NK, Simon EE, He J, Chen J. The association of angiogenic factors and chronic kidney disease. BMC Nephrol 2018; 19:117. [PMID: 29783932 PMCID: PMC5963107 DOI: 10.1186/s12882-018-0909-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 04/27/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND There are limited data on the associations of circulating angiogenic factors with chronic kidney disease (CKD). We investigate the associations of circulating vascular endothelial growth factor (VEGF)-A, angiopoietin-1, angiopoietin-1/VEGF-A ratio, VEGF receptor 1 (VEGFR-1), VEGFR-2, and pentraxin-3 with CKD. METHODS We recruited 201 patients with CKD and 201 community controls without CKD from the greater New Orleans area. CKD was defined as estimated glomerular filtration rate (eGFR) < 60 mL/min/1.73 m2 or presence of albuminuria. Multivariable quantile and logistic regression models were used to examine the relationship between angiogenesis-related factors and CKD adjusting for confounding factors. RESULTS After adjusting for covariables including traditional cardiovascular disease (CVD) risk factors, C-reactive protein, and history of CVD, the medians (interquartile range) were 133.08 (90.39, 204.15) in patients with CKD vs. 114.17 (72.45, 170.32) pg/mL in controls without CKD (p = 0.002 for group difference) for VEGF-A; 3951.2 (2471.9, 6656.6) vs. 4270.5 (2763.7, 6537.2) pg/mL (p = 0.70) for angiopoietin-1; 25.87 (18.09, 47.90) vs. 36.55 (25.71, 61.10) (p = 0.0001) for angiopoietin-1/VEGF-A ratio; 147.81 (122.94, 168.79) vs. 144.16 (123.74, 168.05) ng/mL (p = 0.25) for VEGFR-1; 26.20 (22.67, 29.92) vs. 26.28 (23.10, 29.69) ng/mL (p = 0.31) for VEGFR-2; and 1.01 (0.79, 1.49)vs. 0.89 (0.58, 1.18) ng/mL (p = 0.01) for pentraxin-3, respectively. In addition, an elevated VEGF-A level and decreased angiopoietin-1/VEGF-A ratio were associated with increased odds of CKD. CONCLUSIONS These data indicate that plasma VEGF-A and pentraxin-3 levels were increased and the angiopoietin-1/VEGF-A ratio was decreased in patients with CKD. Future prospective studies are warranted to examine whether angiogenic factors play a role in progression of CKD.
Collapse
Affiliation(s)
- Christopher E. Anderson
- 0000 0001 2217 8588grid.265219.bDepartment of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, room 1504, New Orleans, LA 70112 USA
| | - L. Lee Hamm
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bTulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA USA
| | - Gem Batuman
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Damodar R. Kumbala
- 0000 0004 0608 1972grid.240416.5Department of Nephrology, Ochsner Health System, Ochsner Medical Center, 1514 Jefferson Highway, New Orleans, LA 70121 USA
| | - Chung-Shiuan Chen
- 0000 0001 2217 8588grid.265219.bDepartment of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, room 1504, New Orleans, LA 70112 USA
| | - Swapna G. Kallu
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Ravi Siriki
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Shilpa Gadde
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Myra A. Kleinpeter
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - N. Kevin Krane
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Eric E. Simon
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Jiang He
- 0000 0001 2217 8588grid.265219.bDepartment of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, room 1504, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bTulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA USA
| | - Jing Chen
- 0000 0001 2217 8588grid.265219.bDepartment of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, room 1504, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bTulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA USA
| |
Collapse
|
30
|
Grčević D, Sironi M, Valentino S, Deban L, Cvija H, Inforzato A, Kovačić N, Katavić V, Kelava T, Kalajzić I, Mantovani A, Bottazzi B. The Long Pentraxin 3 Plays a Role in Bone Turnover and Repair. Front Immunol 2018; 9:417. [PMID: 29556234 PMCID: PMC5845433 DOI: 10.3389/fimmu.2018.00417] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 02/15/2018] [Indexed: 01/04/2023] Open
Abstract
Pentraxin 3 (PTX3) is an inflammatory mediator acting as a fluid-phase pattern recognition molecule and playing an essential role in innate immunity and matrix remodeling. Inflammatory mediators also contribute to skeletal homeostasis, operating at multiple levels in physiological and pathological conditions. This study was designed to investigate the role of PTX3 in physiological skeletal remodeling and bone healing. Micro-computed tomography (μCT) and bone histomorphometry of distal femur showed that PTX3 gene-targeted female and male mice (ptx3−/−) had lower trabecular bone volume than their wild-type (ptx3+/+) littermates (BV/TV by μCT: 3.50 ± 1.31 vs 6.09 ± 1.17 for females, p < 0.0001; BV/TV 9.06 ± 1.89 vs 10.47 ± 1.97 for males, p = 0.0435). In addition, μCT revealed lower trabecular bone volume in second lumbar vertebra of ptx3−/− mice. PTX3 was increasingly expressed during osteoblast maturation in vitro and was able to reverse the negative effect of fibroblast growth factor 2 (FGF2) on osteoblast differentiation. This effect was specific for the N-terminal domain of PTX3 that contains the FGF2-binding site. By using the closed transversal tibial fracture model, we found that ptx3−/− female mice formed significantly less mineralized callus during the anabolic phase following fracture injury compared to ptx3+/+ mice (BV/TV 17.05 ± 4.59 vs 20.47 ± 3.32, p = 0.0195). Non-hematopoietic periosteal cells highly upregulated PTX3 expression during the initial phase of fracture healing, particularly CD51+ and αSma+ osteoprogenitor subsets, and callus tissue exhibited concomitant expression of PTX3 and FGF2 around the fracture site. Thus, PTX3 supports maintenance of the bone mass possibly by inhibiting FGF2 and its negative impact on bone formation. Moreover, PTX3 enables timely occurring sequence of callus mineralization after bone fracture injury. These results indicate that PTX3 plays an important role in bone homeostasis and in proper matrix mineralization during fracture repair, a reflection of the function of this molecule in tissue homeostasis and repair.
