1
|
Park Y, Guan X, Han SJ. N-Myc and STAT Interactor is an Endometriosis Suppressor. Int J Mol Sci 2024; 25:8145. [PMID: 39125716 PMCID: PMC11312104 DOI: 10.3390/ijms25158145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
In patients with endometriosis, refluxed endometrial fragments evade host immunosurveillance, developing into endometriotic lesions. However, the mechanisms underlying this evasion have not been fully elucidated. N-Myc and STAT Interactor (NMI) have been identified as key players in host immunosurveillance, including interferon (IFN)-induced cell death signaling pathways. NMI levels are markedly reduced in the stromal cells of human endometriotic lesions due to modulation by the Estrogen Receptor beta/Histone Deacetylase 8 axis. Knocking down NMI in immortalized human endometrial stromal cells (IHESCs) led to elevated RNA levels of genes involved in cell-to-cell adhesion and extracellular matrix signaling following IFNA treatment. Furthermore, NMI knockdown inhibited IFN-regulated canonical signaling pathways, such as apoptosis mediated by Interferon Stimulated Gene Factor 3 and necroptosis upon IFNA treatment. In contrast, NMI knockdown with IFNA treatment activated non-canonical IFN-regulated signaling pathways that promote proliferation, including β-Catenin and AKT signaling. Moreover, NMI knockdown in IHESCs stimulated ectopic lesions' growth in mouse endometriosis models. Therefore, NMI is a novel endometriosis suppressor, enhancing apoptosis and inhibiting proliferation and cell adhesion of endometrial cells upon IFN exposure.
Collapse
Affiliation(s)
- Yuri Park
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Xiaoming Guan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
- Nuclear Receptor, Transcription and Chromatin Biology Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
2
|
Ochoa Bernal MA, Fazleabas AT. The Known, the Unknown and the Future of the Pathophysiology of Endometriosis. Int J Mol Sci 2024; 25:5815. [PMID: 38892003 PMCID: PMC11172035 DOI: 10.3390/ijms25115815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/14/2024] [Accepted: 05/19/2024] [Indexed: 06/21/2024] Open
Abstract
Endometriosis is one of the most common causes of chronic pelvic pain and infertility, affecting 10% of women of reproductive age. A delay of up to 9 years is estimated between the onset of symptoms and the diagnosis of endometriosis. Endometriosis is currently defined as the presence of endometrial epithelial and stromal cells at ectopic sites; however, advances in research on endometriosis have some authors believing that endometriosis should be re-defined as "a fibrotic condition in which endometrial stroma and epithelium can be identified". There are several theories on the etiology of the disease, but the origin of endometriosis remains unclear. This review addresses the role of microRNAs (miRNAs), which are naturally occurring post-transcriptional regulatory molecules, in endometriotic lesion development, the inflammatory environment within the peritoneal cavity, including the role that cytokines play during the development of the disease, and how animal models have helped in our understanding of the pathology of this enigmatic disease.
Collapse
Affiliation(s)
- Maria Ariadna Ochoa Bernal
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA;
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Asgerally T. Fazleabas
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA;
| |
Collapse
|
3
|
Park Y, Guan X, Han SJ. N-Myc and STAT Interactor is an endometriosis suppressor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593227. [PMID: 38766020 PMCID: PMC11100765 DOI: 10.1101/2024.05.08.593227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
In patients with endometriosis, refluxed endometrial fragments evade host immunosurveillance, developing into endometriotic lesions. However, the mechanisms underlying this evasion have not been fully elucidated. N-Myc and STAT Interactor (NMI) have been identified as key players in host immunosurveillance, including interferon (IFN)-induced cell death signaling pathways. NMI levels are markedly reduced in the stromal cells of human endometriotic lesions due to modulation by the Estrogen Receptor beta/Histone Deacetylase 8 axis. Knocking down NMI in immortalized human endometrial stromal cells (IHESCs) led to elevated RNA levels of genes involved in cell-to-cell adhesion and extracellular matrix signaling following IFNA treatment. Furthermore, NMI knockdown inhibited IFN-regulated canonical signaling pathways, such as apoptosis mediated by Interferon Stimulated Gene Factor 3, and necroptosis upon IFNA treatment. In contrast, NMI knockdown with IFNA treatment activated non-canonical IFN-regulated signaling pathways that promote proliferation, including β-Catenin and AKT signaling. Moreover, NMI knockdown in IHESCs stimulated ectopic lesions' growth in mouse endometriosis models. Therefore, NMI is a novel endometriosis suppressor, enhancing apoptosis and inhibiting proliferation and cell adhesion of endometrial cells upon IFN exposure.
