1
|
He Y, Shen X, Zhai K, Nian S. Advances in understanding the role of interleukins in pulmonary fibrosis (Review). Exp Ther Med 2025; 29:25. [PMID: 39650776 PMCID: PMC11619568 DOI: 10.3892/etm.2024.12775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/03/2024] [Indexed: 12/11/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive, irreversible disease characterized by heterogeneous interstitial lung tissue damage. It originates from persistent or repeated lung epithelial injury and leads to the activation and differentiation of fibroblasts into myofibroblasts. Interleukins (ILs) are a group of lymphokines crucial for immunomodulation that are implicated in the pathogenesis of PF. However, different types of ILs exert disparate effects on PF. In the present review, based on the effect on PF, ILs are classified into three categories: i) Promotors of PF; ii) inhibitors of PF; and iii) those that exert dual effects on PF. Several types of ILs can promote PF by provoking inflammation, initiating proliferation and transdifferentiation of epithelial cells, exacerbating lung injury, while other ILs can inhibit PF through suppressing expression of inflammatory factors, modulating the Th1/Th2 balance and autophagy. The present review summarizes the association of ILs and PF, focusing on the roles and mechanisms of ILs underlying PF.
Collapse
Affiliation(s)
- Yuqing He
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xuebin Shen
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Kefeng Zhai
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, P.R. China
| | - Sihui Nian
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
2
|
Weng P, Lan M, Zhang H, Fan H, Wang X, Ran C, Yue Z, Hu J, Xu A, Huang S. Both IRAK3 and IRAK1 Activate the MyD88-TRAF6 Pathway in Zebrafish. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:362-372. [PMID: 38847613 DOI: 10.4049/jimmunol.2400054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/20/2024] [Indexed: 07/17/2024]
Abstract
IL-1R-associated kinases (IRAKs) are signal transducers of the TLR/IL-1R-MyD88-TRAF6 pathways. Vertebrates possess two IRAK lineages, IRAK1/2/3 and IRAK4. In mammals, IRAK4/IRAK1 and IRAK4/IRAK2 are pathway enhancers, whereas IRAK3 is a repressor. However, in bony fish, IRAK2 is absent, and it remains elusive how fish IRAK1/3/4 functionally differ from their mammalian counterparts. In this study, we explored this using the zebrafish model. First, we showed that in human 293T cells, zebrafish IRAK1 and IRAK4 were components of the Myddosome (MyD88-IRAK4-IRAK1) complex, with IRAK1 serving as a potent pathway enhancer. Then, we discovered two zebrafish IRAK3 variants: one (IRAK3a) contains an N-terminal Death domain, a middle pseudokinase domain, and a C-terminal TRAF6-binding domain, whereas the other (IRAK3b) lost both the kinase and TRAF6-binding domains. This truncation of IRAK3 variants could be a conserved phenomenon in fish, because it is also observed in trout and grass carp. We proceeded to show that zebrafish IRAK3a acts as a pathway enhancer by binding with MyD88 and TRAF6, but its activity is milder than IRAK1, possibly because it has no kinase activity. Zebrafish IRAK3b, however, plays a sheer negative role, apparently because of its lack of kinase and TRAF6-binding domains. Moreover, zebrafish IRAK3a/3b inhibit the activity of IRAK1/4, not by interacting with IRAK1/4 but possibly by competing for MyD88 and TRAF6. Finally, we have verified the essential activities of zebrafish IRAK1/3a/3b/4 in zebrafish cells and embryos. In summary, to our knowledge, our findings provide new insights into the molecular functions of fish IRAKs and the evolution of the IRAK functional modes in vertebrates.
Collapse
Affiliation(s)
- Panwei Weng
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Mengjiao Lan
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Hao Zhang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Huiping Fan
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Xiao Wang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Chenrui Ran
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Zirui Yue
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Jiaxuan Hu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Anlong Xu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Shengfeng Huang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| |
Collapse
|
3
|
Lu KP, Zhou XZ. Pin1-catalyzed conformational regulation after phosphorylation: A distinct checkpoint in cell signaling and drug discovery. Sci Signal 2024; 17:eadi8743. [PMID: 38889227 PMCID: PMC11409840 DOI: 10.1126/scisignal.adi8743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
Protein phosphorylation is one of the most common mechanisms regulating cellular signaling pathways, and many kinases and phosphatases are proven drug targets. Upon phosphorylation, protein functions can be further regulated by the distinct isomerase Pin1 through cis-trans isomerization. Numerous protein targets and many important roles have now been elucidated for Pin1. However, no tools are available to detect or target cis and trans conformation events in cells. The development of Pin1 inhibitors and stereo- and phospho-specific antibodies has revealed that cis and trans conformations have distinct and often opposing cellular functions. Aberrant conformational changes due to the dysregulation of Pin1 can drive pathogenesis but can be effectively targeted in age-related diseases, including cancers and neurodegenerative disorders. Here, we review advances in understanding the roles of Pin1 signaling in health and disease and highlight conformational regulation as a distinct signal transduction checkpoint in disease development and treatment.
Collapse
Affiliation(s)
- Kun Ping Lu
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry
- Robarts Research Institute, Schulich School of Medicine & Dentistry
| | - Xiao Zhen Zhou
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry
- Lawson Health Research Institute, Western University, London, ON N6G 2V4, Canada
| |
Collapse
|
4
|
Schettini N, Pacetti L, Corazza M, Borghi A. The Role of OX40-OX40L Axis in the Pathogenesis of Atopic Dermatitis. Dermatitis 2024. [PMID: 38700255 DOI: 10.1089/derm.2024.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
OX40 is a co-stimulatory immune checkpoint molecule that promotes the activation and the effector function of T lymphocytes through interaction with its ligand (OX40L) on antigen-presenting cells. OX40-OX40L axis plays a crucial role in Th1 and Th2 cell expansion, particularly during the late phases or long-lasting response. Atopic dermatitis is characterized by an immune dysregulation of Th2 activity and by an overproduction of proinflammatory cytokines such as interleukin (IL)-4 and IL-13. Other molecules involved in its pathogenesis include thymic stromal lymphopoietin, IL-33, and IL-25, which contribute to the promotion of OX40L expression on dendritic cells. Lesional skin in atopic dermatitis exhibits a higher level of OX40L+-presenting cells compared with other dermatologic diseases or normal skin. Recent clinical trials using antagonizing anti-OX40 or anti-OX40L antibodies have shown symptom improvement and cutaneous manifestation alleviation in patients with atopic dermatitis. These findings suggest the relevance of the OX40-OX40L axis in atopic dermatitis pathogenesis.
Collapse
Affiliation(s)
- Natale Schettini
- From the Section of Dermatology and Infectious Diseases, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Lucrezia Pacetti
- From the Section of Dermatology and Infectious Diseases, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Monica Corazza
- From the Section of Dermatology and Infectious Diseases, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandro Borghi
- From the Section of Dermatology and Infectious Diseases, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
5
|
Wang X, Shields C, Tardo G, Peacock G, Hester E, Anderson M, Williams JM, Cornelius DC. IL-33 supplementation improves uterine artery resistance and maternal hypertension in response to placental ischemia. Am J Physiol Heart Circ Physiol 2024; 326:H1006-H1016. [PMID: 38363211 PMCID: PMC11279736 DOI: 10.1152/ajpheart.00045.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/17/2024]
Abstract
Preeclampsia (PE), a leading cause of maternal/fetal morbidity and mortality, is a hypertensive pregnancy disorder with end-organ damage that manifests after 20 wk of gestation. PE is characterized by chronic immune activation and endothelial dysfunction. Clinical studies report reduced IL-33 signaling in PE. We use the Reduced Uterine Perfusion Pressure (RUPP) rat model, which mimics many PE characteristics including reduced IL-33, to identify mechanisms mediating PE pathophysiology. We hypothesized that IL-33 supplementation would improve blood pressure (BP), inflammation, and oxidative stress (ROS) during placental ischemia. We implanted intraperitoneal mini-osmotic pumps infusing recombinant rat IL-33 (1 µg/kg/day) into normal pregnant (NP) and RUPP rats from gestation day 14 to 19. We found that IL-33 supplementation in RUPP rats reduces maternal blood pressure and improves the uterine artery resistance index (UARI). In addition to physiological improvements, we found decreased circulating and placental cytolytic Natural Killer cells (cNKs) and decreased circulating, placental, and renal TH17s in IL-33-treated RUPP rats. cNK cell cytotoxic activity also decreased in IL-33-supplemented RUPP rats. Furthermore, renal ROS and placental preproendothelin-1 (PPET-1) decreased in RUPP rats treated with IL-33. These findings demonstrate a role for IL-33 in controlling vascular function and maternal BP during pregnancy by decreasing inflammation, renal ROS, and PPET-1 expression. These data suggest that IL-33 may have therapeutic potential in managing PE.NEW & NOTEWORTHY Though decreased IL-33 signaling has been clinically associated with PE, the mechanisms linking this signaling pathway to overall disease pathophysiology are not well understood. This study provides compelling evidence that mechanistically links reduced IL-33 with the inflammatory response and vascular dysfunction observed in response to placental ischemia, such as in PE. Data presented in this study submit the IL-33 signaling pathway as a possible therapeutic target for the treatment of PE.
Collapse
Affiliation(s)
- Xi Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Corbin Shields
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Geilda Tardo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Greg Peacock
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Emily Hester
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Marissa Anderson
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
6
|
Asadi M, Zarredar H, Zafari V, Soleimani Z, Saeedi H, Caner A, Shanehbandi D. Immune Features of Tumor Microenvironment: A Genetic Spotlight. Cell Biochem Biophys 2024; 82:107-118. [PMID: 37870699 DOI: 10.1007/s12013-023-01192-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
A tumor represents a highly intricate tissue entity, characterized by an exceptionally complex microenvironment that starkly contrasts with the typical physiological surroundings of healthy tissues. Within this tumor microenvironment (TME), every component and factor assume paramount importance in the progression of malignancy and exerts a pivotal influence on a patient's clinical outcome. One of the remarkable aspects of the TME is its remarkable heterogeneity, not only across different types of cancers but even within the same histological category of tumors. In-depth research has illuminated the intricate interplay between specific immune cells and molecules and the dynamic characteristics of the TME. Recent investigations have yielded compelling evidence that several mutations harbored by tumor cells possess the capacity to instigate substantial alterations in the TME. These mutations, often acting as drivers of tumorigenesis, can orchestrate a cascade of events that remodel the TME, thereby influencing crucial aspects of cancer behavior, including its invasiveness, immune evasion, and response to therapies. It is within this nuanced context that the present study endeavors to provide a concise yet comprehensive summary of how specific mutations, within the genetic landscape of cancer cells, can instigate profound changes in TME features. By elucidating the intricate relationship between genetic mutations and the TME, this research aims to contribute to a deeper understanding of cancer biology. Ultimately, the knowledge gained from this study holds the potential to inform the development of more targeted and effective treatments, thereby offering new hope to patients grappling with the complexities of cancer.
Collapse
Affiliation(s)
- Milad Asadi
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Venus Zafari
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Zahra Soleimani
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayse Caner
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey.
- The University of Texas, MD Anderson Cancer Center, Houston, USA.
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
7
|
Gürkan B, Poelman H, Pereverzeva L, Kruijswijk D, de Vos AF, Groenen AG, Nollet EE, Wichapong K, Lutgens E, van der Poll T, Du J, Wiersinga WJ, Nicolaes GAF, van ‘t Veer C. The IRAK-M death domain: a tale of three surfaces. Front Mol Biosci 2024; 10:1265455. [PMID: 38268724 PMCID: PMC10806146 DOI: 10.3389/fmolb.2023.1265455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 12/29/2023] [Indexed: 01/26/2024] Open
Abstract
The anti-inflammatory interleukin-1 receptor associated kinase-M (IRAK-M) is a negative regulator of MyD88/IRAK-4/IRAK-1 signaling. However, IRAK-M has also been reported to activate NF-κB through the MyD88/IRAK-4/IRAK-M myddosome in a MEKK-3 dependent manner. Here we provide support that IRAK-M uses three surfaces of its Death Domain (DD) to activate NF-κB downstream of MyD88/IRAK-4/IRAK-M. Surface 1, with central residue Trp74, binds to MyD88/IRAK-4. Surface 2, with central Lys60, associates with other IRAK-M DDs to form an IRAK-M homotetramer under the MyD88/IRAK-4 scaffold. Surface 3; with central residue Arg97 is located on the opposite side of Trp74 in the IRAK-M DD tetramer, lacks any interaction points with the MyD88/IRAK-4 complex. Although the IRAK-M DD residue Arg97 is not directly involved in the association with MyD88/IRAK-4, Arg97 was responsible for 50% of the NF-κB activation though the MyD88/IRAK-4/IRAK-M myddosome. Arg97 was also found to be pivotal for IRAK-M's interaction with IRAK-1, and important for IRAK-M's interaction with TRAF6. Residue Arg97 was responsible for 50% of the NF-κB generated by MyD88/IRAK-4/IRAK-M myddosome in IRAK-1/MEKK3 double knockout cells. By structural modeling we found that the IRAK-M tetramer surface around Arg97 has excellent properties that allow formation of an IRAK-M homo-octamer. This model explains why mutation of Arg97 results in an IRAK-M molecule with increased inhibitory properties: it still binds to myddosome, competing with myddosome IRAK-1 binding, while resulting in less NF-κB formation. The findings further identify the structure-function properties of IRAK-M, which is a potential therapeutic target in inflammatory disease.