Collapse
Affiliation(s)
- Danka Grčević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Marina Sironi
- Humanitas Clinical and Research Center, Milan, Italy
| | | | - Livija Deban
- Humanitas Clinical and Research Center, Milan, Italy.,Oxford BioTherapeutics Ltd., Abingdon, United Kingdom
| | - Hrvoje Cvija
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Antonio Inforzato
- Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Nataša Kovačić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vedran Katavić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Tomislav Kelava
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivo Kalajzić
- Department of Reconstructive Sciences, School of Dental Medicine, UConn Health, Farmingam, CT, United States
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
31
|
Watanabe R, Sato Y, Ozawa N, Takahashi Y, Koba S, Watanabe T. Emerging Roles of Tumor Necrosis Factor-Stimulated Gene-6 in the Pathophysiology and Treatment of Atherosclerosis. Int J Mol Sci 2018; 19:E465. [PMID: 29401724 PMCID: PMC5855687 DOI: 10.3390/ijms19020465] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/22/2018] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor necrosis factor-stimulated gene-6 (TSG-6) is a 35-kDa glycoprotein that has been shown to exert anti-inflammatory effects in experimental models of arthritis, acute myocardial infarction, and acute cerebral infarction. Several lines of evidence have shed light on the pathophysiological roles of TSG-6 in atherosclerosis. TSG-6 suppresses inflammatory responses of endothelial cells, neutrophils, and macrophages as well as macrophage foam cell formation and vascular smooth muscle cell (VSMC) migration and proliferation. Exogenous TSG-6 infusion and endogenous TSG-6 attenuation with a neutralizing antibody for four weeks retards and accelerates, respectively, the development of aortic atherosclerotic lesions in ApoE-deficient mice. TSG-6 also decreases the macrophage/VSMC ratio (a marker of plaque instability) and promotes collagen fibers in atheromatous plaques. In patients with coronary artery disease (CAD), plasma TSG-6 levels are increased and TSG-6 is abundantly expressed in the fibrous cap within coronary atheromatous plaques, indicating that TSG-6 increases to counteract the progression of atherosclerosis and stabilize the plaque. These findings indicate that endogenous TSG-6 enhancement and exogenous TSG-6 replacement treatments are expected to emerge as new lines of therapy against atherosclerosis and related CAD. Therefore, this review provides support for the clinical utility of TSG-6 in the diagnosis and treatment of atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Rena Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Yuki Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Nana Ozawa
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Yui Takahashi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan.
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| |
Collapse
|
32
|
Day AJ, Milner CM. TSG-6: A multifunctional protein with anti-inflammatory and tissue-protective properties. Matrix Biol 2018; 78-79:60-83. [PMID: 29362135 DOI: 10.1016/j.matbio.2018.01.011] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 02/06/2023]
Abstract
Tumor necrosis factor- (TNF) stimulated gene-6 (TSG-6) is an inflammation-associated secreted protein that has been implicated as having important and diverse tissue protective and anti-inflammatory properties, e.g. mediating many of the immunomodulatory and beneficial activities of mesenchymal stem/stromal cells. TSG-6 is constitutively expressed in some tissues, which are either highly metabolically active or subject to challenges from the environment, perhaps providing protection in these contexts. The diversity of its functions are dependent on the binding of TSG-6 to numerous ligands, including matrix molecules such as glycosaminoglycans, as well as immune regulators and growth factors that themselves interact with these linear polysaccharides. It is becoming apparent that TSG-6 can directly affect matrix structure and modulate the way extracellular signalling molecules interact with matrix. In this review, we focus mainly on the literature for TSG-6 over the last 10 years, summarizing its expression, structure, ligand-binding properties, biological functions and highlighting TSG-6's potential as a therapeutic for a broad range of disease indications.
Collapse
Affiliation(s)
- Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK.
| | - Caroline M Milner
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
33
|
C-reactive protein and pentraxin-3 binding of factor H-like protein 1 differs from complement factor H: implications for retinal inflammation. Sci Rep 2018; 8:1643. [PMID: 29374201 PMCID: PMC5786067 DOI: 10.1038/s41598-017-18395-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/11/2017] [Indexed: 12/31/2022] Open
Abstract
Retinal inflammation plays a key role in the progression of age-related macular degeneration (AMD), a condition that leads to loss of central vision. The deposition of the acute phase pentraxin C-reactive protein (CRP) in the macula activates the complement system, thereby contributing to dysregulated inflammation. The complement protein factor H (FH) can bind CRP and down-regulate an inflammatory response. However, it is not known whether a truncated form of FH, called factor H-like protein 1 (FHL-1), which plays a significant regulatory role in the eye, also interacts with CRP. Here, we compare the binding properties of FHL-1 and FH to both CRP and the related protein pentraxin-3 (PTX3). We find that, unlike FH, FHL-1 can bind pro-inflammatory monomeric CRP (mCRP) as well as the circulating pentameric form. Furthermore, the four-amino acid C-terminal tail of FHL-1 (not present in FH) plays a role in mediating its binding to mCRP. PTX3 was found to be present in the macula of donor eyes and the AMD-associated Y402H polymorphism altered the binding of FHL-1 to PTX3. Our findings reveal that the binding characteristics of FHL-1 differ from those of FH, likely underpinning independent immune regulatory functions in the context of the human retina.