Collapse
|
4
|
Huang E, Wang X, Chen L. Regulated Cell Death in Endometriosis. Biomolecules 2024; 14:142. [PMID: 38397379 PMCID: PMC10886833 DOI: 10.3390/biom14020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
Regulated cell death (RCD) represents a distinct mode of cell demise, differing from accidental cell death (ACD), characterized by specific signaling cascades orchestrated by diverse biomolecules. The regular process of cell death plays a crucial role in upholding internal homeostasis, acting as a safeguard against biological or chemical damage. Nonetheless, specific programmed cell deaths have the potential to activate an immune-inflammatory response, potentially contributing to diseases by enlisting immune cells and releasing pro-inflammatory factors. Endometriosis, a prevalent gynecological ailment, remains incompletely understood despite substantial progress in unraveling associated signaling pathways. Its complexity is intricately tied to the dysregulation of inflammatory immune responses, with various RCD processes such as apoptosis, autophagic cell death, pyroptosis, and ferroptosis implicated in its development. Notably, limited research explores the association between endometriosis and specific RCD pathways like pyroptosis and cuproptosis. The exploration of regulated cell death in the context of endometriosis holds tremendous potential for further advancements. This article thoroughly reviews the molecular mechanisms governed by regulated cell death and their implications for endometriosis. A comprehensive understanding of the regulated cell death mechanism in endometriosis has the potential to catalyze the development of promising therapeutic strategies and chart the course for future research directions in the field.
Collapse
Affiliation(s)
| | | | - Lijuan Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (E.H.)
| |
Collapse
|
5
|
Higashi K, Oda S, Fujii M, Nishida F, Matsumoto H, Morise J, Oka S, Nonaka M. Construction of a T7 phage random peptide library by combining seamless cloning with in vitro translation. J Biochem 2023; 175:85-93. [PMID: 37795834 DOI: 10.1093/jb/mvad077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/07/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023] Open
Abstract
T7 phage libraries displaying random peptides are powerful tools for screening peptide sequences that bind to various target molecules. The T7 phage system has the advantage of less biased peptide distribution compared to the M13 phage system. However, the construction of T7 phage DNA is challenging due to its long 36 kb linear DNA. Furthermore, the diversity of the libraries depends strongly on the efficiency of commercially available packaging extracts. To address these issues, we examined the combination of seamless cloning with cell-free translation systems. Seamless cloning technologies have been widely used to construct short circular plasmid DNA, and several recent studies showed that cell-free translation can achieve more diverse phage packaging. In this study, we combined these techniques to construct four libraries (CX7C, CX9C, CX11C and CX13C) with different random regions lengths. The libraries thus obtained all showed diversity > 109 plaque forming units (pfu). Evaluating our libraries with an anti-FLAG monoclonal antibody yielded the correct epitope sequence. The results indicate that our libraries are useful for screening peptide epitopes against antibodies. These findings suggest that our system can efficiently construct T7 phage libraries with greater diversity than previous systems.