Collapse
Affiliation(s)
- Berke Gürkan
- Center of Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Hessel Poelman
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
- Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Liza Pereverzeva
- Center of Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Danielle Kruijswijk
- Center of Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Alex F. de Vos
- Center of Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Anouk G. Groenen
- Center of Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Edgar E. Nollet
- Center of Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Kanin Wichapong
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Esther Lutgens
- Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Division of Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Jiangfeng Du
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - W. Joost Wiersinga
- Center of Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Division of Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Gerry A. F. Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
- Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Cornelis van ‘t Veer
- Center of Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
8
|
Li W, Liu M, Chu M. Strategies targeting IL-33/ST2 axis in the treatment of allergic diseases. Biochem Pharmacol 2023; 218:115911. [PMID: 37981174 DOI: 10.1016/j.bcp.2023.115911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023]
Abstract
Interleukin-33 (IL-33) and its receptor Serum Stimulation-2 (ST2, also called Il1rl1) are members of the IL-1 superfamily that plays a crucial role in allergic diseases. The interaction of IL-33 and ST2 mainly activates NF-κB signaling and MAPK signaling via the MyD88/IRAK/TRAF6 module, resulting in the production and secretion of pro-inflammatory cytokines. The IL-33/ST2 axis participates in the pathogenesis of allergic diseases, and therefore serves as a promising strategy for allergy treatment. In recent years, strategies blocking IL-33/ST2 through targeting regulation of IL-33 and ST2 or targeting the molecules involved in the signal transduction have been extensively studied mostly in animal models. These studies provide various potential therapeutic agents other than antibodies, such as small molecules, nucleic acids and traditional Chinese medicines. Herein, we reviewed potential targets and agents targeting IL-33/ST2 axis in the treatment of allergic diseases, providing directions for further investigations on treatments for IL-33 induced allergic diseases.
Collapse
Affiliation(s)
- Wenran Li
- Department of Immunology, School of Basic Medical Sciences, Health Science Centre, Peking University. Beijing, China
| | - Mengqi Liu
- Department of Immunology, School of Basic Medical Sciences, Health Science Centre, Peking University. Beijing, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Sciences, Health Science Centre, Peking University. Beijing, China; Beijing Life Science Academy, Beijing, China.
| |
Collapse
|
9
|
Qing J, Guo Q, Lv L, Zhang X, Liu Y, Heng BC, Li Z, Zhang P, Zhou Y. Organoid Culture Development for Skeletal Systems. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:545-557. [PMID: 37183418 DOI: 10.1089/ten.teb.2023.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Organoids are widely considered to be ideal in vitro models that have been widely applied in many fields, including regenerative medicine, disease research and drug screening. It is distinguished from other three-dimensional in vitro culture model systems by self-organization and sustainability in long-term culture. The three core components of organoid culture are cells, exogenous factors, and culture matrix. Due to the complexity of bone tissue, and heterogeneity of osteogenic stem/progenitor cells, it is challenging to construct organoids for modeling skeletal systems. In this study, we examine current progress in the development of skeletal system organoid culture systems and analyze the current research status of skeletal stem cells, their microenvironmental factors, and various potential organoid culture matrix candidates to provide cues for future research trajectory in this field. Impact Statement The emergence of organoids has brought new opportunities for the development of many biomedical fields. The bone organoid field still has much room for exploration. This review discusses the characteristics distinguishing organoids from other three-dimensional model systems and examines current progress in the organoid production of skeletal systems. In addition, based on core elements of organoid cultures, three main problems that need to be solved in bone organoid generation are further analyzed. These include the heterogeneity of skeletal stem cells, their microenvironmental factors, and potential organoid culture matrix candidates. This information provides direction for the future research of bone organoids.
Collapse
Affiliation(s)
- Jia Qing
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Qian Guo
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Boon Chin Heng
- The Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| |
Collapse
|
10
|
Mahmoud IS, Jarrar YB, Febrimarsa. Modulation of IRAK enzymes as a therapeutic strategy against SARS-CoV-2 induced cytokine storm. Clin Exp Med 2023; 23:2909-2923. [PMID: 37061574 PMCID: PMC10105542 DOI: 10.1007/s10238-023-01064-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/02/2023] [Indexed: 04/17/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the current pandemic coronavirus disease 2019 (COVID-19). Dysregulated and excessive production of cytokines and chemokines, known as cytokine storm, is frequently seen in patients with severe COVID-19 disease and it can provoke a severe systematic inflammation in the patients. The IL-1R/TLRs/IRAKs signaling network is a key pathway in immune cells that plays a central role in regulating innate immunity and inflammatory responses via stimulating the expression and production of various proinflammatory molecules including cytokines. Modulation of IRAKs activity has been proposed to be a promising strategy in the treatment of inflammatory disorders. In this review, we highlight the biochemical properties of IRAKs and their role in regulating inflammatory molecular signaling pathways and discuss the potential targeting of IRAKs to suppress the SARS-CoV-2-induced cytokine storm in COVID-19 patients.
Collapse
Affiliation(s)
- Ismail Sami Mahmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan.
| | - Yazun Bashir Jarrar
- Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, As-Salt, Jordan
| | - Febrimarsa
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway, Republic of Ireland
| |
Collapse
|
11
|
Lan T, Tang T, Li Y, Duan Y, Yuan Q, Liu W, Ren Y, Li N, Liu X, Zhang Y, Li X, Jin G, Wang S, Guo J. FTZ polysaccharides ameliorate kidney injury in diabetic mice by regulating gut-kidney axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154935. [PMID: 37364420 DOI: 10.1016/j.phymed.2023.154935] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/26/2023] [Accepted: 06/19/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND The Fufang-zhenzhu-tiaozhi formula (FTZ), a traditional Chinese medicine (TCM) commonly used to treat metabolic diseases, potentially impacts the microbial ecosystem. Increasing evidence suggests that polysaccharides, bioactive components of TCMs, have great potential on kinds of diseases such as DKD by regulating intestinal flora. PURPOSE This study aimed to investigate whether the polysaccharide components in FTZ (FTZPs) have beneficial effects in DKD mice via the gut-kidney axis. STUDY DESIGN AND METHODS The DKD model in mice was established by streptozotocin combined with a high-fat diet (STZ/HFD). Losartan was used as a positive control, and FTZPs were administered at doses of 100 and 300 mg/kg daily. Renal histological changes were measured by H&E and Masson staining. Western blotting, quantitative real-time polymerase chain reaction (q-PCR) and immunohistochemistry were performed to analyze the effects of FTZPs on renal inflammation and fibrosis, which were further confirmed using RNA sequencing. Immunofluorescence was used to analyze the effects of FTZPs on colonic barrier function in DKD mice. Faecal microbiota transplantation (FMT) was used to evaluate the contribution of intestinal flora. 16S rRNA sequencing was utilized to analyze the composition of intestinal bacteria, and UPLC-QTOF-MS-based untargeted metabolomics was used to identify the metabolite profiles. RESULTS Treatment with FTZPs attenuated kidney injury, as indicated by the decreased urinary albumin/creatinine ratio and improved renal architecture. FTZPs downregulated the expression of renal genes associated with inflammation, fibrosis, and systematically blunted related pathways. FTZPs also restored the colonic mucosal barrier and increased the expression of tight junction proteins (E-cadherin). The FMT experiment confirmed the substantial contribution of the FTZPs-reshaped microbiota to relieving DKD symptoms. Moreover, FTZPs elevated the content of short-chain fatty acids (propionic acid and butanoic acid) and increased the level of the SCFAs transporter Slc22a19. Intestinal flora disorders caused by diabetes, including the growth of the genera Weissella, Enterococcus and Akkermansia, were inhibited by FTZPs treatment. Spearman's analysis revealed that these bacteria were positively correlated with indicators of renal damage. CONCLUSION These results show that oral administration of FTZPs, by altering SCFAs levels and the gut microbiome, is a therapeutic strategy for the treatment of DKD.
Collapse
Affiliation(s)
- Tian Lan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China.
| | - Tang Tang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China
| | - Ying Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China
| | - Yingling Duan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China
| | - Qin Yuan
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Wen Liu
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yuqing Ren
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China
| | - Ning Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China
| | - Xuenan Liu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China
| | - Yu Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China
| | - Xinglong Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China
| | - Guifang Jin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou 510006, China.
| |
Collapse
|
12
|
Turek I, Nguyen TH, Galea C, Abad I, Freihat L, Manallack DT, Velkov T, Irving H. Mutations in the Vicinity of the IRAK3 Guanylate Cyclase Center Impact Its Subcellular Localization and Ability to Modulate Inflammatory Signaling in Immortalized Cell Lines. Int J Mol Sci 2023; 24:ijms24108572. [PMID: 37239919 DOI: 10.3390/ijms24108572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Interleukin-1 receptor-associated kinase 3 (IRAK3) modulates the magnitude of cellular responses to ligands perceived by interleukin-1 receptors (IL-1Rs) and Toll-like receptors (TLRs), leading to decreases in pro-inflammatory cytokines and suppressed inflammation. The molecular mechanism of IRAK3's action remains unknown. IRAK3 functions as a guanylate cyclase, and its cGMP product suppresses lipopolysaccharide (LPS)-induced nuclear factor kappa-light-chain-enhancer of activated B cell (NFκB) activity. To understand the implications of this phenomenon, we expanded the structure-function analyses of IRAK3 through site-directed mutagenesis of amino acids known or predicted to impact different activities of IRAK3. We verified the capacity of the mutated IRAK3 variants to generate cGMP in vitro and revealed residues in and in the vicinity of its GC catalytic center that impact the LPS-induced NFκB activity in immortalized cell lines in the absence or presence of an exogenous membrane-permeable cGMP analog. Mutant IRAK3 variants with reduced cGMP generating capacity and differential regulation of NFκB activity influence subcellular localization of IRAK3 in HEK293T cells and fail to rescue IRAK3 function in IRAK3 knock-out THP-1 monocytes stimulated with LPS unless the cGMP analog is present. Together, our results shed new light on the mechanism by which IRAK3 and its enzymatic product control the downstream signaling, affecting inflammatory responses in immortalized cell lines.
Collapse
Affiliation(s)
- Ilona Turek
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC 3552, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC 3552, Australia
| | - Trang H Nguyen
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC 3552, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC 3552, Australia
| | - Charles Galea
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia
| | - Isaiah Abad
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia
| | - Lubna Freihat
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia
| | - David T Manallack
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia
| | - Tony Velkov
- Department of Microbiology, Monash University, Wellington Rd, Clayton, VIC 3800, Australia
| | - Helen Irving
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC 3552, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC 3552, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia
| |
Collapse
|
13
|
Li J, Zheng Z, Liu Y, Zhang H, Zhang Y, Gao J. IRAK-M has effects in regulation of lung epithelial inflammation. Respir Res 2023; 24:103. [PMID: 37029363 PMCID: PMC10082527 DOI: 10.1186/s12931-023-02406-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/22/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Epithelial barrier is important for asthma development by shaping immune responses. Airway expressing-IL-1 receptor-associated kinase (IRAK)-M of Toll-like receptor pathway was involved in immunoregulation of airway inflammation through influencing activities of macrophages and dendritic cells or T cell differentiation. Whether IRAK-M has effect on cellular immunity in airway epithelial cells upon stimulation remains unclear. METHODS We modeled cellular inflammation induced by IL-1β, TNF-α, IL-33, and house dust mite (HDM) in BEAS-2B and A549 cells. Cytokine production and pathway activation were used to reflect the effects of IRAK-M siRNA knockdown on epithelial immunity. Genotyping an asthma-susceptible IRAK-M SNP rs1624395 and measurement of serum CXCL10 levels were performed in asthma patients. RESULTS IRAK-M expression was significantly induced in BEAS-2B and A549 cells after inflammatory stimulation. IRAK-M knockdown increased the lung epithelial production of cytokines and chemokines, including IL-6, IL-8, CXCL10, and CXCL11, at both mRNA and protein levels. Upon stimulation, IRAK-M silencing led to overactivation of JNK and p38 MAPK in lung epithelial cells. While antagonizing JNK or p38 MAPK inhibited increased secretion of CXCL10 in IRAK-M silenced-lung epithelium. Asthma patients carrying G/G genotypes had significantly higher levels of serum CXCL10 than those carrying homozygote A/A. CONCLUSION Our findings suggested that IRAK-M has effect on lung epithelial inflammation with an influence on epithelial secretion of CXCL10 partly mediated through JNK and p38 MAPK pathways. IRAK-M modulation might indicate a new insight into asthma pathogenesis from disease origin.