Collapse
|
34
|
Casula M, Montecucco F, Bonaventura A, Liberale L, Vecchié A, Dallegri F, Carbone F. Update on the role of Pentraxin 3 in atherosclerosis and cardiovascular diseases. Vascul Pharmacol 2017; 99:1-12. [PMID: 29051088 DOI: 10.1016/j.vph.2017.10.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/11/2017] [Accepted: 10/15/2017] [Indexed: 12/12/2022]
Abstract
Pentraxin 3 (PTX3) is an acute-phase protein that was recently demonstrated to play pleiotropic activities in cardiovascular (CV) diseases. Tumor necrosis factor and interleukins up-regulates PTX3 transcription in different cell types (i.e. endothelial cells, phagocytes, smooth muscle cells, fibroblasts and glial cells) involved in atherogenesis. By interacting with numerous ligands, PTX3 acts as a modulatory molecule of complement system, inflammatory response, angiogenesis, and vascular/tissue remodeling. Experimental data point to a beneficial role of PTX3 in atherosclerotic plaque development and vulnerability. Animal studies indicated a protective role of PTX3 signaling in ischemic/reperfusion injury and failing heart. Clinical studies have so far provided contrasting results, highlighting a debated role of PTX3 as an active mediator of endothelial dysfunction, atherosclerotic plaque vulnerability and worse outcome after ischemic events. Therefore, substantial evidence suggests a dual role of PTX3 as modulator or amplifiers of the innate immune response. The final result of PTX3 activation might be determined by a fine tuning of time, space and environmental signals. The aim of this review is to provide an overview of biological properties of PTX3 in CV diseases and to discuss the ability of PTX3 to act as a crossroad between pro- and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Matteo Casula
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland
| | - Alessandra Vecchié
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| |
Collapse
|
35
|
Long pentraxin 3: A novel multifaceted player in cancer. Biochim Biophys Acta Rev Cancer 2017; 1869:53-63. [PMID: 29175552 DOI: 10.1016/j.bbcan.2017.11.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/22/2017] [Accepted: 11/22/2017] [Indexed: 01/12/2023]
Abstract
Since its discovery in 1992, long pentraxin 3 (PTX3) has been characterized as soluble patter recognition receptor, a key player of the innate immunity arm with non-redundant functions in pathogen recognition and inflammatory responses. As a component of the extra-cellular matrix milieu, PTX3 has been implicated also in wound healing/tissue remodeling, cardiovascular diseases, fertility, and infectious diseases. Consequently, PTX3 levels in biological fluids have been proposed as a fluid-phase biomarker in different pathological conditions. In the last decade, experimental evidences have shown that PTX3 may exert a significant impact also on different aspects of cancer biology, including tumor onset, angiogenesis, metastatic dissemination and immune-modulation. However, it remains unclear whether PTX3 acts as a good cop or bad cop in cancer. In this review, we will summarize and discuss the scientific literature data focusing on the role of PTX3 in experimental and human tumors, including its putative translational implications.
Collapse
|
36
|
Impairment of PTX3 expression in osteoblasts: a key element for osteoporosis. Cell Death Dis 2017; 8:e3125. [PMID: 29022895 PMCID: PMC5682679 DOI: 10.1038/cddis.2017.514] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/19/2017] [Accepted: 08/30/2017] [Indexed: 01/21/2023]
Abstract
Pentraxin 3 (PTX3) is a multifunctional glycoprotein regulating inflammatory response, cell proliferation and migration and deposition and remodelling of the extracellular matrix by a variety of cells. In this study, we investigated the possible role of PTX3 in bone homeostasis. To this end, we compared the expression and function of PTX3 in human osteoblasts of osteoporotic, osteoarthritic patients and young subjects not affected by bone diseases. Immunohistochemical analysis performed on bone head biopsies showed a close association between bone health and the number of osteoblasts expressing PTX3. Noteworthy, the proportion of PTX3-positive osteoblasts resulted to be significantly lower in osteoporotic patients compared with both young patients and osteoarthritic patients of the same age. Ex vivo culture of osteoblasts isolated from the three groups of patients confirmed in vivo observation. Specifically, we observed rare runt-related transcription factor 2 (RUNX2) immunopositive osteoblasts expressing PTX3 in cell cultures derived from osteoporotic patients and western blotting analysis showed 80% reduction of PTX3 in the corresponding culture extracts compared with young and osteoarthritic patients. The treatment of human osteoblast primary cultures derived from young patients with anti-PTX3 antibody dramatically affected osteoblast behaviour. Indeed, they lost the morphological and molecular features typical of mature osteoblasts, acquiring fibroblast-like shape and highly decreasing nuclear factor kappa-B ligand (RANKL) and RUNX2 expression. Also, the inhibition of PTX3 negatively affected osteoblast proliferation and their ability to form cell clusters and microhydroxyapatite crystals. Altogether, these results suggest a central role of PTX3 in bone homeostasis showing its involvement in osteoblast proliferation, differentiation and function.
Collapse
|
37
|
Gesteira TF, Sun M, Coulson-Thomas YM, Yamaguchi Y, Yeh LK, Hascall V, Coulson-Thomas VJ. Hyaluronan Rich Microenvironment in the Limbal Stem Cell Niche Regulates Limbal Stem Cell Differentiation. Invest Ophthalmol Vis Sci 2017; 58:4407-4421. [PMID: 28863216 PMCID: PMC5584473 DOI: 10.1167/iovs.17-22326] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/31/2017] [Indexed: 02/07/2023] Open
Abstract
Purpose Limbal epithelial stem cells (LSCs), located in the basal layer of the corneal epithelium in the corneal limbus, are vital for maintaining the corneal epithelium. LSCs have a high capacity of self-renewal with increased potential for error-free proliferation and poor differentiation. To date, limited research has focused on unveiling the composition of the limbal stem cell niche, and, more important, on the role the specific stem cell niche may have in LSC differentiation and function. Our work investigates the composition of the extracellular matrix in the LSC niche and how it regulates LSC differentiation and function. Methods Hyaluronan (HA) is naturally synthesized by hyaluronan synthases (HASs), and vertebrates have the following three types: HAS1, HAS2, and HAS3. Wild-type and HAS and TSG-6 knockout mice-HAS1-/-;HAS3-/-, HAS2Δ/ΔCorEpi, TSG-6-/--were used to determine the importance of the HA niche in LSC differentiation and specification. Results Our data demonstrate that the LSC niche is composed of a HA rich extracellular matrix. HAS1-/-;HAS3-/-, HAS2Δ/ΔCorEpi, and TSG-6-/- mice have delayed wound healing and increased inflammation after injury. Interestingly, upon insult the HAS knock-out mice up-regulate HA throughout the cornea through a compensatory mechanism, and in turn this alters LSC and epithelial cell specification. Conclusions The LSC niche is composed of a specialized HA matrix that differs from that present in the rest of the corneal epithelium, and the disruption of this specific HA matrix within the LSC niche leads to compromised corneal epithelial regeneration. Finally, our findings suggest that HA has a major role in maintaining the LSC phenotype.