Collapse
Affiliation(s)
- Katsuaki Higashi
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Sakiho Oda
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mai Fujii
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Fumiya Nishida
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hayato Matsumoto
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Jyoji Morise
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shogo Oka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Motohiro Nonaka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
6
|
Cho MK, Jin JS, Jo Y, Han JH, Shin S, Bae SJ, Ryu D, Joo J, Park JK, Ha KT. Frankincense ameliorates endometriosis via inducing apoptosis and reducing adhesion. Integr Med Res 2023; 12:100947. [PMID: 37168676 PMCID: PMC10165193 DOI: 10.1016/j.imr.2023.100947] [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: 10/27/2022] [Revised: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 04/05/2023] Open
Abstract
Background Frankincense, a resin derived from trees of the Boswellia genus, has been used as an incense and a type of herbal medicine for treating inflammatory diseases such arthritis, chronic bowel illness, and asthma. While endometriosis is a well-known inflammatory gynecological illness caused by the ectopic attachment and development of uterine tissue over the menstrual cycle, the impact of frankincense on this illness is poorly understood. The purpose of this study was to explore the effects of frankincense on endometriosis. Methods We used a network pharmacological assessment, in vitro and in vivo investigations with a human endometriotic cell line as well as a syngeneic uterine transfer mouse model. High-performance liquid chromatographic analysis was used to compare water-extracted frankincense (Fr) to its reference compounds and validate the sample. Results A network pharmacological analysis suggested a positive effect of Fr on endometriosis. Fr relieved endometriosis by reducing ectopic endometrial adherence and development, according to both in vivo and in vitro models. We suggested that the ER stress/p53-apoptosis and chemokine-migration/adhesion pathways underlie Fr's anti-endometriotic action using RNA sequencing and bioinformatic analysis. Conclusion This study revealed the potential effect of Fr on endometriosis using an experimental investigation. Fr may have the potential to be an effective and safe treatment for endometriosis.
Collapse
Affiliation(s)
- Min Kyoung Cho
- Korean Medical Research Center for Healthy Aging, Pusan National University, Gyeongsangnam-do, Republic of Korea
- Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan, Republic of Korea
| | - Jung-Sook Jin
- Korean Medical Research Center for Healthy Aging, Pusan National University, Gyeongsangnam-do, Republic of Korea
| | - Yunju Jo
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Jung Ho Han
- Korean Medical Research Center for Healthy Aging, Pusan National University, Gyeongsangnam-do, Republic of Korea
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Gyeongsangnam-do, Republic of Korea
| | - Su Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Gyeongsangnam-do, Republic of Korea
| | - Sung-Jin Bae
- Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan, Republic of Korea
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Jongkil Joo
- Department of Korean Obstetrics and Gynecology, Pusan National University Korean Medicine Hospital, Gyeongsangnam-do, Republic of Korea
| | - Jang-Kyung Park
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Busan, Republic of Korea
| | - Ki-Tae Ha
- Korean Medical Research Center for Healthy Aging, Pusan National University, Gyeongsangnam-do, Republic of Korea
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Gyeongsangnam-do, Republic of Korea
| |
Collapse
|
7
|
Efficient antigen delivery by dendritic cell-targeting peptide via nucleolin confers superior vaccine effects in mice. iScience 2022; 25:105324. [PMID: 36304121 PMCID: PMC9593262 DOI: 10.1016/j.isci.2022.105324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/28/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022] Open
Abstract
Efficient delivery of subunit vaccines to dendritic cells (DCs) is necessary to improve vaccine efficacy, because the vaccine antigen alone cannot induce sufficient protective immunity. Here, we identified DC-targeting peptides using a phage display system and demonstrated the potential of these peptides as antigen-delivery carriers to improve subunit vaccine effectiveness in mice. The fusion of antigen proteins and peptides with DC-targeting peptides induced strong antigen-specific IgG responses, even in the absence of adjuvants. In addition, the DC-targeting peptide improved the distribution of antigens to DCs and antigen presentation by DCs. The combined use of an adjuvant with a DC-targeting peptide improved the effectiveness of the vaccine. Furthermore, nucleolin, located on the DC surface, was identified as the receptor for DC-targeting peptide, and nucleolin was indispensable for the vaccine effect of the DC-targeting peptide. Overall, the findings of this study could be useful for developing subunit vaccines against infectious diseases. We successfully identified an efficient DC-targeting peptide using a phage display system Fusion of the peptide improves the efficacy of vaccine even in the absence of adjuvants The peptide improves the distribution of antigens to DCs and antigen presentation by DCs Nucleolin is indispensable for the vaccine effect of the DC-targeting peptide