Collapse
Affiliation(s)
- Jia Li
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Zhoude Zheng
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yi Liu
- Department of Respiratory Medicine, Civil Aviation General Hospital, Beijing, 100123, China
| | - Hongbing Zhang
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Youming Zhang
- Section of Genomic and Environmental Medicine National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Jinming Gao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
14
|
Malter JS. Pin1 and Alzheimer's disease. Transl Res 2023; 254:24-33. [PMID: 36162703 PMCID: PMC10111655 DOI: 10.1016/j.trsl.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/29/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Alzheimer's disease (AD) is an immense and growing public health crisis. Despite over 100 years of investigation, the etiology remains elusive and therapy ineffective. Despite current gaps in knowledge, recent studies have identified dysfunction or loss-of-function of Pin1, a unique cis-trans peptidyl prolyl isomerase, as an important step in AD pathogenesis. Here I review the functionality of Pin1 and its role in neurodegeneration.
Collapse
Affiliation(s)
- James S Malter
- Department of Pathology, UT Southwestern Medical Center, 5333 Harry Hines Blvd, Dallas, TX 75390.
| |
Collapse
|
15
|
Zheng Z, Li J, Cui Y, Wang W, Zhang M, Zhang Y, Bai Y, Ying S, Gao J. IRAK-M Regulates Proliferative and Invasive Phenotypes of Lung Fibroblasts. Inflammation 2023; 46:763-778. [PMID: 36577924 DOI: 10.1007/s10753-022-01772-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/25/2022] [Accepted: 12/07/2022] [Indexed: 12/30/2022]
Abstract
Lung fibroblasts play an important role in subepithelial fibrosis, one feature for airway remodeling. IL-1 receptor-associated kinase (IRAK)-M was shown to involve fibrosis formation in airways and lung through regulation of inflammatory responses. IRAK-M is expressed by lung fibroblasts, whether IRAK-M has direct impact on lung fibroblasts remains unclear. In this investigation, we evaluated in vitro effect of IRAK-M on phenotypes of lung fibroblasts by silencing or overexpressing IRAK-M. Murine lung fibroblasts (MLg) were stimulated with house dust mite (HDM), IL-33, and transforming growth factor (TGF) β1. Techniques of small interfering RNA or expression plasmid were employed to silence or overexpress IRAK-M in MLg fibroblast cells. Proliferation, migration, invasiveness, and fibrosis-related events were evaluated. Significant upregulation of IRAK-M expression in MLg cells was caused by these stimuli. Silencing IRAK-M significantly increased proliferation, migration, and invasiveness of lung fibroblasts regardless of stimulating conditions. By contrast, IRAK-M overexpression significantly inhibited proliferation and motility of MLg lung fibroblasts. IRAK-M overexpression also significantly decreased the expression of fibronectin, collagen I, and α-SMA in MLg cells. Under stimulation with TGFβ1 or IL-33, IRAK-M silencing reduced MMP9 production, while IRAK-M overexpression increased MMP9 production. Modulation of IRAK-M expression affected cytokines production, either decreased or increased expression of TNFα and CXCL10 by the cells regardless of stimulation. Our in vitro data reveal that IRAK-M directly impacts on lung fibroblasts through modulation of cellular motility, release of inflammatory, and fibrotic cytokines of lung fibroblasts. These might suggest a new target by regulation of IRAK-M in slowing airway remodeling.
Collapse
Affiliation(s)
- Zhoude Zheng
- Department of Pulmonary and Critical Care Medicine, Dongcheng District, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Beijing, 100730, China
| | - Jia Li
- Department of Pulmonary and Critical Care Medicine, Dongcheng District, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Beijing, 100730, China
| | - Ye Cui
- Department of Immunology, Capital Medical University, Beijing, 100069, China
| | - Wei Wang
- Department of Immunology, Capital Medical University, Beijing, 100069, China
| | - Mingqiang Zhang
- Department of Pulmonary and Critical Care Medicine, Dongcheng District, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Beijing, 100730, China
- Department of Pulmonary and Critical Care Medicine, Tsinghua Changgung Hospital, Beijing, 102218, China
| | - Youming Zhang
- Section of Genomic and Environmental Medicine, National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Yan Bai
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sun Ying
- Department of Immunology, Capital Medical University, Beijing, 100069, China
| | - Jinming Gao
- Department of Pulmonary and Critical Care Medicine, Dongcheng District, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Beijing, 100730, China.
| |
Collapse
|
16
|
Ottaiano A, Santorsola M, Circelli L, Trotta AM, Izzo F, Perri F, Cascella M, Sabbatino F, Granata V, Correra M, Tarotto L, Stilo S, Fiore F, Martucci N, Rocca AL, Picone C, Muto P, Borzillo V, Belli A, Patrone R, Mercadante E, Tatangelo F, Ferrara G, Di Mauro A, Scognamiglio G, Berretta M, Capuozzo M, Lombardi A, Galon J, Gualillo O, Pace U, Delrio P, Savarese G, Scala S, Nasti G, Caraglia M. Oligo-Metastatic Cancers: Putative Biomarkers, Emerging Challenges and New Perspectives. Cancers (Basel) 2023; 15:cancers15061827. [PMID: 36980713 PMCID: PMC10047282 DOI: 10.3390/cancers15061827] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Some cancer patients display a less aggressive form of metastatic disease, characterized by a low tumor burden and involving a smaller number of sites, which is referred to as "oligometastatic disease" (OMD). This review discusses new biomarkers, as well as methodological challenges and perspectives characterizing OMD. Recent studies have revealed that specific microRNA profiles, chromosome patterns, driver gene mutations (ERBB2, PBRM1, SETD2, KRAS, PIK3CA, SMAD4), polymorphisms (TCF7L2), and levels of immune cell infiltration into metastases, depending on the tumor type, are associated with an oligometastatic behavior. This suggests that OMD could be a distinct disease with specific biological and molecular characteristics. Therefore, the heterogeneity of initial tumor burden and inclusion of OMD patients in clinical trials pose a crucial methodological question that requires responses in the near future. Additionally, a solid understanding of the molecular and biological features of OMD will be necessary to support and complete the clinical staging systems, enabling a better distinction of metastatic behavior and tailored treatments.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Luisa Circelli
- AMES, Centro Polidiagnostico Strumentale SRL, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy
| | - Anna Maria Trotta
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Francesco Izzo
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
| | - Vincenza Granata
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Marco Correra
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Luca Tarotto
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Salvatore Stilo
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Francesco Fiore
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Nicola Martucci
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Antonello La Rocca
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Carmine Picone
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Paolo Muto
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Valentina Borzillo
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Andrea Belli
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Renato Patrone
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Edoardo Mercadante
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Fabiana Tatangelo
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Gerardo Ferrara
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Annabella Di Mauro
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Giosué Scognamiglio
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | | | - Angela Lombardi
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio 7, 80138 Naples, Italy
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Ugo Pace
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Paolo Delrio
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale SRL, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy
| | - Stefania Scala
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Guglielmo Nasti
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio 7, 80138 Naples, Italy
| |
Collapse
|
17
|
Barrio-Hernandez I, Schwartzentruber J, Shrivastava A, Del-Toro N, Gonzalez A, Zhang Q, Mountjoy E, Suveges D, Ochoa D, Ghoussaini M, Bradley G, Hermjakob H, Orchard S, Dunham I, Anderson CA, Porras P, Beltrao P. Network expansion of genetic associations defines a pleiotropy map of human cell biology. Nat Genet 2023; 55:389-398. [PMID: 36823319 PMCID: PMC10011132 DOI: 10.1038/s41588-023-01327-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/30/2023] [Indexed: 02/25/2023]
Abstract
Interacting proteins tend to have similar functions, influencing the same organismal traits. Interaction networks can be used to expand the list of candidate trait-associated genes from genome-wide association studies. Here, we performed network-based expansion of trait-associated genes for 1,002 human traits showing that this recovers known disease genes or drug targets. The similarity of network expansion scores identifies groups of traits likely to share an underlying genetic and biological process. We identified 73 pleiotropic gene modules linked to multiple traits, enriched in genes involved in processes such as protein ubiquitination and RNA processing. In contrast to gene deletion studies, pleiotropy as defined here captures specifically multicellular-related processes. We show examples of modules linked to human diseases enriched in genes with known pathogenic variants that can be used to map targets of approved drugs for repurposing. Finally, we illustrate the use of network expansion scores to study genes at inflammatory bowel disease genome-wide association study loci, and implicate inflammatory bowel disease-relevant genes with strong functional and genetic support.
Collapse
Affiliation(s)
- Inigo Barrio-Hernandez
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Jeremy Schwartzentruber
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
- Wellcome Sanger Institute, Cambridge, UK
| | - Anjali Shrivastava
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Noemi Del-Toro
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Asier Gonzalez
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Qian Zhang
- Wellcome Sanger Institute, Cambridge, UK
| | - Edward Mountjoy
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Daniel Suveges
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - David Ochoa
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Maya Ghoussaini
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Glyn Bradley
- Computational Biology, Genomic Sciences, GSK, Stevenage, UK
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Sandra Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Ian Dunham
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
- Wellcome Sanger Institute, Cambridge, UK
| | - Carl A Anderson
- Open Targets, Cambridge, UK
- Wellcome Sanger Institute, Cambridge, UK
| | - Pablo Porras
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
- Open Targets, Cambridge, UK
| | - Pedro Beltrao
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK.
- Open Targets, Cambridge, UK.
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
18
|
Pereira M, Gazzinelli RT. Regulation of innate immune signaling by IRAK proteins. Front Immunol 2023; 14:1133354. [PMID: 36865541 PMCID: PMC9972678 DOI: 10.3389/fimmu.2023.1133354] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
The Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1R) families are of paramount importance in coordinating the early immune response to pathogens. Signaling via most TLRs and IL-1Rs is mediated by the protein myeloid differentiation primary-response protein 88 (MyD88). This signaling adaptor forms the scaffold of the myddosome, a molecular platform that employs IL-1R-associated kinase (IRAK) proteins as main players for transducing signals. These kinases are essential in controlling gene transcription by regulating myddosome assembly, stability, activity and disassembly. Additionally, IRAKs play key roles in other biologically relevant responses such as inflammasome formation and immunometabolism. Here, we summarize some of the key aspects of IRAK biology in innate immunity.
Collapse
Affiliation(s)
- Milton Pereira
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States,*Correspondence: Milton Pereira, ; Ricardo T. Gazzinelli,
| | - Ricardo T. Gazzinelli
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States,Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil,Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, MG, Brazil,Plataforma de Medicina Translacional, Fundação Oswaldo Cruz, Ribeirão Preto, SP, Brazil,*Correspondence: Milton Pereira, ; Ricardo T. Gazzinelli,
| |
Collapse
|
19
|
Esmael A, Petro TM. IL-33 promotes increased replication of Theiler's Murine Encephalomyelitis Virus in RAW264.7 macrophage cells with an IRF3-dependent response. Virus Res 2023; 323:199007. [PMID: 36414191 PMCID: PMC10194383 DOI: 10.1016/j.virusres.2022.199007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/03/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Interleukin-33 (IL-33), which promotes M2 macrophage development, may influence the control of viruses, such as Theiler's Murine Encephalomyelitis Virus (TMEV) that infect macrophages. Because Interferon Regulatory Factor-3 (IRF3) is also critical to control of TMEV infection in macrophages, information on the relationship between IL-33 and IRF3 is important. Thus, RAW264.7 Lucia murine macrophage lineage cells with an endogenous IRF3-ISRE promoter driving secreted luciferase and IRF3KO RAW Lucia, a subline deficient in IRF3, were challenged with TMEV. After the challenge, considerable TMEV RNA detected at 18 and 24 h in RAW cells was significantly elevated in IRF3KO RAW cells. TMEV induction of ISRE-IRF3 promoter activity, IFN-β and IL-33 gene expression, and IL-6 and IL-10 protein production, which was strong in RAW cells, was less in IRF3KO RAW cells. In contrast, expression of CD206 and ARG1, classical M2 macrophage markers, was significantly elevated in IRF3KO RAW cells. Moreover, RAW and IRF3KO RAW cells produced extracellular IL-33 prior to and after infection with TMEV and antibody blockade of the IL-33 receptor, ST2, reduced CD206 and ARG1 expression, but increased IL-6 gene expression. Pre-treating both RAW and IRF3KO RAW cells with IL-33 prior to challenge significantly increased TMEV infection, but also increased IL-33, IL-10, IL-6 mRNA expression, and NO production without increasing IFN-β. Notably, IL-33 induction of IL-33, IRF3-ISRE promoter activity, and IL-10 by TMEV or poly I:C/IFN-γ was significantly dependent upon IRF3. The results show that the expression of IL-33 and the repression of M2 macrophage phenotypic markers are dependent on IRF3 and that IL-33 decreases the ability of macrophages to control infection with macrophage-tropic viruses.