Collapse
MESH Headings
- Animals
- Burns, Chemical/metabolism
- Cell Differentiation/physiology
- Cellular Microenvironment/physiology
- Disease Models, Animal
- Epithelium, Corneal/metabolism
- Eye Burns/chemically induced
- Glucuronosyltransferase/metabolism
- Hyaluronan Synthases
- Hyaluronic Acid/genetics
- Hyaluronic Acid/metabolism
- Immunohistochemistry
- Limbus Corneae/cytology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal
- Microscopy, Electron, Transmission
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Sodium Hydroxide
- Stem Cell Niche/physiology
- Stem Cells/metabolism
- Wound Healing/physiology
Collapse
Affiliation(s)
| | - Mingxia Sun
- College of Optometry, University of Houston, Houston, Texas, United States
| | | | - Yu Yamaguchi
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States
| | - Lung-Kun Yeh
- Department of Ophthalmology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Linko, Taiwan
| | | | | |
Collapse
|
38
|
Daigo K, Inforzato A, Barajon I, Garlanda C, Bottazzi B, Meri S, Mantovani A. Pentraxins in the activation and regulation of innate immunity. Immunol Rev 2017; 274:202-217. [PMID: 27782337 DOI: 10.1111/imr.12476] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Humoral fluid phase pattern recognition molecules (PRMs) are a key component of the activation and regulation of innate immunity. Humoral PRMs are diverse. We focused on the long pentraxin PTX3 as a paradigmatic example of fluid phase PRMs. PTX3 acts as a functional ancestor of antibodies and plays a non-redundant role in resistance against selected microbes in mouse and man and in the regulation of inflammation. This molecule interacts with complement components, thus modulating complement activation. In particular, PTX3 regulates complement-driven macrophage-mediated tumor progression, acting as an extrinsic oncosuppressor in preclinical models and selected human tumors. Evidence collected over the years suggests that PTX3 is a biomarker and potential therapeutic agent in humans, and pave the way to translation of this molecule into the clinic.
Collapse
Affiliation(s)
- Kenji Daigo
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Antonio Inforzato
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Italy
| | | | - Cecilia Garlanda
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Barbara Bottazzi
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Seppo Meri
- Immunobiology Research Program, Research Programs Unit, Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki , Helsinki , Finland
| | - Alberto Mantovani
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, Rozzano (Milan), Italy.,Humanitas University, Rozzano, Italy
| |
Collapse
|
39
|
Naturil-Alfonso C, Peñaranda DS, Vicente JS, Marco-Jiménez F. Feed restriction regime in a rabbit line selected for growth rate alters oocyte maturation manifested by alteration in MSY2 gene expression. Reprod Domest Anim 2017. [PMID: 28627068 DOI: 10.1111/rda.13006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Young rabbit females selected for growth rate may have nutritional needs, which may not be met with the common practice of feed restriction during rearing in commercial rabbit production. The aim of this study was to analyse whether two different feeding programmes: ad libitum or restricted (130 g/day) feeding, applied in young rabbit females for 1 month at the end of rearing, could modulate the origin of ovulation process and the quality of the oocytes. At 16 weeks of age, 34 females were randomly assigned to restricted or ad libitum feeding, maintaining these conditions for a month. Then, in an initial experiment, transcriptional profiling of hypothalamus-hypophysis tissue was performed to assess failure to ovulate. In the second experiment, the gene expression analysis of some candidate genes related to oocytes quality was performed. Our results demonstrated that neither of the two feeding programmes modified the transcription of hypothalamus-hypophysis tissue, while the only differences in MSYR expression were found in in vivo mature oocytes ready for successful fertilization. Specifically, MSYR was over-expressed in oocytes from females fed ad libitum. MSYR is one of the most abundant proteins in the oocyte and has proven to be a key regulator of maternal RNA transcription and translation. This finding suggests that MSYR gene is a promising gene in our understanding of the relationship between high growth rate and reproductive performance decline.
Collapse
Affiliation(s)
- C Naturil-Alfonso
- Institute of Science and Animal Technology, Laboratorio de Biotecnología de la Reproducción, Universitat Politécnica de València, Valencia, Spain
| | - D S Peñaranda
- Institute of Science and Animal Technology, Laboratorio de Biotecnología de la Reproducción, Universitat Politécnica de València, Valencia, Spain
| | - J S Vicente
- Institute of Science and Animal Technology, Laboratorio de Biotecnología de la Reproducción, Universitat Politécnica de València, Valencia, Spain
| | - F Marco-Jiménez
- Institute of Science and Animal Technology, Laboratorio de Biotecnología de la Reproducción, Universitat Politécnica de València, Valencia, Spain
| |
Collapse
|
40
|
Wirestam L, Enocsson H, Skogh T, Eloranta ML, Rönnblom L, Sjöwall C, Wetterö J. Interferon-α coincides with suppressed levels of pentraxin-3 (PTX3) in systemic lupus erythematosus and regulates leucocyte PTX3 in vitro. Clin Exp Immunol 2017; 189:83-91. [PMID: 28257596 DOI: 10.1111/cei.12957] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2017] [Indexed: 12/20/2022] Open
Abstract
Dysfunctional elimination of cell debris, and the role of opsonins such as pentraxins, is of interest regarding systemic lupus erythematosus (SLE) pathogenesis. Interferon (IFN)-α is typically elevated during SLE flares, and inhibits hepatocyte production of the pentraxin 'C-reactive protein' (CRP), partly explaining the poor correlation between CRP levels and SLE disease activity. The extrahepatically produced 'pentraxin 3' (PTX3) shares waste disposal functions with CRP, but has not been studied extensively in SLE. We analysed serum PTX3 in SLE, and assessed its interference with IFN-α in vitro. Serum samples from 243 patients with SLE and 100 blood donors were analysed regarding PTX3. Patient sera were analysed for IFN-α, and genotyped for three PTX3 single nucleotide polymorphisms reported previously to associate with PTX3 levels. Stimulated PTX3 release was assessed in the presence or absence of IFN-α in blood donor neutrophils and peripheral blood mononuclear cells (PBMC). Serum PTX3 was 44% lower in patients with SLE compared to blood donors (P < 0·0001) and correlated with leucocyte variables. Patients with undetectable IFN-α had 29% higher median PTX3 level than patients with detectable IFN-α (P = 0·01). PTX3 production by PBMC was inhibited by IFN-α, whereas neutrophil degranulation of PTX3 was increased. No differences in PTX3 levels were observed between the SNPs. In conclusion, median serum PTX3 is lower in SLE (especially when IFN-α is detectable) compared to blood donors. In addition to its potential consumption during waste disposal, it is plausible that IFN-α also attenuates PTX3 by inhibiting synthesis by PBMC and/or exhausting PTX3 storage in neutrophil granules.