Collapse
|
8
|
Shi Y, Wei L, Xing L, Wu S, Yue F, Xia K, Zhang D. Sex Difference is a Determinant of Gut Microbes and Their Metabolites SCFAs/MCFAs in High Fat Diet Fed Rats. Curr Microbiol 2022; 79:347. [PMID: 36209302 DOI: 10.1007/s00284-022-03025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 09/04/2022] [Indexed: 11/24/2022]
Abstract
There are gender differences in obesity and related metabolic diseases, but the mechanism of these differences has not been elucidated. Gut microbiota has been recently recognized as a pivotal determinant of obesity and related diseases. The aim of the present study was to investigate sex differences in gut microbiota and its metabolites in an obesity rat model induced by prolonged high-fat-diet (HFD) feeding. In this study, male and female Sprague-Dawley rats were fed normal chow or HFD for 16 weeks (n = 8 for each group). We found that comparing with male rats on HFD (MHFD), female rats on HFD (FHFD) gained more body weight percentage, while had lower body weight gain efficiency and less severity of hepatic steatosis. HFD induced decreased taxon diversity and richness of gut microbiota, and FHFD group had even lower diversity than MHFD group. Among key genera, HFD induced increased Bilophila in male rats but not in female rats. Compared with the MHFD group, FHFD group possessed increases of Akkermansia and Murimonas, and decreases of Acetanaerobacterium, Bacteroides, Bilophila, Blautia and Romboutsia. The levels of total SCFAs in colon contents were increased in tendency in HFD-fed rats of both sexes. FHFD group had increased propionate and decreased ratio of acetate to propionate and butyrate than MHFD group. For MCFAs, HFD induced increases in undecanoic acid and lauric acid in female rats but not in males. In conclusion, HFD induced sex-related alterations in gut microbiome and short/medium-chain fatty acids in rats.
Collapse
Affiliation(s)
- Ying Shi
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Lin Wei
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Lin Xing
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Shanyu Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Fangzhi Yue
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Ke Xia
- Department of Cardiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| | - Dongmei Zhang
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
| |
Collapse
|
9
|
Simón-Gracia L, Kiisholts K, Petrikaitė V, Tobi A, Saare M, Lingasamy P, Peters M, Salumets A, Teesalu T. Homing Peptide-Based Targeting of Tenascin-C and Fibronectin in Endometriosis. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:3257. [PMID: 34947606 PMCID: PMC8708492 DOI: 10.3390/nano11123257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 11/16/2022]
Abstract
The current diagnostic and therapeutic strategies for endometriosis are limited. Although endometriosis is a benign condition, some of its traits, such as increased cell invasion, migration, tissue inflammation, and angiogenesis are similar to cancer. Here we explored the application of homing peptides for precision delivery of diagnostic and therapeutic compounds to endometriotic lesions. First, we audited a panel of peptide phages for the binding to the cultured immortalized endometriotic epithelial 12Z and eutopic stromal HESC cell lines. The bacteriophages displaying PL1 peptide that engages with angiogenic extracellular matrix overexpressed in solid tumors showed the strongest binding to both cell lines. The receptors of PL1 peptide, tenascin C domain C (TNC-C) and fibronectin Extra Domain-B (Fn-EDB), were expressed in both cells. Silver nanoparticles functionalized with synthetic PL1 peptide showed specific internalization in 12Z and HESC cells. Treatment with PL1-nanoparticles loaded with the potent antimitotic drug monomethyl auristatin E decreased the viability of endometriotic cells in 2D and 3D cultures. Finally, PL1-nanoparticless bound to the cryosections of clinical peritoneal endometriotic lesions in the areas positive for TNC-C and Fn-EDB immunoreactivities and not to sections of normal endometrium. Our findings suggest potential applications for PL1-guided nanoparticles in precision diagnosis and therapy of endometriosis.
Collapse
Affiliation(s)
- Lorena Simón-Gracia
- Laboratory of Precision and Nanomedicine, Department of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (L.S.-G.); (A.T.); (P.L.)
| | - Kristina Kiisholts
- Competence Centre on Health Technologies, 50411 Tartu, Estonia; (K.K.); (M.S.); (M.P.); (A.S.)
| | - Vilma Petrikaitė
- Laboratory of Drug Target Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
- Life Sciences Center, Institute of Biotechnology, Vilnius University, 10257 Vilnius, Lithuania
| | - Allan Tobi
- Laboratory of Precision and Nanomedicine, Department of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (L.S.-G.); (A.T.); (P.L.)
| | - Merli Saare
- Competence Centre on Health Technologies, 50411 Tartu, Estonia; (K.K.); (M.S.); (M.P.); (A.S.)