Collapse
Affiliation(s)
- Ahmed Esmael
- Nebraska Center for Virology, University of Nebraska Lincoln, Lincoln, NE 68583, USA; Botany and Microbiology Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Thomas M Petro
- Nebraska Center for Virology, University of Nebraska Lincoln, Lincoln, NE 68583, USA; Dept. of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, 68583, USA.
| |
Collapse
|
20
|
Li Y, Tang M, Zhang FJ, Huang Y, Zhang J, Li J, Wang Y, Yang J, Zhu S. Screening of ulcerative colitis biomarkers and potential pathways based on weighted gene co-expression network, machine learning and ceRNA hypothesis. Hereditas 2022; 159:42. [PMID: 36419192 PMCID: PMC9685902 DOI: 10.1186/s41065-022-00259-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 11/12/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Ulcerative colitis (UC) refers to an intractable intestinal inflammatory disease. Its increasing incidence rate imposes a huge burden on patients and society. The UC etiology has not been determined, so screening potential biomarkers is critical to preventing disease progression and selecting optimal therapeutic strategies more effectively. METHODS The microarray datasets of intestinal mucosal biopsy of UC patients were selected from the GEO database, and integrated with R language to screen differentially expressed genes and draw proteins interaction network diagrams. GO, KEGG, DO and GSEA enrichment analyses were performed to explore their biological functions. Through machine learning and WGCNA analysis, targets that can be used as UC potential biomarkers are screened out. ROC curves were drawn to verify the reliability of the results and predicted the mechanism of marker genes from the aspects of immune cell infiltration, co-expression analysis, and competitive endogenous network (ceRNA). RESULTS Two datasets GSE75214 and GSE87466 were integrated for screening, and a total of 107 differentially expressed genes were obtained. They were mainly related to biological functions such as humoral immune response and inflammatory response. Further screened out five marker genes, and found that they were associated with M0 macrophages, quiescent mast cells, M2 macrophages, and activated NK cells in terms of immune cell infiltration. The co-expression network found significant co-expression relationships between 54 miRNAs and 5 marker genes. According to the ceRNA hypothesis, NEAT1-miR-342-3p/miR-650-SLC6A14, NEAT1-miR-650-IRAK3, and XIST-miR-342-3p-IRAK3 axes were found as potential regulatory pathways in UC. CONCLUSION This study screened out five biomarkers that can be used for the diagnosis and treatment of UC, namely SLC6A14, TIMP1, IRAK3, HMGCS2, and APOBEC3B. Confirmed that they play a role in the occurrence and development of UC at the level of immune infiltration, and proposed a potential RNA regulatory pathway that controls the progression of UC.
Collapse
Affiliation(s)
- Ying Li
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Jinan, China ,grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, The First College for Clinical Medicine, Jinan, China
| | - Mengyao Tang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, College of Innovation and Research of Traditional Chinese Medicine, Jinan, 250000 China
| | - Feng Jun Zhang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Jinan, China ,grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, The First College for Clinical Medicine, Jinan, China
| | - Yihan Huang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Jinan, China
| | - Jing Zhang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Jinan, China
| | - Junqi Li
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Jinan, China
| | - Yunpeng Wang
- grid.479672.9Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Department of Gastroenterology, Jinan, China
| | - Jinguang Yang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, The First College for Clinical Medicine, Jinan, China
| | - Shu Zhu
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, College of Innovation and Research of Traditional Chinese Medicine, Jinan, 250000 China
| |
Collapse
|
21
|
Kanamoto M, Takahagi S, Aoyama S, Kido Y, Nakanishi M, Naito M, Kanna M, Yamamotoya T, Tanaka A, Hide M, Asano T, Nakatsu Y. The expression of prolyl isomerase Pin1 is expanded in the skin of patients with atopic dermatitis and facilitates
IL
‐33 expression in
HaCaT
cells. J Dermatol 2022; 50:462-471. [PMID: 37006202 DOI: 10.1111/1346-8138.16633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/23/2022]
Abstract
Atopic dermatitis (AD) is attributable to both a genetic predisposition and environmental factors. Among numerous cytokines involved in the pathogenesis of AD, interleukin-33 (IL-33), reportedly escaping exocytotically in response to a scratch, is abundantly expressed in the skin tissues of patients with AD and is postulated to induce inflammatory and autoimmune responses. In this study, we first demonstrated that peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (Pin1), a unique enzyme which isomerizes the proline residues of target proteins, is abundantly expressed in keratinocytes, and that the areas where it is present in the skin tissues of AD patients became expanded due to hyperkeratosis. Thus, we investigated the effects of Pin1 on the regulation of IL-33 expression using the human keratinocyte cell line HaCaT. Interestingly, silencing of the Pin1 gene or treatment with Pin1 inhibitors dramatically reduced IL-33 expressions in HaCaT cells, although Pin1 overexpression did not elevate it. Subsequently, we showed that Pin1 binds to STAT1 and the nuclear factor-kappaB (NF-κB) subunit p65. Silencing the Pin1 gene with small interfering RNAs significantly reduced the phosphorylation of p65, while no marked effects of Pin1 on the STAT1 pathway were detected. Thus, it is likely that Pin1 contributes to increased expression of IL-33 via the NF-κB subunit p65 in HaCaT cells, at least modestly. Nevertheless, further study is necessary to demonstrate the pathogenic roles of Pin1 and IL-33 in AD development.
Collapse
Affiliation(s)
- Mayu Kanamoto
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
- Department of Dermatology, Institute of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Shunsuke Takahagi
- Department of Dermatology, Institute of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Shunya Aoyama
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Yuri Kido
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Mikako Nakanishi
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Miki Naito
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Machi Kanna
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Takeshi Yamamotoya
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Akio Tanaka
- Department of Dermatology, Institute of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
| | - Michihiro Hide
- Department of Dermatology, Institute of Biomedical & Health Sciences Hiroshima University Hiroshima Japan
- Department of Dermatology Hiroshima Citizens Hospital Hiroshima Japan
| | - Tomoichiro Asano
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Yusuke Nakatsu
- Department of Medical Chemistry, Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| |
Collapse
|
22
|
Yan X, Tsuji G, Hashimoto-Hachiya A, Furue M. Galactomyces Ferment Filtrate Potentiates an Anti-Inflammaging System in Keratinocytes. J Clin Med 2022; 11:6338. [PMID: 36362566 PMCID: PMC9657190 DOI: 10.3390/jcm11216338] [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: 09/16/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 12/24/2023] Open
Abstract
Skincare products play a crucial role in preventing the dry skin induced by various causes. Certain ingredients can help to improve the efficacy of skincare products. Galactomyces ferment filtrate (GFF) is such a functional ingredient. Its use originated from the empirical observation that the hands of sake brewers who deal with yeast fermentation retain a beautiful and youthful appearance. Consequently, skincare products based on GFF are widely used throughout the world. Recent studies have demonstrated that GFF activates an aryl hydrocarbon receptor (AHR) and upregulates the expression of filaggrin, a pivotal endogenous source of natural moisturizing factors, in epidermal keratinocytes. It also activates nuclear factor erythroid-2-related factor 2 (NRF2), the antioxidative master transcription factor, and exhibits potent antioxidative activity against oxidative stress induced by ultraviolet irradiation and proinflammatory cytokines, which also accelerate inflammaging. GFF-mediated NRF2 activation downregulates the expression of CDKN2A, which is known to be overexpressed in senescent keratinocytes. Moreover, GFF enhances epidermal terminal differentiation by upregulating the expression of caspase-14, claudin-1, and claudin-4. It also promotes the synthesis of the antiinflammatory cytokine IL-37 and downregulates the expression of proallergic cytokine IL-33 in keratinocytes. In addition, GFF downregulates the expression of the CXCL14 and IL6R genes, which are involved in inflammaging. These beneficial properties might underpin the potent barrier-protecting and anti-inflammaging effects of GFF-containing skin formulae.
Collapse
Affiliation(s)
- Xianghong Yan
- SK-II Science Communications, Kobe Innovation Center, Procter and Gamble Innovation, Kobe 651-0088, Japan
| | - Gaku Tsuji
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, Fukuoka 812-8582, Japan
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Akiko Hashimoto-Hachiya
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
23
|
Guo H, Bossila EA, Ma X, Zhao C, Zhao Y. Dual Immune Regulatory Roles of Interleukin-33 in Pathological Conditions. Cells 2022; 11:cells11203237. [PMID: 36291105 PMCID: PMC9600220 DOI: 10.3390/cells11203237] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/20/2022] Open
Abstract
Interleukin-33 (IL-33), a member of the IL-1 cytokine family and a multifunctional cytokine, plays critical roles in maintaining host homeostasis and in pathological conditions, such as allergy, infectious diseases, and cancer, by acting on multiple types of immune cells and promoting type 1 and 2 immune responses. IL-33 is rapidly released by immune and non-immune cells upon stimulation by stress, acting as an “alarmin” by binding to its receptor, suppression of tumorigenicity 2 (ST2), to trigger downstream signaling pathways and activate inflammatory and immune responses. It has been recognized that IL-33 displays dual-functioning immune regulatory effects in many diseases and has both pro- and anti-tumorigenic effects, likely depending on its primary target cells, IL-33/sST2 expression levels, cellular context, and the cytokine microenvironment. Herein, we summarize our current understanding of the biological functions of IL-33 and its roles in the pathogenesis of various conditions, including inflammatory and autoimmune diseases, infections, cancers, and cases of organ transplantation. We emphasize the nature of context-dependent dual immune regulatory functions of IL-33 in many cells and diseases and review systemic studies to understand the distinct roles of IL-33 in different cells, which is essential to the development of more effective diagnoses and therapeutic approaches for IL-33-related diseases.
Collapse
Affiliation(s)
- Han Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
| | - Elhusseny A. Bossila
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
- Biotechnology Department, Faculty of Agriculture Al-Azhar University, Cairo 11311, Egypt
| | - Xinran Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
| | - Chenxu Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
- Beijing Institute for Stem Cell and Regeneration, Beijing 100101, China
- Correspondence: ; Tel.: +86-10-64807302; Fax: +86-10-64807313
| |
Collapse
|
24
|
Cramer M, Pineda Molina C, Hussey G, Turnquist HR, Badylak SF. Transcriptomic Regulation of Macrophages by Matrix-Bound Nanovesicle-Associated Interleukin-33. Tissue Eng Part A 2022; 28:867-878. [PMID: 35770892 PMCID: PMC9634988 DOI: 10.1089/ten.tea.2022.0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/14/2022] [Indexed: 11/12/2022] Open
Abstract
The innate immune response, particularly the phenotype of responding macrophages, has significant clinical implications in the remodeling outcome following implantation of biomaterials and engineered tissues. In general, facilitation of an anti-inflammatory (M2-like) phenotype is associated with tissue repair and favorable outcomes, whereas pro-inflammatory (M1-like) activation can contribute to chronic inflammation and a classic foreign body response. Biologic scaffolds composed of extracellular matrix (ECM) and, more recently, matrix-bound nanovesicles (MBV) embedded within the ECM are known to direct macrophages toward an anti-inflammatory phenotype and stimulate a constructive remodeling outcome. The mechanisms of MBV-mediated macrophage activation are not fully understood, but interleukin-33 (IL-33) within the MBV appears critical for M2-like activation. Previous work has shown that IL-33 is encapsulated within the lumen of MBV and stimulates phenotypical changes in macrophages independent of its canonical surface receptor stimulation-2 (ST2). In the present study, we used next-generation RNA sequencing to determine the gene signature of macrophages following exposure to MBV with and without intraluminal IL-33. MBV-associated IL-33 instructed an anti-inflammatory phenotype in both wild-type and st2-/- macrophages by upregulating M2-like and downregulating M1-like genes. The repertoire of genes regulated by ST2-independent IL-33 signaling were broadly related to the inflammatory response and crosstalk between cells of both the innate and adaptive immune systems. These results signify the importance of the MBV intraluminal protein IL-33 in stimulating a pro-remodeling M2-like phenotype in macrophages and provides guidance for the designing of next-generation biomaterials and tissue engineering strategies. Impact statement The phenotype of responding macrophages is predictive of the downstream remodeling response to an implanted biomaterial. The clinical impact of macrophage phenotype has motivated studies to investigate the factors that regulate macrophage activation. Matrix-bound nanovesicles (MBV) embedded within the extracellular matrix direct macrophages toward an anti-inflammatory (M2)-like phenotype that is indicative of a favorable remodeling response. Although the mechanisms of MBV-mediated macrophage activation are not fully understood, the intraluminal protein interleukin-33 (IL-33) is clearly a contributing signaling molecule. The present study identifies those genes regulated by MBV-associated IL-33 that promote a pro-remodeling M2-like macrophage activation state and can guide future therapies in regenerative medicine.