Collapse
Affiliation(s)
- L Wirestam
- Rheumatology/Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - H Enocsson
- Rheumatology/Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - T Skogh
- Rheumatology/Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - M L Eloranta
- Rheumatology, Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - L Rönnblom
- Rheumatology, Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - C Sjöwall
- Rheumatology/Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - J Wetterö
- Rheumatology/Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
41
|
O'Neill CL, Guduric-Fuchs J, Chambers SEJ, O'Doherty M, Bottazzi B, Stitt AW, Medina RJ. Endothelial cell-derived pentraxin 3 limits the vasoreparative therapeutic potential of circulating angiogenic cells. Cardiovasc Res 2016; 112:677-688. [PMID: 27659714 PMCID: PMC5157134 DOI: 10.1093/cvr/cvw209] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 08/09/2016] [Accepted: 08/17/2016] [Indexed: 01/14/2023] Open
Abstract
Aims Circulating angiogenic cells (CACs) promote revascularization of ischaemic tissues although their underlying mechanism of action and the consequences of delivering varying number of these cells for therapy remain unknown. This study investigates molecular mechanisms underpinning CAC modulation of blood vessel formation. Methods and results CACs at low (2 × 105 cells/mL) and mid (2 × 106 cells/mL) cellular densities significantly enhanced endothelial cell tube formation in vitro, while high density (HD) CACs (2 × 107 cells/mL) significantly inhibited this angiogenic process. In vivo, Matrigel-based angiogenesis assays confirmed mid-density CACs as pro-angiogenic and HD CACs as anti-angiogenic. Secretome characterization of CAC-EC conditioned media identified pentraxin 3 (PTX3) as only present in the HD CAC-EC co-culture. Recombinant PTX3 inhibited endothelial tube formation in vitro and in vivo. Importantly, our data revealed that the anti-angiogenic effect observed in HD CAC-EC co-cultures was significantly abrogated when PTX3 bioactivity was blocked using neutralizing antibodies or PTX3 siRNA in endothelial cells. We show evidence for an endothelial source of PTX3, triggered by exposure to HD CACs. In addition, we confirmed that PTX3 inhibits fibroblast growth factor (FGF) 2-mediated angiogenesis, and that the PTX3 N-terminus, containing the FGF-binding site, is responsible for such anti-angiogenic effects. Conclusion Endothelium, when exposed to HD CACs, releases PTX3 which markedly impairs the vascular regenerative response in an autocrine manner. Therefore, CAC density and accompanying release of angiocrine PTX3 are critical considerations when using these cells as a cell therapy for ischaemic disease.
Collapse
Affiliation(s)
- Christina L O'Neill
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast BT12 6BA, UK
| | - Jasenka Guduric-Fuchs
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast BT12 6BA, UK
| | - Sarah E J Chambers
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast BT12 6BA, UK
| | - Michelle O'Doherty
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast BT12 6BA, UK
| | - Barbara Bottazzi
- Humanitas Clinical and Research Centre, Rozzano 20089 Milan, Italy
| | - Alan W Stitt
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast BT12 6BA, UK
| | - Reinhold J Medina
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast BT12 6BA, UK
| |
Collapse
|
42
|
Fornai F, Carrizzo A, Forte M, Ambrosio M, Damato A, Ferrucci M, Biagioni F, Busceti C, Puca AA, Vecchione C. The inflammatory protein Pentraxin 3 in cardiovascular disease. IMMUNITY & AGEING 2016; 13:25. [PMID: 27559355 PMCID: PMC4995820 DOI: 10.1186/s12979-016-0080-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/15/2016] [Indexed: 12/12/2022]
Abstract
The acute phase protein Pentraxin 3 (PTX3) plays a non-redundant role as a soluble pattern recognition receptor for selected pathogens and it represents a rapid biomarker for primary local activation of innate immunity and inflammation. Recent evidence indicates that PTX3 exerts an important role in modulating the cardiovascular system in humans and experimental models. In particular, there are conflicting points concerning the effects of PTX3 in cardiovascular diseases (CVD) since several observations indicate a cardiovascular protective effect of PTX3 while others speculate that the increased plasma levels of PTX3 in subjects with CVD correlate with disease severity and with poor prognosis in elderly patients. In the present review, we discuss the multifaceted effects of PTX3 on the cardiovascular system focusing on its involvement in atherosclerosis, endothelial function, hypertension, myocardial infarction and angiogenesis. This may help to explain how the specific modulation of PTX3 such as the use of different dosing, time, and target organs could help to contain different vascular diseases. These opposite actions of PTX3 will be emphasized concerning the modulation of cardiovascular system where potential therapeutic implications of PTX3 in humans are discussed.
Collapse
Affiliation(s)
- Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy ; I.R.C.C.S. Neuromed, Pozzilli, IS Italy
| | | | | | | | | | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | | | - Annibale A Puca
- Vascular Physiopathology Unit, I.R.C.C.S. Multimedica, Milan, Italy ; Department of Medicine and Surgery, University of Salerno, Via S. Allende, Baronissi, SA 84081 Italy
| | - Carmine Vecchione
- I.R.C.C.S. Neuromed, Pozzilli, IS Italy ; Department of Medicine and Surgery, University of Salerno, Via S. Allende, Baronissi, SA 84081 Italy
| |
Collapse
|
43
|
Shindo A, Maki T, Mandeville ET, Liang AC, Egawa N, Itoh K, Itoh N, Borlongan M, Holder JC, Chuang TT, McNeish JD, Tomimoto H, Lok J, Lo EH, Arai K. Astrocyte-Derived Pentraxin 3 Supports Blood-Brain Barrier Integrity Under Acute Phase of Stroke. Stroke 2016; 47:1094-100. [PMID: 26965847 DOI: 10.1161/strokeaha.115.012133] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/09/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE Pentraxin 3 (PTX3) is released on inflammatory responses in many organs. However, roles of PTX3 in brain are still mostly unknown. Here we asked whether and how PTX3 contributes to blood-brain barrier dysfunction during the acute phase of ischemic stroke. METHODS In vivo, spontaneously hypertensive rats were subjected to focal cerebral ischemia by transient middle cerebral artery occlusion. At day 3, brains were analyzed to evaluate the cellular origin of PTX3 expression. Correlations with blood-brain barrier breakdown were assessed by IgG staining. In vitro, rat primary astrocytes and rat brain endothelial RBE.4 cells were cultured to study the role of astrocyte-derived PTX3 on vascular endothelial growth factor-mediated endothelial permeability. RESULTS During the acute phase of stroke, reactive astrocytes in the peri-infarct area expressed PTX3. There was negative correlation between gradients of IgG leakage and PTX3-positive astrocytes. Cell culture experiments showed that astrocyte-conditioned media increased levels of tight junction proteins and reduced endothelial permeability under normal conditions. Removing PTX3 from astrocyte-conditioned media by immunoprecipitation increased endothelial permeability. PTX3 strongly bound vascular endothelial growth factor in vitro and was able to decrease vascular endothelial growth factor-induced endothelial permeability. CONCLUSIONS Astrocytes in peri-infarct areas upregulate PTX3, which may support blood-brain barrier integrity by regulating vascular endothelial growth factor-related mechanisms. This response in astrocytes may comprise a compensatory mechanism for maintaining blood-brain barrier function after ischemic stroke.