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
| | - Prakash Lingasamy
- Laboratory of Precision and Nanomedicine, Department of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (L.S.-G.); (A.T.); (P.L.)
| | - Maire Peters
- Competence Centre on Health Technologies, 50411 Tartu, Estonia; (K.K.); (M.S.); (M.P.); (A.S.)
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, 50411 Tartu, Estonia; (K.K.); (M.S.); (M.P.); (A.S.)
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
- Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 14152 Stockholm, Sweden
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Department of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (L.S.-G.); (A.T.); (P.L.)
- Center for Nanomedicine, Department of Cell, Molecular and Developmental Biology, University of California at Santa Barbara, Santa Barbara, CA 93106, USA
| |
Collapse
|
10
|
Zhao Y, Wang Q, Xie C, Cai Y, Chen X, Hou Y, He L, Li J, Yao M, Chen S, Wu W, Chen X, Hong A. Peptide ligands targeting FGF receptors promote recovery from dorsal root crush injury via AKT/mTOR signaling. Am J Cancer Res 2021; 11:10125-10147. [PMID: 34815808 PMCID: PMC8581430 DOI: 10.7150/thno.62525] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/10/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Fibroblast growth factor receptors (FGFRs) are key targets for nerve regeneration and repair. The therapeutic effect of exogenous recombinant FGFs in vivo is limited due to their high molecular weight. Small peptides with low molecular weight, easy diffusion, low immunogenicity, and nontoxic metabolite formation are potential candidates. The present study aimed to develop a novel low-molecular-weight peptide agonist of FGFR to promote nerve injury repair. Methods: Phage display technology was employed to screen peptide ligands targeting FGFR2. The peptide ligand affinity for FGFRs was detected by isothermal titration calorimetry. Structural biology-based computer virtual analysis was used to characterize the interaction between the peptide ligand and FGFR2. The peptide ligand effect on axon growth, regeneration, and behavioral recovery of sensory neurons was determined in the primary culture of sensory neurons and dorsal root ganglia (DRG) explants in vitro and a rat spinal dorsal root injury (DRI) model in vivo. The peptide ligand binding to other membrane receptors was characterized by surface plasmon resonance (SPR) and liquid chromatography-mass spectrometry (LC-MS)/MS. Intracellular signaling pathways primarily affected by the peptide ligand were characterized by phosphoproteomics, and related pathways were verified using specific inhibitors. Results: We identified a novel FGFR-targeting small peptide, CH02, with seven amino acid residues. CH02 activated FGFR signaling through high-affinity binding with the extracellular segment of FGFRs and also had an affinity for several receptor tyrosine kinase (RTK) family members, including VEGFR2. In sensory neurons cultured in vitro, CH02 maintained the survival of neurons and promoted axon growth. Simultaneously, CH02 robustly enhanced nerve regeneration and sensory-motor behavioral recovery after DRI in rats. CH02-induced activation of FGFR signaling promoted nerve regeneration primarily via AKT and ERK signaling downstream of FGFRs. Activation of mTOR downstream of AKT signaling augmented axon growth potential in response to CH02. Conclusion: Our study revealed the significant therapeutic effect of CH02 on strengthening nerve regeneration and suggested a strategy for treating peripheral and central nervous system injuries.