Collapse
Affiliation(s)
- Madeline Cramer
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Catalina Pineda Molina
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - George Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery and School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Heth R. Turnquist
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery and School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephen F. Badylak
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery and School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
25
|
Poudel M, Bhattarai PY, Shrestha P, Choi HS. Regulation of Interleukin-36γ/IL-36R Signaling Axis by PIN1 in Epithelial Cell Transformation and Breast Tumorigenesis. Cancers (Basel) 2022; 14:cancers14153654. [PMID: 35954317 PMCID: PMC9367291 DOI: 10.3390/cancers14153654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Members of the interleukin (IL)-1 cytokine family exhibit dual functions in the regulation of inflammation and cancer. Recent studies have shown the critical role of IL-36γ, the newly identified IL-1 family member, in the regulation of cellular processes implicated in the progression of cancer. Therefore, the underlying mechanism of IL-36γ in tumor development is of considerable interest. Here, we identified the pivotal role of IL-36γ in the proliferation of breast cancer cells. Consistently, IL-36γ was found to promote epithelial cell transformation via the activation of c-Fos, c-Jun, and AP-1 transcription factors, followed by the IL36R-mediated MEK/ERK and JNK/c-Jun cascades. Furthermore, our findings demonstrate the critical role of PIN1 in the regulation of IL-36γ-induced mammary gland tumorigenesis. Abstract Given the increasing recognition of the relationship between IL-1 cytokines, inflammation, and cancer, the significance of distinct members of the IL-1 cytokine family in the etiology of cancer has been widely researched. In the present study, we investigated the underlying mechanism of the IL-36γ/IL-36R axis during breast cancer progression, which has not yet been elucidated. Initially, we determined the effects of IL-36γ on the proliferation and epithelial cell transformation of JB6 Cl41 mouse epidermal and MCF7 human breast cancer cells using BrdU incorporation and anchorage-independent growth assays. We found that treatment with IL-36γ increased the proliferation and colony formation of JB6 Cl41 and MCF7 cells. Analysis of the mechanism underlying the neoplastic cell transformation revealed that IL-36γ induced IL-36R-mediated phosphorylation of MEK1/2, ERK1/2, JNK1/2, and c-Jun, resulting in increased c-Fos, c-Jun, and AP-1 activities in JB6 Cl41 and MCF7 cells. Furthermore, the IL-36γ-induced tumorigenic capacity of MCF7 cells was considerably enhanced by PIN1, following MEK/ERK and JNK/c-Jun signaling. Interestingly, blocking PIN1 activity using juglone suppressed the IL-36γ-induced increase in the anchorage-independent growth of 4T1 metastatic mouse breast cancer cells. Finally, in a syngeneic mouse model, IL-36γ-induced tumor growth in the breast mammary gland was significantly inhibited following PIN1 knockout.
Collapse
Affiliation(s)
| | | | | | - Hong Seok Choi
- Correspondence: ; Tel.: +82-622306379; Fax: +82-622225414
| |
Collapse
|
26
|
Xu S, Yuan H. A three-methylation-driven genes based deep learning model for tuberculosis diagnosis in people with and without human immunodeficiency virus co-infection. Microbiol Immunol 2022; 66:317-323. [PMID: 35510555 DOI: 10.1111/1348-0421.12983] [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: 02/28/2022] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 11/29/2022]
Abstract
Improved diagnostic tests for tuberculosis (TB) among people living with human immunodeficiency virus (HIV) are urgently required. We hypothesized that methylation-driven genes of host blood could be used to diagnosis patients co-infected with HIV/TB. In this study, we identified three methylation-driven genes (MDGs) between patients with HIV mono-infection and with HIV/TB co-infection using R package MethylMix. Then, we developed a deep learning model using three MDGs screened which distinguished HIV/TB co-infection from HIV mono-infection with a sensitivity of 95.2% and a specificity of 88.3%. On the two independent datasets, the sensitivity was 80% to 92.8%, respectively; the specificity was 72.7% to 87.5%, respectively. Besides, our deep learning model also accurately classified TB from healthy controls (75.0-100% sensitivity, 91.3-98.1% specificity) and other respiratory disorders (ORDs) (72.7-75.0% sensitivity, 70.9-72.7% specificity). This study will contribute to improve molecular diagnosis for HIV/TB co-infection. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shaohua Xu
- Drug Clinical Trial Center, Gansu Wuwei Tumor Hospital, Wuwei, Gansu, People's Republic of China
| | - Huicheng Yuan
- Drug Clinical Trial Center, Gansu Wuwei Tumor Hospital, Wuwei, Gansu, People's Republic of China
| |
Collapse
|
27
|
Modulation of Inflammatory Cytokine Production in Human Monocytes by cGMP and IRAK3. Int J Mol Sci 2022; 23:ijms23052552. [PMID: 35269704 PMCID: PMC8909980 DOI: 10.3390/ijms23052552] [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] [Received: 02/03/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2022] Open
Abstract
Interleukin-1 receptor-associated kinase-3 (IRAK3) is a critical checkpoint molecule of inflammatory responses in the innate immune system. The pseudokinase domain of IRAK3 contains a guanylate cyclase (GC) centre that generates small amounts of cyclic guanosine monophosphate (cGMP) associated with IRAK3 functions in inflammation. However, the mechanisms of IRAK3 actions are poorly understood. The effects of low cGMP levels on inflammation are unknown, therefore a dose–response effect of cGMP on inflammatory markers was assessed in THP-1 monocytes challenged with lipopolysaccharide (LPS). Sub-nanomolar concentrations of membrane permeable 8-Br-cGMP reduced LPS-induced NFκB activity, IL-6 and TNF-α cytokine levels. Pharmacologically upregulating cellular cGMP levels using a nitric oxide donor reduced cytokine secretion. Downregulating cellular cGMP using a soluble GC inhibitor increased cytokine levels. Knocking down IRAK3 in THP-1 cells revealed that unlike the wild type cells, 8-Br-cGMP did not suppress inflammatory responses. Complementation of IRAK3 knockdown cells with wild type IRAK3 suppressed cytokine production while complementation with an IRAK3 mutant at GC centre only partially restored this function. Together these findings indicate low levels of cGMP form a critical component in suppressing cytokine production and in mediating IRAK3 action, and this may be via a cGMP enriched nanodomain formed by IRAK3 itself.
Collapse
|
28
|
Jia F, Chen L, Fang L, Chen W. IRAK-M deletion aggravates acute inflammatory response and mitochondrial respiratory dysfunction following myocardial infarction: A bioinformatics analysis. J Proteomics 2022; 257:104512. [DOI: 10.1016/j.jprot.2022.104512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/11/2022] [Accepted: 01/31/2022] [Indexed: 10/19/2022]
|
29
|
Poelman H, Ippel H, Gürkan B, Boelens R, Vriend G, Veer CV', Lutgens E, Nicolaes GAF. Structural anomalies in a published NMR-derived structure of IRAK-M. J Mol Graph Model 2021; 111:108061. [PMID: 34837785 DOI: 10.1016/j.jmgm.2021.108061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022]
Abstract
Signaling by Toll-Like Receptors and the Interleukin-1 Receptor (IL1-R) involves intracellular binding of MyD88, followed by assembly of IL1-R Associated Kinases (IRAKs) into the so-called Myddosome. Using NMR, Nechama et al. determined the structure of the IRAK-M death domain monomer (PDBid: 5UKE). With this structure, they performed a docking study to model the location of IRAK-M in the Myddosome. Based on this, they present a molecular basis for selectivity of IRAK-M towards IRAK1 over IRAK2 binding. When we attempted to use 5UKE as a homology modeling template, we noticed that our 5UKE-based models had structural issues, such as disallowed torsion angles and solvent exposed tryptophans. We therefore analyzed the NMR ensemble of 5UKE using structure validation tools and we compared 5UKE with homologous high-resolution X-ray structures. We identified several structural anomalies in 5UKE, including packing issues, frayed helices and improbable side chain conformations. We used Yasara to build a homology model, based on two high resolution death domain crystal structures, as an alternative model for the IRAK-M death domain (atomic coordinates, modeling details and validation are available at https://swift.cmbi.umcn.nl/gv/service/5uke/). Our model agrees better with known death domain structure information than 5UKE and also with the chemical shift data that was deposited for 5UKE.
Collapse
Affiliation(s)
- Hessel Poelman
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Hans Ippel
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Berke Gürkan
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Rolf Boelens
- Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands
| | - Gert Vriend
- BIPS, Poblacion BACO, 5201, Mindoro, Philippines
| | - Cornelis van 't Veer
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Esther Lutgens
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany & German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80539, Munich, Germany
| | - Gerry A F Nicolaes
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
| |
Collapse
|
30
|
Ottaiano A, Santorsola M, Caraglia M, Circelli L, Gigantino V, Botti G, Nasti G. Genetic regressive trajectories in colorectal cancer: A new hallmark of oligo-metastatic disease? Transl Oncol 2021; 14:101131. [PMID: 34034007 PMCID: PMC8144733 DOI: 10.1016/j.tranon.2021.101131] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) originates as consequence of multiple genetic alterations. Some of the involved genes have been extensively studied (APC, TP53, KRAS, SMAD4, PIK3CA, MMR genes) in highly heterogeneous and poly-metastatic cohorts. However, about 10% of metastatic CRC patients presents with an indolent oligo-metastatic disease differently from other patients with poly-metastatic and aggressive clinical course. Which are the genetic dynamics underlying the differences between oligo- and poly-metastatic CRC? The understanding of the genetic trajectories (primary→metastatic) of CRC, in patients selected to represent homogenous clinical models, is crucial to make genotype/phenotype correlations and to identify the molecular events pushing the disease towards an increasing malignant phenotype. This information is crucial to plan innovative therapeutic strategies aimed to reverse or inhibit these phenomena. In the present study, we review the genetic evolution of CRC with the intent to give a developmental perspective on the border line between oligo- and poly-metastatic diseases.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy.
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via L. De Crecchio, 7 80138, Naples, Italy; Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, 83031, Ariano Irpino, Italy
| | - Luisa Circelli
- AMES-Centro Polidiagnostico Strumentale, 80013, Casalnuovo di Napoli, Italy
| | - Valerio Gigantino
- Innovalab scarl, Molecular Biology, Centro Direzionale, isola A2, 80143, Naples, Italy
| | - Gerardo Botti
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| | - Guglielmo Nasti
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| |
Collapse
|
31
|
Furue M, Furue M. OX40L-OX40 Signaling in Atopic Dermatitis. J Clin Med 2021; 10:jcm10122578. [PMID: 34208041 PMCID: PMC8230615 DOI: 10.3390/jcm10122578] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/26/2021] [Accepted: 06/10/2021] [Indexed: 12/17/2022] Open
Abstract
OX40 is one of the co-stimulatory molecules expressed on T cells, and it is engaged by OX40L, primarily expressed on professional antigen-presenting cells such as dendritic cells. The OX40L-OX40 axis is involved in the sustained activation and expansion of effector T and effector memory T cells, but it is not active in naïve and resting memory T cells. Ligation of OX40 by OX40L accelerates both T helper 1 (Th1) and T helper 2 (Th2) effector cell differentiation. Recent therapeutic success in clinical trials highlights the importance of the OX40L-OX40 axis as a promising target for the treatment of atopic dermatitis.