Collapse
Affiliation(s)
- Akihiro Shindo
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Takakuni Maki
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Emiri T Mandeville
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Anna C Liang
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Naohiro Egawa
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Kanako Itoh
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Naoki Itoh
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Mia Borlongan
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Julie C Holder
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Tsu Tshen Chuang
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - John D McNeish
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Hidekazu Tomimoto
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Josephine Lok
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Eng H Lo
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.)
| | - Ken Arai
- From the Neuroprotection Research Laboratory, Departments of Radiology and Neurology (A.S., T.M., E.T.M., A.C.L., N.E., K.I., N.I., M.B., J.L., E.H.L., K.A.) and Pediatrics (J.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown, Boston; Department of Vascular Biology, GlaxoSmithKline, Harlow, United Kingdom (J.C.H., T.T.C., J.D.M.); and Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan (A.S., H.T.).
| |
Collapse
|
44
|
Guo S, Lok J, Zhao S, Leung W, Som AT, Hayakawa K, Wang Q, Xing C, Wang X, Ji X, Zhou Y, Lo EH. Effects of Controlled Cortical Impact on the Mouse Brain Vasculome. J Neurotrauma 2016; 33:1303-16. [PMID: 26528928 DOI: 10.1089/neu.2015.4101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Perturbations in blood vessels play a critical role in the pathophysiology of brain injury and neurodegeneration. Here, we use a systematic genome-wide transcriptome screening approach to investigate the vasculome after brain trauma in mice. Mice were subjected to controlled cortical impact and brains were extracted for analysis at 24 h post-injury. The core of the traumatic lesion was removed and then cortical microvesels were isolated from nondirectly damaged ipsilateral cortex. Compared to contralateral cortex and normal cortex from sham-operated mice, we identified a wide spectrum of responses in the vasculome after trauma. Up-regulated pathways included those involved in regulation of inflammation and extracellular matrix processes. Decreased pathways included those involved in regulation of metabolism, mitochondrial function, and transport systems. These findings suggest that microvascular perturbations can be widespread and not necessarily localized to core areas of direct injury per se and may further provide a broader gene network context for existing knowledge regarding inflammation, metabolism, and blood-brain barrier alterations after brain trauma. Further efforts are warranted to map the vasculome with higher spatial and temporal resolution from acute to delayed phase post-trauma. Investigating the widespread network responses in the vasculome may reveal potential mechanisms, therapeutic targets, and biomarkers for traumatic brain injury.
Collapse
Affiliation(s)
- Shuzhen Guo
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts
| | - Josephine Lok
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital , Harvard Medical School, Boston, Massachusetts
| | - Song Zhao
- 3 The Department of Spine Surgery, the First Hospital of Jilin University , Changchun, China
| | - Wendy Leung
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts
| | - Angel T Som
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts
| | - Kazuhide Hayakawa
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts
| | - Qingzhi Wang
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts
| | - Changhong Xing
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts
| | - Xiaoying Wang
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts
| | - Xunming Ji
- 4 Cerebrovascular Research Center, Department of Neurosurgery, Xuanwu Hospital, Capital Medical University , Beijing, China
| | - Yiming Zhou
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts
| | - Eng H Lo
- 1 Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital , Harvard Medical School, Charlestown, Massachusetts
| |
Collapse
|
45
|
Gorin C, Rochefort GY, Bascetin R, Ying H, Lesieur J, Sadoine J, Beckouche N, Berndt S, Novais A, Lesage M, Hosten B, Vercellino L, Merlet P, Le-Denmat D, Marchiol C, Letourneur D, Nicoletti A, Vital SO, Poliard A, Salmon B, Muller L, Chaussain C, Germain S. Priming Dental Pulp Stem Cells With Fibroblast Growth Factor-2 Increases Angiogenesis of Implanted Tissue-Engineered Constructs Through Hepatocyte Growth Factor and Vascular Endothelial Growth Factor Secretion. Stem Cells Transl Med 2016; 5:392-404. [PMID: 26798059 DOI: 10.5966/sctm.2015-0166] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/07/2015] [Indexed: 12/18/2022] Open
Abstract
Tissue engineering strategies based on implanting cellularized biomaterials are promising therapeutic approaches for the reconstruction of large tissue defects. A major hurdle for the reliable establishment of such therapeutic approaches is the lack of rapid blood perfusion of the tissue construct to provide oxygen and nutrients. Numerous sources of mesenchymal stem cells (MSCs) displaying angiogenic potential have been characterized in the past years, including the adult dental pulp. Establishment of efficient strategies for improving angiogenesis in tissue constructs is nevertheless still an important challenge. Hypoxia was proposed as a priming treatment owing to its capacity to enhance the angiogenic potential of stem cells through vascular endothelial growth factor (VEGF) release. The present study aimed to characterize additional key factors regulating the angiogenic capacity of such MSCs, namely, dental pulp stem cells derived from deciduous teeth (SHED). We identified fibroblast growth factor-2 (FGF-2) as a potent inducer of the release of VEGF and hepatocyte growth factor (HGF) by SHED. We found that FGF-2 limited hypoxia-induced downregulation of HGF release. Using three-dimensional culture models of angiogenesis, we demonstrated that VEGF and HGF were both responsible for the high angiogenic potential of SHED through direct targeting of endothelial cells. In addition, FGF-2 treatment increased the fraction of Stro-1+/CD146+ progenitor cells. We then applied in vitro FGF-2 priming to SHED before encapsulation in hydrogels and in vivo subcutaneous implantation. Our results showed that FGF-2 priming is more efficient than hypoxia at increasing SHED-induced vascularization compared with nonprimed controls. Altogether, these data demonstrate that FGF-2 priming enhances the angiogenic potential of SHED through the secretion of both HGF and VEGF.