Collapse
Affiliation(s)
- Masutaka Furue
- Department of Dermatology, Kyushu University, Higashiku, Fukuoka 812-8582, Japan
- Correspondence: ; Tel.: +81-90-2518-9125
| | - Mihoko Furue
- 1-19-20 Momochi, Sawara-ku, Fukuoka 814-0006, Japan;
| |
Collapse
|
32
|
Hou X, Yang F, Li A, Zhao D, Ma N, Chen L, Lin S, Lin Y, Wang L, Yan X, Zheng M, Lee TH, Zhou XZ, Lu KP, Liu H. The Pin1-CaMKII-AMPA Receptor Axis Regulates Epileptic Susceptibility. Cereb Cortex 2021; 31:3082-3095. [PMID: 33569579 DOI: 10.1093/cercor/bhab004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 10/28/2020] [Accepted: 12/27/2020] [Indexed: 12/11/2022] Open
Abstract
Pin1 is a unique isomerase that regulates protein conformation and function after phosphorylation. Pin1 aberration contributes to some neurological diseases, notably Alzheimer's disease, but its role in epilepsy is not fully understood. We found that Pin1-deficient mice had significantly increased seizure susceptibility in multiple chemical inducing models and developed age-dependent spontaneous epilepsy. Electrophysiologically, Pin1 ablation enhanced excitatory synaptic transmission to prefrontal cortex (PFC) pyramidal neurons without affecting their intrinsic excitability. Biochemically, Pin1 ablation upregulated AMPA receptors and GluA1 phosphorylation by acting on phosphorylated CaMKII. Clinically, Pin1 was decreased significantly, whereas phosphorylated CaMKII and GluA1 were increased in the neocortex of patients with epilepsy. Moreover, Pin1 expression restoration in the PFC of Pin1-deficient mice using viral gene transfer significantly reduced phosphorylated CaMKII and GluA1 and effectively suppressed their seizure susceptibility. Thus, Pin1-CaMKII-AMPA receptors are a novel axis controlling epileptic susceptibility, highlighting attractive new therapeutic strategies.
Collapse
Affiliation(s)
- Xiaojun Hou
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China.,Fuzhou Children's Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
| | - Fan Yang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Angcheng Li
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Debao Zhao
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Nengjun Ma
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Linying Chen
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China.,The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Suijin Lin
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Yuanxiang Lin
- The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Long Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Xingxue Yan
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Min Zheng
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Tae Ho Lee
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Xiao Zhen Zhou
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Kun Ping Lu
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Hekun Liu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| |
Collapse
|
33
|
Liu O, Xu J, Wang F, Jin W, Zanvit P, Wang D, Goldberg N, Cain A, Guo N, Han Y, Bynum A, Ma G, Wang S, Tang Z, Chen W. Adipose-mesenchymal stromal cells suppress experimental Sjögren syndrome by IL-33-driven expansion of ST2 + regulatory T cells. iScience 2021; 24:102446. [PMID: 33997712 PMCID: PMC8105666 DOI: 10.1016/j.isci.2021.102446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/11/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived mesenchymal stromal cells (ADSCs) play important roles in the alleviation of inflammation and autoimmune diseases. Interleukin-33 (IL-33), a member of the IL-1 family, has been shown to regulate innate and adaptive immunity. However, it is still unknown whether ADSCs regulate immune responses via IL-33. We show here that ADSCs produced IL-33 in response to IL-1β stimulation, which depended on TAK1, ERK, and p38 pathways. ADSCs-derived IL-33 drove the proliferation of CD4+Foxp3+ST2+ regulatory T cells (Tregs) and alleviated experimental autoimmune Sjögren syndrome in mice. Importantly, human ADSCs also produced IL-33 in response to IL-1β. Thus, we have revealed a previously unrecognized immunoregulatory function of ADSCs by IL-33 production in experimental autoimmunity, which may have clinical applications for human immunopathology. Human and mouse ADSCs express IL-33 in response to IL-β stimulation mADSC-derived IL-33 inhibits inflammation in salivary glands in SS model mADSC-derived IL-33 expand ST2+ Tregs in vitro and in SS model
Collapse
Affiliation(s)
- Ousheng Liu
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Junji Xu
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Fu Wang
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
- Dalian Medical University, School of Stomatology, Dalian 116044, China
| | - Wenwen Jin
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Peter Zanvit
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Dandan Wang
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Nathan Goldberg
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Alexander Cain
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Nancy Guo
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Yichen Han
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Andrew Bynum
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Guowu Ma
- Dalian Medical University, School of Stomatology, Dalian 116044, China
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Zhangui Tang
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Wanjun Chen
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
- Corresponding author
| |
Collapse
|
34
|
Targeting Pin1 for Modulation of Cell Motility and Cancer Therapy. Biomedicines 2021; 9:biomedicines9040359. [PMID: 33807199 PMCID: PMC8065645 DOI: 10.3390/biomedicines9040359] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 01/09/2023] Open
Abstract
Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) specifically binds and isomerizes the phosphorylated serine/threonine-proline (pSer/Thr-Pro) motif, which leads to changes in protein conformation and function. Pin1 is widely overexpressed in cancers and plays an important role in tumorigenesis. Mounting evidence has revealed that targeting Pin1 is a potential therapeutic approach for various cancers by inhibiting cell proliferation, reducing metastasis, and maintaining genome stability. In this review, we summarize the underlying mechanisms of Pin1-mediated upregulation of oncogenes and downregulation of tumor suppressors in cancer development. Furthermore, we also discuss the multiple roles of Pin1 in cancer hallmarks and examine Pin1 as a desirable pharmaceutical target for cancer therapy. We also summarize the recent progress of Pin1-targeted small-molecule compounds for anticancer activity.
Collapse
|
35
|
Zhang X, Zhang M, Li L, Chen W, Zhou W, Gao J. IRAK-M knockout promotes allergic airway inflammation, but not airway hyperresponsiveness, in house dust mite-induced experimental asthma model. J Thorac Dis 2021; 13:1413-1426. [PMID: 33841934 PMCID: PMC8024803 DOI: 10.21037/jtd-20-2133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background IL-1 receptor associated-kinase (IRAK)-M, expressed by airway epithelium and macrophages, was shown to regulate acute and chronic airway inflammation exhibiting a biphasic response in an OVA-based animal model. House dust mite (HDM) is a common real-life aeroallergen highly relevant to asthma pathogenesis. The role of IRAK-M in HDM-induced asthma remains unknown. This study was aimed to investigate the effect of IRAK-M on allergic airway inflammation induced by HDM using IRAK-M knockout (KO) mice and the potential underlying mechanisms. Methods IRAK-M KO and wild-type (WT) mice were sensitized and challenged with HDM. The differences in airway inflammation were evaluated 24 hours after the last challenge between the two genotypes of mice using a number of cellular and molecular biological techniques. In vitro mechanistic investigation was also involved. Results Lung expression of IRAK-M was significantly upregulated by HDM in the WT mice. Compared with the WT controls, HDM-treated IRAK-M KO mice showed exacerbated infiltration of inflammatory cells, particularly Th2 cells, in the airways and mucus overproduction, higher epithelial mediators IL-25, IL-33 and TSLP and Th2 cytokines in bronchoalveolar lavage (BAL) fluid. Lung IRAK-M KO macrophages expressed higher percentage of costimulatory molecules OX40L and CD 80 and exhibited enhanced antigen uptake. However, IRAK-M KO didn’t impact the airway hyperreactivity (AHR) indirectly induced by HDM. Conclusions The findings indicate that IRAK-M protects allergic airway inflammation, not AHR, by modifying activation and antigen uptake of lung macrophages following HDM stimulation. Optimal regulation of IRAK-M might indicate an intriguing therapeutic avenue for allergic airway inflammation.
Collapse
Affiliation(s)
- Xudong Zhang
- Departments of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mingqiang Zhang
- Departments of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lun Li
- Departments of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Chen
- Departments of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wexun Zhou
- Departments of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinming Gao
- Departments of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
36
|
Dong R, Xue Z, Fan G, Zhang N, Wang C, Li G, Da Y. Pin1 Promotes NLRP3 Inflammasome Activation by Phosphorylation of p38 MAPK Pathway in Septic Shock. Front Immunol 2021; 12:620238. [PMID: 33717117 PMCID: PMC7947244 DOI: 10.3389/fimmu.2021.620238] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Pin1 is the only known peptidyl-prolyl cis-trans isomerase (PPIase) that can specifically recognize and isomerize the phosphorylated Serine/Threonine-Proline (pSer/Thr-Pro) motif, change the conformation of proteins through protein phosphorylation, thus regulate various cellular processes in the body. Pin1 plays an important role in cancer, Alzheimer’s disease, and autoimmune diseases. However, the specific mechanism of Pin1 regulation in LPS-induced septic shock is unclear. Here, we found that lack of Pin1 reduced shock mortality and organ damage in mice, and NLRP3 inflammasome activation also was reduced in this process. We further confirmed that Pin1 can affect the expression of NLRP3, ASC, Caspase1, and this process can be regulated through the p38 MAPK pathway. We analyzed that p38 MAPK signaling pathway was highly expressed in septic shock and showed a positive correlation with Pin1 in the Gene Expression Omnibus database. We found that Pin1 could affect the phosphorylation of p38 MAPK, have no obvious difference in extracellular signal-regulated kinases (ERK) and Jun-amino-terminal kinase (JNK) signaling. We further found that Pin1 and p-p38 MAPK interacted, but not directly. In addition, Pin1 deficiency inhibited the cleavage of gasdermin D (GSDMD) and promoted the death of macrophages with LPS treatment, and reduced secretion of inflammatory cytokines including IL-1β and IL-18. In general, our results suggest that Pin1 regulates the NLRP3 inflammasome activation by p38 MAPK signaling pathway in macrophages. Thus, Pin1 may be a potential target for the treatment of inflammatory diseases such as septic shock.
Collapse
Affiliation(s)
- Ruijie Dong
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Zhenyi Xue
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Guangyue Fan
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Na Zhang
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Chengzhi Wang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Guangliang Li
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Yurong Da
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW The alarmins, thymic stromal lymphopoietin (TSLP), interleukin (IL)-25 and IL-33, are upstream regulators of T2 (type 2) inflammation and found to be expressed at high levels in airway epithelium of patients with T2 asthma. This review will summarize how alarmins regulate the inflamed asthmatic airways through previously described and newly identified mechanisms. RECENT FINDINGS Alarmins drive allergic and nonallergic asthma through activation of innate lymphoid cell 2 (ILC2), which are a rich source of cytokines such as IL-5 and IL-13, with resulting effects on eosinophilopoeisis and remodelling, respectively. Findings from bronchial allergen challenges have illustrated widespread expression of alarmins and their receptors across many effector cells in airways, and recent studies have emphasized alarmin regulation of CD4 T lymphocytes, eosinophils and basophils, and their progenitors. Furthermore, a link between alarmins and lipid mediators is being uncovered. SUMMARY Alarmins can drive well defined inflammatory pathways through activation of dendritic cells and polarizing T cells to produce type 2 cytokines, as well as they can directly activate many other effector cells that play a central role in allergic and nonallergic asthma. Clinical trials support a central role for TSLP in driving airway inflammation and asthma exacerbations, while ongoing trials blocking IL-33 and IL-25 will help to define their respective role in asthma.
Collapse
|
38
|
Ni H, Pan W, Jin Q, Xie Y, Zhang N, Chen K, Lin T, Lin C, Xie Y, Wu J, Ni P, Wu L. Label-free proteomic analysis of serum exosomes from paroxysmal atrial fibrillation patients. Clin Proteomics 2021; 18:1. [PMID: 33407078 PMCID: PMC7789314 DOI: 10.1186/s12014-020-09304-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Background Atrial fibrillation (AF) is the most common cardiac heterogeneous rhythm disorder. It represents a major cause of mortality and morbidity, mainly related to embolic events and heart failure. Mechanisms of AF are complex and remain incompletely understood. Recent evidence suggests exosomes are membrane-coated objects released by many cell-types. Their presence in body fluids and the variable surface composition and content render them attractive as a mechanism for potential biomarkers. However, the content of serum exosomes of AF patients has not been fully delineated. Methods In this work, the serum exosomes from AF patients and healthy donors were used to compare changes in the exosome protein content. Exosomes were isolated from serum of AF patients and healthy donors and their purity was confirmed by Western blotting assays and transmission electron microscopy (TEM). Label-free LC–MS/MS quantitative proteomic analysis was applied to analyze protein content of serum exosomes. Results A total of 440 exosomal protein groups were identified, differentially expressed proteins were filtrated with fold change ≥ 2.0 (AF/controls protein abundance ratio ≥ 2 or ≤ 0.5) and p value less than 0.05 (p < 0.05), significantly changed in abundance group contains 39 elevated proteins and 18 reduced proteins, while consistent presence/absence expression profile group contains 40 elevated proteins and 75 reduced proteins. Bioinformatic analysis of differential exosomal proteins confirmed the significant enrichment of components involved in the anticoagulation, complement system and protein folding. Parallel-Reaction Monitoring Relative Quantitative Analysis (PRM) further suggested that AF related to complement system and protein folding. Conclusions These results revealed the composition and potential function of AF serum exosomes, thus providing a new perspective on the complement system and protein folding to AF.