Collapse
Affiliation(s)
- Caroline Gorin
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France Assistance Publique des Hôpitaux de Paris (AP-HP) Département d'Odontologie, Hôpitaux Universitaires PNVS, Paris, France
| | - Gael Y Rochefort
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Rumeyza Bascetin
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Hanru Ying
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Julie Lesieur
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Jérémy Sadoine
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Nathan Beckouche
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Sarah Berndt
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Anita Novais
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Matthieu Lesage
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Benoit Hosten
- INSERM UMR-S1144, Université Paris Descartes-Paris Diderot Sorbonne Paris Cité, AP-HP, Hôpital St. Louis, Unité Claude Kellershohn, Paris, France
| | - Laetitia Vercellino
- Université Paris Diderot, AP-HP, Hôpital St. Louis, Unité Claude Kellershohn, Paris, France
| | - Pascal Merlet
- Université Paris Diderot, AP-HP, Hôpital St. Louis, Unité Claude Kellershohn, Paris, France
| | - Dominique Le-Denmat
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Carmen Marchiol
- Institut Cochin, Plateforme Imagerie du vivant, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Didier Letourneur
- INSERM U1148, Laboratory of Vascular Translational Science, Université Paris Diderot Sorbonne Paris Cité, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, and Département Hospitalo-Universitaire Fibrosis, Inflammation, and Remodeling, Paris, France
| | - Antonino Nicoletti
- INSERM U1148, Laboratory of Vascular Translational Science, Université Paris Diderot Sorbonne Paris Cité, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, and Département Hospitalo-Universitaire Fibrosis, Inflammation, and Remodeling, Paris, France
| | - Sibylle Opsahl Vital
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France Assistance Publique des Hôpitaux de Paris (AP-HP) Département d'Odontologie, Hôpitaux Universitaires PNVS, Paris, France
| | - Anne Poliard
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Benjamin Salmon
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France Assistance Publique des Hôpitaux de Paris (AP-HP) Département d'Odontologie, Hôpitaux Universitaires PNVS, Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Catherine Chaussain
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France Assistance Publique des Hôpitaux de Paris (AP-HP) Département d'Odontologie, Hôpitaux Universitaires PNVS, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| |
Collapse
|
46
|
Kim DK, Choi H, Nishida H, Oh JY, Gregory C, Lee RH, Yu JM, Watanabe J, An SY, Bartosh TJ, Prockop DJ. Scalable Production of a Multifunctional Protein (TSG-6) That Aggregates with Itself and the CHO Cells That Synthesize It. PLoS One 2016; 11:e0147553. [PMID: 26793973 PMCID: PMC4721919 DOI: 10.1371/journal.pone.0147553] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 01/05/2016] [Indexed: 01/11/2023] Open
Abstract
TNF-α stimulated gene/protein 6 (TNFAIP6/TSG-6) is a multifunctional protein that has a number of potential therapeutic applications. Experiments and clinical trials with TSG-6, however, have been limited by the technical difficulties of producing the recombinant protein. We prepared stable clones of CHO cells that expressed recombinant human TSG-6 (rhTSG-6) as a secreted glycoprotein. Paradoxically, both cell number and protein production decreased dramatically when the clones were expanded. The decreases occurred because the protein aggregated the synthesizing CHO cells by binding to the brush border of hyaluronan that is found around many cultured cells. In addition, the rhTSG-6 readily self-aggregated. To address these problems, we added to the medium an inhibitor of hyaluronan synthesis and heparin to compete with the binding of TSG-6 to hyaluronan. Also, we optimized the composition of the culture medium, and transferred the CHO cells from a spinner culture system to a bioreactor that controlled pH and thereby decreased pH-dependent binding properties of the protein. With these and other improvements in the culture conditions, we obtained 57.0 mg ± 9.16 S.D. of rhTSG-6 in 5 or 6 liter of medium. The rhTSG-6 accounted for 18.0% ± 3.76 S.D. of the total protein in the medium. We then purified the protein with a Ni-chelate column that bound the His tag engineered into the C-terminus of the protein followed by an anion exchange column. The yield of the purified monomeric rhTSG-6 was 4.1 mg to 5.6 mg per liter of culture medium. After intravenous injection into mice, the protein had a longer plasma half-life than commercially available rhTSG-6 isolated from a mammalian cell lysate, apparently because it was recovered as a secreted glycoprotein. The bioactivity of the rhTSG-6 in suppressing inflammation was demonstrated in a murine model.
Collapse
Affiliation(s)
- Dong-Ki Kim
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Hosoon Choi
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Hidetaka Nishida
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Joo Youn Oh
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Carl Gregory
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Ryang Hwa Lee
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Ji Min Yu
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Jun Watanabe
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Su Yeon An
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Thomas J. Bartosh
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Darwin J. Prockop
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
- * E-mail:
| |
Collapse
|
47
|
Rajkovic I, Denes A, Allan SM, Pinteaux E. Emerging roles of the acute phase protein pentraxin-3 during central nervous system disorders. J Neuroimmunol 2016; 292:27-33. [PMID: 26943955 DOI: 10.1016/j.jneuroim.2015.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 12/13/2015] [Accepted: 12/16/2015] [Indexed: 12/24/2022]
Abstract
Pentraxin-3 (PTX3) is an acute phase protein (APP) and a member of the long pentraxin family that is recognised for its role in peripheral immunity and vascular inflammation in response to injury, infection and diseases such as atherosclerosis, cancer and respiratory disease. Systemic levels of PTX3 are highly elevated in these conditions, and PTX3 is now recognised as a new biomarker of disease risk and progression. There is extensive evidence demonstrating that central nervous system (CNS) disorders are primarily characterised by central activation of innate immunity, as well as activation of a potent peripheral acute phase response (APR) that influences central inflammation and contributes to poor outcome. PTX3 has been recently recognised to play important roles in CNS disorders, having both detrimental and neuroprotective effects. The present review aims to give an up-to-date account of the emerging roles of PTX3 in CNS disorders, and to provide a critical comparison between peripheral and central actions of PTX3 in inflammatory diseases.