Collapse
Affiliation(s)
- Hanwen Ni
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Wenqi Pan
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Qi Jin
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Yucai Xie
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Ning Zhang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Kang Chen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Tianyou Lin
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Changjian Lin
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Yun Xie
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Jiemin Wu
- Faculty of Medical Laboratory Science, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China
| | - Peihua Ni
- Faculty of Medical Laboratory Science, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China.
| | - Liqun Wu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197# Ruijin Rd, Huangpu District, Shanghai, 200025, China.
| |
Collapse
|
39
|
Nguyen TH, Turek I, Meehan-Andrews T, Zacharias A, Irving H. Analysis of interleukin-1 receptor associated kinase-3 (IRAK3) function in modulating expression of inflammatory markers in cell culture models: A systematic review and meta-analysis. PLoS One 2020; 15:e0244570. [PMID: 33382782 PMCID: PMC7774834 DOI: 10.1371/journal.pone.0244570] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/13/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND IRAK3 is a critical modulator of inflammation in innate immunity. IRAK3 is associated with many inflammatory diseases, including sepsis, and is required in endotoxin tolerance to maintain homeostasis of inflammation. The impact of IRAK3 on inflammatory markers such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in cell culture models remains controversial. OBJECTIVE To analyse temporal effects of IRAK3 on inflammatory markers after one- or two-challenge interventions in cell culture models. METHODS A systematic search was performed to identify in vitro cell studies reporting outcome measures of expression of IRAK3 and inflammatory markers. Meta-analyses were performed where sufficient data were available. Comparisons of outcome measures were performed between different cell lines and human and mouse primary cells. RESULTS The literature search identified 7766 studies for screening. After screening titles, abstracts and full-texts, a total of 89 studies were included in the systematic review. CONCLUSIONS The review identifies significant effects of IRAK3 on decreasing NF-κB DNA binding activity in cell lines, TNF-α protein level at intermediate time intervals (4h-15h) in cell lines or at long term intervals (16h-48h) in mouse primary cells following one-challenge. The patterns of TNF-α protein expression in human cell lines and human primary cells in response to one-challenge are more similar than in mouse primary cells. Meta-analyses confirm a negative correlation between IRAK3 and inflammatory cytokine (IL-6 and TNF-α) expression after two-challenges.
Collapse
Affiliation(s)
- Trang Hong Nguyen
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Ilona Turek
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Terri Meehan-Andrews
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Anita Zacharias
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Helen Irving
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| |
Collapse
|
40
|
Furue M. Regulation of Skin Barrier Function via Competition between AHR Axis versus IL-13/IL-4‒JAK‒STAT6/STAT3 Axis: Pathogenic and Therapeutic Implications in Atopic Dermatitis. J Clin Med 2020; 9:E3741. [PMID: 33233866 PMCID: PMC7700181 DOI: 10.3390/jcm9113741] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Atopic dermatitis (AD) is characterized by skin inflammation, barrier dysfunction, and chronic pruritus. As the anti-interleukin-4 (IL-4) receptor α antibody dupilumab improves all three cardinal features of AD, the type 2 cytokines IL-4 and especially IL-13 have been indicated to have pathogenic significance in AD. Accumulating evidence has shown that the skin barrier function is regulated via competition between the aryl hydrocarbon receptor (AHR) axis (up-regulation of barrier) and the IL-13/IL-4‒JAK‒STAT6/STAT3 axis (down-regulation of barrier). This latter axis also induces oxidative stress, which exacerbates inflammation. Conventional and recently developed agents for treating AD such as steroid, calcineurin inhibitors, cyclosporine, dupilumab, and JAK inhibitors inhibit the IL-13/IL-4‒JAK‒STAT6/STAT3 axis, while older remedies such as coal tar and glyteer are antioxidative AHR agonists. In this article, I summarize the pathogenic and therapeutic implications of the IL-13/IL-4‒JAK‒STAT6/STAT3 axis and the AHR axis in AD.
Collapse
Affiliation(s)
- Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; ; Tel.: +81-92-642-5581; Fax: +81-92-642-5600
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, Fukuoka 812-8582, Japan
| |
Collapse
|
41
|
Ottaiano A, Caraglia M, Di Mauro A, Botti G, Lombardi A, Galon J, Luce A, D’Amore L, Perri F, Santorsola M, Hermitte F, Savarese G, Tatangelo F, Granata V, Izzo F, Belli A, Scala S, Delrio P, Circelli L, Nasti G. Evolution of Mutational Landscape and Tumor Immune-Microenvironment in Liver Oligo-Metastatic Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12103073. [PMID: 33096795 PMCID: PMC7589866 DOI: 10.3390/cancers12103073] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/14/2020] [Accepted: 10/18/2020] [Indexed: 12/17/2022] Open
Abstract
Simple Summary About 10% of colorectal cancer patients presents with oligo-metastatic disease. The aim of our study was to assess genetic and immunologic dynamics underlying the oligo-metastatic status, evaluating genotype-phenotype correlations in a clean and homogeneous clinical model of liver-limited metastatic colorectal cancer. We show that loss of KRAS and SMAD4 mutations characterizes the oligo-metastatic disease while a progressive mutational evolution (gain in KRAS, PI3KCA, BRAF and SMAD4) is observed in poly-metastatic evolving disease. Furthermore, high granzyme-B+ T-cells infiltration is found in oligo-metastatic lesions. This study can support innovative strategies to monitor clinical evolution and to induce regressive genetic trajectories in cancer. Abstract Genetic dynamics underlying cancer progression are largely unknown and several genes involved in highly prevalent illnesses (e.g., hypertension, obesity, and diabetes) strongly concur to cancer phenotype heterogeneity. To study genotype-phenotype relationships contributing to the mutational evolution of colorectal cancer (CRC) with a focus on liver metastases, we performed genome profiling on tumor tissues of CRC patients with liver metastatic disease and no co-morbidities. We studied 523 cancer-related genes and tumor-immune microenvironment characteristics in primary and matched metastatic tissues. We observed a loss of KRAS and SMAD4 alterations and a high granzyme-B+ T-cell infiltration when the disease did not progress. Conversely, gain in KRAS, PIK3CA and SMAD4 alterations and scarce granzyme-B+ T-cells infiltration were observed when the tumor evolved towards a poly-metastatic spread. These findings provide novel insights into the identification of tumor oligo-metastatic status, indicating that some genes are on a boundary line between these two clinical settings (oligo- vs. poly-metastatic CRC). We speculate that the identification of these genes and modification of their evolution could be a new approach for anti-cancer therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Department of Abdominal Oncology, SSD-Innovative Therapies for Abdominal Cancers, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.)
- Correspondence: ; Tel.: +39-081-590-3510; Fax: +39-081-771-4224
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (M.C.); (A.L.); (A.L.)
- Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, 83031 Ariano Irpino, Italy
| | - Annabella Di Mauro
- Department of Pathology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (A.D.M.); (G.B.); (F.T.)
| | - Gerardo Botti
- Department of Pathology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (A.D.M.); (G.B.); (F.T.)
| | - Angela Lombardi
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (M.C.); (A.L.); (A.L.)
- Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, 83031 Ariano Irpino, Italy
| | - Jerome Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Université Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, F-75006 Paris, France;
| | - Amalia Luce
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (M.C.); (A.L.); (A.L.)
- Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, 83031 Ariano Irpino, Italy
| | - Luigi D’Amore
- AMES-Centro Polidiagnostico Strumentale, Srl, 80013 Naples, Italy; (L.D.); (G.S.); (L.C.)
| | - Francesco Perri
- Head and Neck Cancer Medical Oncology Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy;
| | - Mariachiara Santorsola
- Department of Abdominal Oncology, SSD-Innovative Therapies for Abdominal Cancers, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.)
| | | | - Giovanni Savarese
- AMES-Centro Polidiagnostico Strumentale, Srl, 80013 Naples, Italy; (L.D.); (G.S.); (L.C.)
| | - Fabiana Tatangelo
- Department of Pathology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (A.D.M.); (G.B.); (F.T.)
| | - Vincenza Granata
- Department of Radiology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy;
| | - Francesco Izzo
- Hepatic Surgery Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.I.); (A.B.)
| | - Andrea Belli
- Hepatic Surgery Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.I.); (A.B.)
| | - Stefania Scala
- Functional Genomics, Istituto Nazionale Tumori, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy;
| | - Paolo Delrio
- Colorectal Abdominal Surgery Division, Istituto Nazionale Tumori, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy;
| | - Luisa Circelli
- AMES-Centro Polidiagnostico Strumentale, Srl, 80013 Naples, Italy; (L.D.); (G.S.); (L.C.)
| | - Guglielmo Nasti
- Department of Abdominal Oncology, SSD-Innovative Therapies for Abdominal Cancers, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.)
| |
Collapse
|
42
|
Kim WJ, Shin HL, Kim BS, Kim HJ, Ryoo HM. RUNX2-modifying enzymes: therapeutic targets for bone diseases. Exp Mol Med 2020; 52:1178-1184. [PMID: 32788656 PMCID: PMC8080656 DOI: 10.1038/s12276-020-0471-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/01/2023] Open
Abstract
RUNX2 is a master transcription factor of osteoblast differentiation. RUNX2 expression in the bone and osteogenic front of a suture is crucial for cranial suture closure and membranous bone morphogenesis. In this manner, the regulation of RUNX2 is precisely controlled by multiple posttranslational modifications (PTMs) mediated by the stepwise recruitment of multiple enzymes. Genetic defects in RUNX2 itself or in its PTM regulatory pathways result in craniofacial malformations. Haploinsufficiency in RUNX2 causes cleidocranial dysplasia (CCD), which is characterized by open fontanelle and hypoplastic clavicles. In contrast, gain-of-function mutations in FGFRs, which are known upstream stimulating signals of RUNX2 activity, cause craniosynostosis (CS) characterized by premature suture obliteration. The identification of these PTM cascades could suggest suitable drug targets for RUNX2 regulation. In this review, we will focus on the mechanism of RUNX2 regulation mediated by PTMs, such as phosphorylation, prolyl isomerization, acetylation, and ubiquitination, and we will summarize the therapeutics associated with each PTM enzyme for the treatment of congenital cranial suture anomalies.
Collapse
Affiliation(s)
- Woo-Jin Kim
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hye-Lim Shin
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Bong-Soo Kim
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hyun-Jung Kim
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hyun-Mo Ryoo
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea.
| |
Collapse
|
43
|
Nakatsu Y, Matsunaga Y, Ueda K, Yamamotoya T, Inoue Y, Inoue MK, Mizuno Y, Kushiyama A, Ono H, Fujishiro M, Ito H, Okabe T, Asano T. Development of Pin1 Inhibitors and their Potential as Therapeutic Agents. Curr Med Chem 2020; 27:3314-3329. [PMID: 30394205 DOI: 10.2174/0929867325666181105120911] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 12/26/2022]
Abstract
The prolyl isomerase Pin1 is a unique enzyme, which isomerizes the cis-trans conformation between pSer/pThr and proline and thereby regulates the function, stability and/or subcellular distribution of its target proteins. Such regulations by Pin1 are involved in numerous physiological functions as well as the pathogenic mechanisms underlying various diseases. Notably, Pin1 deficiency or inactivation is a potential cause of Alzheimer's disease, since Pin1 induces the degradation of Tau. In contrast, Pin1 overexpression is highly correlated with the degree of malignancy of cancers, as Pin1 controls a number of oncogenes and tumor suppressors. Accordingly, Pin1 inhibitors as anti-cancer drugs have been developed. Interestingly, recent intensive studies have demonstrated Pin1 to be responsible for the onset or development of nonalcoholic steatosis, obesity, atherosclerosis, lung fibrosis, heart failure and so on, all of which have been experimentally induced in Pin1 deficient mice. In this review, we discuss the possible applications of Pin1 inhibitors to a variety of diseases including malignant tumors and also introduce the recent advances in Pin1 inhibitor research, which have been reported.