Collapse
Affiliation(s)
- Ivana Rajkovic
- Faculty of Life Sciences, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Adam Denes
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest H-1450, Hungary
| | - Stuart M Allan
- Faculty of Life Sciences, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Emmanuel Pinteaux
- Faculty of Life Sciences, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
48
|
Fornai F, Carrizzo A, Ferrucci M, Damato A, Biagioni F, Gaglione A, Puca AA, Vecchione C. Brain diseases and tumorigenesis: The good and bad cops of pentraxin3. Int J Biochem Cell Biol 2015; 69:70-4. [DOI: 10.1016/j.biocel.2015.10.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 12/12/2022]
|
49
|
Ronca R, Giacomini A, Di Salle E, Coltrini D, Pagano K, Ragona L, Matarazzo S, Rezzola S, Maiolo D, Torrella R, Moroni E, Mazzieri R, Escobar G, Mor M, Colombo G, Presta M. Long-Pentraxin 3 Derivative as a Small-Molecule FGF Trap for Cancer Therapy. Cancer Cell 2015; 28:225-39. [PMID: 26267536 DOI: 10.1016/j.ccell.2015.07.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 04/10/2015] [Accepted: 07/10/2015] [Indexed: 11/16/2022]
Abstract
The fibroblast growth factor (FGF)/FGF receptor (FGFR) system plays a crucial role in cancer by affecting tumor growth, angiogenesis, drug resistance, and escape from anti-angiogenic anti-vascular endothelial growth factor therapy. The soluble pattern recognition receptor long-pentraxin 3 (PTX3) acts as a multi-FGF antagonist. Here we demonstrate that human PTX3 overexpression in transgenic mice driven by the Tie2 promoter inhibits tumor growth, angiogenesis, and metastasis in heterotopic, orthotopic, and autochthonous FGF-dependent tumor models. Using pharmacophore modeling of the interaction of a minimal PTX3-derived FGF-binding pentapeptide with FGF2, we identified a small-molecule chemical (NSC12) that acts as an extracellular FGF trap with significant implications in cancer therapy.
Collapse
Affiliation(s)
- Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Emanuela Di Salle
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Daniela Coltrini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Katiuscia Pagano
- NMR Laboratory, Istituto per lo Studio delle Macromolecole, CNR, 20133 Milan, Italy
| | - Laura Ragona
- NMR Laboratory, Istituto per lo Studio delle Macromolecole, CNR, 20133 Milan, Italy
| | - Sara Matarazzo
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Daniele Maiolo
- Chemistry for Technologies Laboratory and INSTM, School of Engineering, University of Brescia, 25123 Brescia, Italy
| | - Rubben Torrella
- Istituto di Chimica del Riconoscimento Molecolare, CNR, 20133 Milan, Italy
| | - Elisabetta Moroni
- Istituto di Chimica del Riconoscimento Molecolare, CNR, 20133 Milan, Italy
| | - Roberta Mazzieri
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Giulia Escobar
- San Raffaele Telethon Institute for Gene Therapy, 20132 Milan, Italy; Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Marco Mor
- Department of Pharmacy, University of Parma, 43121 Parma, Italy
| | - Giorgio Colombo
- Istituto di Chimica del Riconoscimento Molecolare, CNR, 20133 Milan, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
50
|
Rodriguez-Grande B, Varghese L, Molina-Holgado F, Rajkovic O, Garlanda C, Denes A, Pinteaux E. Pentraxin 3 mediates neurogenesis and angiogenesis after cerebral ischaemia. J Neuroinflammation 2015; 12:15. [PMID: 25616391 PMCID: PMC4308938 DOI: 10.1186/s12974-014-0227-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 12/20/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The acute phase protein pentraxin 3 (PTX3) is a new biomarker of stroke severity and is a key regulator of oedema resolution and glial responses after cerebral ischaemia, emerging as a possible target for brain repair after stroke. Neurogenesis and angiogenesis are essential events in post-stroke recovery. Here, we investigated for the first time the role of PTX3 in neurogenesis and angiogenesis after stroke. METHODS PTX3 knockout (KO) or wild-type (WT) mice were subjected to experimental cerebral ischaemia (induced by middle cerebral artery occlusion (MCAo)). Poststroke neurogenesis was assessed by nestin, doublecortin (DCX) and bromodeoxyuridine (BrdU) immunostaining, whereas angiogenesis was assessed by BrdU, vascular endothelial growth factor receptor 2 (VEGFR2) and PECAM-1 immunostaining. In vitro neurogenesis and angiogenesis assays were carried out on neurospheres derived from WT or interleukin-1β (IL-1β) KO mice, and mouse endothelial cell line bEnd.5 respectively. Behavioural function was assessed in WT and PTX3 KO mice using open-field, motor and Y-maze tests. RESULTS Neurogenesis was significantly reduced in the dentate gyrus (DG) of the hippocampus of PTX3 KO mice, compared to WT mice, 6 days after MCAo. In addition, recombinant PTX3 was neurogenic in vitro when added to neurospheres, which was mediated by IL-1β. In vivo poststroke angiogenesis was significantly reduced in PTX3 KO mice compared to WT mice 14 days after MCAo, as revealed by reduced vascular density, less newly formed blood vessels and decreased expression of VEGFR2. In vitro, recombinant PTX3 induced marked endothelial cellular proliferation and promoted formation of tube-like structures of endothelial cell line bEnd.5. Finally, a lack of PTX3 potentiated motor deficits 14 days after MCAo. CONCLUSIONS These results indicate that PTX3 mediates neurogenesis and angiogenesis and contributes to functional recovery after stroke, highlighting a key role of PTX3 as a mediator of brain repair and suggesting that PTX3 could be used as a new target for stroke therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Emmanuel Pinteaux
- Faculty of Life Sciences, A,V, Hill Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|