Collapse
Affiliation(s)
- Yusuke Nakatsu
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Yasuka Matsunaga
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Koji Ueda
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Takeshi Yamamotoya
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Yuki Inoue
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Masa-Ki Inoue
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Yu Mizuno
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| | - Akifumi Kushiyama
- The Division of Diabetes and Metabolism, Institute for Adult Diseases, Asahi Life Foundation, Chuo-ku, Tokyo 103-0002, Japan
| | - Hiraku Ono
- Department of Clinical Cell Biology, Chiba University Graduate School of Medicine, Chiba City, Chiba 260-8677, Japan
| | - Midori Fujishiro
- The Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan
| | - Hisanaka Ito
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Takayoshi Okabe
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomoichiro Asano
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima City, Hiroshima 734-8553, Japan
| |
Collapse
|
44
|
Berghi NO, Dumitru M, Vrinceanu D, Ciuluvica RC, Simioniuc-Petrescu A, Caragheorgheopol R, Tucureanu C, Cornateanu RS, Giurcaneanu C. Relationship between chemokines and T lymphocytes in the context of respiratory allergies (Review). Exp Ther Med 2020; 20:2352-2360. [PMID: 32765714 PMCID: PMC7401840 DOI: 10.3892/etm.2020.8961] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Allergic diseases have been classified in the last decades using various theories. The main classes of the newest classification in allergic respiratory diseases focus on the characterization of the endotype (which takes into account biomarkers related to determinant pathophysiological mechanisms) and of the phenotype (based on the description of the disease). Th2, Th1 and Th17 lymphocytes and the type of inflammatory response mediated by them represent the basis for Th2 and non-Th2 endotype classification. In addition, new lymphocytes were also used to characterize allergic diseases: Th9 lymphocytes, Th22 lymphocytes, T follicular helper cells (TFH) lymphocytes and invariant natural killer T (iNKT) lymphocytes. In the last decade, a growing body of evidence focused on chemokines, chemoattractant cytokines, which seems to have an important contribution to the pathogenesis of this pathology. This review presents the interactions between chemokines and Th lymphocytes in the context of Th2/non-Th2 endotype classification of respiratory allergies.
Collapse
Affiliation(s)
- Nicolae Ovidiu Berghi
- Department of Oncologic Dermatology, 'Elias' Emergency University Hospital, 'Carol Davila' University of Medicine and Pharmacy, 011461 Bucharest, Romania
| | - Mihai Dumitru
- Anatomy Department, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Daniela Vrinceanu
- ENT Department, 'Carol Davila' University of Medicine and Pharmacy, 010271 Bucharest, Romania
| | | | - Anca Simioniuc-Petrescu
- ENT Department, 'Carol Davila' University of Medicine and Pharmacy, 010271 Bucharest, Romania
| | - Ramona Caragheorgheopol
- Immunology Laboratory, 'Cantacuzino' National Military-Medical Institute for Research and Development, 050096 Bucharest, Romania
| | - Catalin Tucureanu
- Immunology Laboratory, 'Cantacuzino' National Military-Medical Institute for Research and Development, 050096 Bucharest, Romania
| | - Roxana Sfrent Cornateanu
- Department of Physiopathology and Immunology, 'Carol Davila' University of Medicine and Pharmacy, 041914 Bucharest, Romania
| | - Calin Giurcaneanu
- Department of Oncologic Dermatology, 'Elias' Emergency University Hospital, 'Carol Davila' University of Medicine and Pharmacy, 011461 Bucharest, Romania
| |
Collapse
|
45
|
Zharichenko N, Njoku DB. The Role of Pro-Inflammatory and Regulatory Signaling by IL-33 in the Brain and Liver: A Focused Systematic Review of Mouse and Human Data and Risk of Bias Assessment of the Literature. Int J Mol Sci 2020; 21:ijms21113933. [PMID: 32486265 PMCID: PMC7312033 DOI: 10.3390/ijms21113933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-33 is a member of the IL-1 family of proteins that have multiple roles in organ-specific inflammation. Many studies suggest diagnostic and therapeutic implications of this cytokine. Many studies have reported pro-inflammatory roles for IL-33 in innate immune responses involving the heart and lung. Recent studies also describe pro-inflammatory and regulatory roles for IL-33 in the pathogenesis of brain and liver disorders in addition to regulatory roles for this cytokine in the heart and lung. In this focused systematic review, we will review the literature regarding pro-inflammatory and regulatory effects of IL-33 in the brain and liver. We will also assess the potential risk of bias in the published literature in order to uncover gaps in the knowledge that will be useful for the scientific community. We utilized guidelines set by preferred reporting items for systemic reviews and meta-analyses. The electronic database was PubMed. Eligibility criteria included organ-specific inflammation in mice and humans, organ-specific inflammation in the central nervous and hepatic systems, and IL-33. Outcomes were pro-inflammatory or regulatory effects of IL-33. Risk of bias in individual studies and across studies was addressed by adapting the Cochrane Rob 2.0 tool. We discovered that a source of bias across the studies was a lack of randomization in human studies. Additionally, because the majority of studies were performed in mice, this could be perceived as a potential risk of bias. Regarding the central nervous system, roles for IL-33 in the development and maturation of neuronal circuits were reported; however, exact mechanisms by which this occurred were not elucidated. IL-33 was produced by astrocytes and endothelial cells while IL-33 receptors were expressed by microglia and astrocytes, demonstrating that these cells are first responders for IL-33; however, in the CNS, IL-33 seems to induce Th1 cytokines such as IL-1β and TNF-α chemokines such as RANTES, MCP-1, MIP-1α, and IP-10, as well as nitric oxide. In the liver, similar risks of bias were determined because of the lack of randomized controlled trials in humans and because the majority of studies were performed in mice. Interestingly, the strain of mouse utilized in the study seemed to affect the role of IL-33 in liver inflammation. Lastly, similar to the brain, IL-33 appeared to have ST2-independent regulatory functions in the liver. Our results reveal plausible gaps in what is known regarding IL-33 in the pathogenesis of brain and liver disorders. We highlight key studies in the lung and heart as examples of advancements that likely occurred because of countless basic and translational studies in this area. More research is needed in these areas in order to assess the diagnostic or therapeutic potential of IL-33 in these disorders.
Collapse
Affiliation(s)
- Nika Zharichenko
- Department of Anesthesiology and Critical Care Medicine Johns Hopkins University, Baltimore, MD 21287, USA;
| | - Dolores B. Njoku
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University, The Charlotte R. Bloomberg Childrens Center, 1800 Orleans Street, Suite 6349D, Baltimore, MD 21287, USA
- Correspondence: ; Tel.: +1-410-955-7610
| |
Collapse
|
46
|
Rost J, Muralidharan S, Lee NA. A label-free shotgun proteomics analysis of macadamia nut. Food Res Int 2020; 129:108838. [DOI: 10.1016/j.foodres.2019.108838] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 11/12/2019] [Accepted: 11/18/2019] [Indexed: 12/18/2022]
|
47
|
Emerging Roles of Interleukin-33-responsive Kidney Group 2 Innate Lymphoid Cells in Acute Kidney Injury. Int J Mol Sci 2020; 21:ijms21041544. [PMID: 32102434 PMCID: PMC7073188 DOI: 10.3390/ijms21041544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/22/2020] [Accepted: 02/22/2020] [Indexed: 12/24/2022] Open
Abstract
Interleukin (IL)-33, a member of the IL-1 family of cytokines, is involved in innate and adaptive immune responses. IL-33 triggers pleiotropic immune functions in multiple types of immune cells, which express the IL-33 receptor, ST2. Recent studies have revealed the potential applications of IL-33 for treating acute kidney injury in preclinical animal models. However, IL-33 and IL-33-responding immune cells are reported to exhibit both detrimental and beneficial roles. The IL-33-mediated immunomodulatory functions have been investigated using loss-of-function approaches, such as IL33-deficient mice, IL-33 antagonists, or administration of exogenous IL-33 recombinant protein. This review will discuss the key findings on IL-33-mediated activation of kidney resident group 2 innate lymphoid cells (ILC2s) and summarize the current understanding of the differential functions of endogenous IL-33 and exogenous IL-33 and their potential implications in treating acute kidney injury.
Collapse
|
48
|
Kılıç A, Ameli A, Park JA, Kho AT, Tantisira K, Santolini M, Cheng F, Mitchel JA, McGill M, O'Sullivan MJ, De Marzio M, Sharma A, Randell SH, Drazen JM, Fredberg JJ, Weiss ST. Mechanical forces induce an asthma gene signature in healthy airway epithelial cells. Sci Rep 2020; 10:966. [PMID: 31969610 PMCID: PMC6976696 DOI: 10.1038/s41598-020-57755-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/23/2019] [Indexed: 12/27/2022] Open
Abstract
Bronchospasm compresses the bronchial epithelium, and this compressive stress has been implicated in asthma pathogenesis. However, the molecular mechanisms by which this compressive stress alters pathways relevant to disease are not well understood. Using air-liquid interface cultures of primary human bronchial epithelial cells derived from non-asthmatic donors and asthmatic donors, we applied a compressive stress and then used a network approach to map resulting changes in the molecular interactome. In cells from non-asthmatic donors, compression by itself was sufficient to induce inflammatory, late repair, and fibrotic pathways. Remarkably, this molecular profile of non-asthmatic cells after compression recapitulated the profile of asthmatic cells before compression. Together, these results show that even in the absence of any inflammatory stimulus, mechanical compression alone is sufficient to induce an asthma-like molecular signature.
Collapse
Affiliation(s)
- Ayşe Kılıç
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Asher Ameli
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Physics, Northeastern University, Boston, MA, USA
| | - Jin-Ah Park
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Alvin T Kho
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
| | - Kelan Tantisira
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Marc Santolini
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Centre for Research and Interdisciplinarity (CRI), Paris, F-75014, France
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106, USA
| | - Jennifer A Mitchel
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Maureen McGill
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Michael J O'Sullivan
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Margherita De Marzio
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Amitabh Sharma
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffrey M Drazen
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Jeffrey J Fredberg
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
49
|
Cohen P, Kelsall IR, Nanda SK, Zhang J. HOIL-1, an atypical E3 ligase that controls MyD88 signalling by forming ester bonds between ubiquitin and components of the Myddosome. Adv Biol Regul 2020; 75:100666. [PMID: 31615747 PMCID: PMC7132539 DOI: 10.1016/j.jbior.2019.100666] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
Components of bacteria and viruses activate Toll-Like Receptors in host cells, triggering the formation of the Myddosome and a signalling network that culminates in the production and release of the inflammatory mediators required to combat pathogenic infection. The Myddosome initiates signalling by recruiting and activating five E3 ligases that generate hybrid ubiquitin chains and attach them to components of the Myddosome. These ubiquitin chains act as a scaffold for the recruitment and activation of ubiquitin-binding proteins, which include the "master" protein kinases TAK1 and IKKβ that drive inflammatory mediator production, as well as other proteins like ABIN1 and A20 that restrict activation of the network to prevent the overproduction of these substances that can lead to autoimmunity and organ damage. Here we review recent developments in our understanding of this network, focusing on the unexpected discovery that the E3 ligase HOIL-1 initiates the formation of hybrid ubiquitin chains by forming an ester bond between the first ubiquitin and the protein components of the Myddosome.
Collapse
Affiliation(s)
- Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, United Kingdom.
| | - Ian R Kelsall
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, United Kingdom
| | - Sambit K Nanda
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, United Kingdom
| | - Jiazhen Zhang
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, United Kingdom
| |
Collapse
|
50
|
Acevedo LA, Korson NE, Williams JM, Nicholson LK. Tuning a timing device that regulates lateral root development in rice. JOURNAL OF BIOMOLECULAR NMR 2019; 73:493-507. [PMID: 31407206 PMCID: PMC7141409 DOI: 10.1007/s10858-019-00258-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/18/2019] [Indexed: 06/10/2023]
Abstract
Peptidyl Prolyl Isomerases (PPIases) accelerate cis-trans isomerization of prolyl peptide bonds. In rice, the PPIase LRT2 is essential for lateral root initiation. LRT2 displays in vitro isomerization of a highly conserved W-P peptide bond (104W-P105) in the natural substrate OsIAA11. OsIAA11 is a transcription repressor that, in response to the plant hormone auxin, is targeted to ubiquitin-mediated proteasomal degradation via specific recognition of the cis isomer of its 104W-P105 peptide bond. OsIAA11 controls transcription of specific genes, including its own, that are required for lateral root development. This auxin-responsive negative feedback circuit governs patterning and development of lateral roots along the primary root. The ability to tune LRT2 activity via mutagenesis is crucial for understanding and modeling the role of this bimodal switch in the auxin circuit and lateral root development. We present characterization of the thermal stability and isomerization rates of several LRT2 mutants acting on the OsIAA11 substrate. The thermally stable mutants display activities lower than that of wild-type (WT) LRT2. These include binding diminished but catalytically active P125K, binding incompetent W128A, and binding capable but catalytically incompetent H133Q mutations. Additionally, LRT2 homologs hCypA from human, TaCypA from Triticum aestivum (wheat) and PPIB from E. coli were shown to have 110, 50 and 60% of WT LRT2 activity on the OsIAA11 substrate. These studies identify several thermally stable LRT2 mutants with altered activities that will be useful for establishing relationships between cis-trans isomerization, auxin circuit dynamics, and lateral root development in rice.
Collapse
Affiliation(s)
- Lucila Andrea Acevedo
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, 14853, USA
- Department of Biochemistry and Biophysics and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nathan E Korson
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Justin M Williams
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Linda K Nicholson
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|