Collapse
|
11
|
Cell-Penetrating Peptide and siRNA-Mediated Therapeutic Effects on Endometriosis and Cancer In Vitro Models. Pharmaceutics 2021; 13:pharmaceutics13101618. [PMID: 34683911 PMCID: PMC8541686 DOI: 10.3390/pharmaceutics13101618] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 12/17/2022] Open
Abstract
Gene therapy is a powerful tool for the development of new treatment strategies for various conditions, by aiming to transport biologically active nucleic acids into diseased cells. To achieve that goal, we used highly potential delivery vectors, cell-penetrating peptides (CPPs), as oligonucleotide carriers for the development of a therapeutic approach for endometriosis and cancer. Despite marked differences, both of these conditions still exhibit similarities, like excessive, uncoordinated, and autonomous cellular proliferation and invasion, accompanied by overlapping gene expression patterns. Thus, in the current study, we investigated the therapeutic effects of CPP and siRNA nanoparticles using in vitro models of benign endometriosis and malignant glioblastoma. We demonstrated that CPPs PepFect6 and NickFect70 are highly effective in transfecting cell lines, primary cell cultures, and three-dimensional spheroids. CPP nanoparticles are capable of inducing siRNA-specific knockdown of therapeutic genes, ribonucleotide reductase subunit M2 (RRM2), and vascular endothelial growth factor (VEGF), which results in the reduction of in vitro cellular proliferation, invasion, and migration. In addition, we proved that it is possible to achieve synergistic suppression of endometriosis cellular proliferation and invasion by combining gene therapy and hormonal treatment approaches by co-administering CPP/siRNA nanoparticles together with the endometriosis-drug danazol. We suggest a novel target, RRM2, for endometriosis therapy and as a proof-of-concept, we propose a CPP-mediated gene therapy approach for endometriosis and cancer.
Collapse
|
12
|
Lynch KM, Strain CR, Johnson C, Patangia D, Stanton C, Koc F, Gil-Martinez J, O'Riordan P, Sahin AW, Ross RP, Arendt EK. Extraction and characterisation of arabinoxylan from brewers spent grain and investigation of microbiome modulation potential. Eur J Nutr 2021; 60:4393-4411. [PMID: 34057578 PMCID: PMC8572209 DOI: 10.1007/s00394-021-02570-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/19/2021] [Indexed: 12/17/2022]
Abstract
Purpose Brewers’ spent grain (BSG) represents the largest by-product of the brewing industry. Its utilisation as an animal feed has become less practical today; however, its high fibre and protein content make it a promising untapped resource for human nutrition. BSG contains mainly insoluble fibre. This fibre, along with protein, is trapped with the complex lignocellulosic cell structure and must be solubilised to release components which may be beneficial to health through modulation of the gut microbiota. Methods In this study, the application of a simultaneous saccharification and fermentation process for the extraction and solubilisation of arabinoxylan from BSG is demonstrated. Results Processing of the BSG was varied to modulate the physicochemical and molecular characteristic of the released arabinoxylan. The maximum level of arabinoxylan solubilisation achieved was approximately 21%, compared to the unprocessed BSG which contained no soluble arabinoxylan (AX). Concentration of the solubilised material produced a sample containing 99% soluble AX. Samples were investigated for their microbiome modulating capacity in in-vitro faecal fermentation trials. Many samples promoted increased Lactobacillus levels (approx. twofold). One sample that contained the highest level of soluble AX was shown to be bifidogenic, increasing the levels of this genus approx. 3.5-fold as well as acetate (p = 0.018) and propionate (p < 0.001) production. Conclusion The findings indicate that AX extracted from BSG has prebiotic potential. The demonstration that BSG is a source of functional fibre is a promising step towards the application of this brewing side-stream as a functional food ingredient for human nutrition. Supplementary Information The online version contains supplementary material available at 10.1007/s00394-021-02570-8.
Collapse
Affiliation(s)
- Kieran M Lynch
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - Conall R Strain
- Teagasc Food Research Centre, Fermoy, Co., Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Crystal Johnson
- Teagasc Food Research Centre, Fermoy, Co., Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Dhrati Patangia
- Teagasc Food Research Centre, Fermoy, Co., Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Fermoy, Co., Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Fatma Koc
- Teagasc Food Research Centre, Fermoy, Co., Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jorge Gil-Martinez
- Global Innovation and Technology Centre, Anheuser-Busch InBev nv/sa, Brouwerijplein 1, 3000, Leuven, Belgium
| | - Patrick O'Riordan
- Global Innovation and Technology Centre, Anheuser-Busch InBev nv/sa, Brouwerijplein 1, 3000, Leuven, Belgium
| | - Aylin W Sahin
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Elke K Arendt
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland. .,APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
13
|
Song Y, Joshi NR, Vegter E, Hrbek S, Lessey BA, Fazleabas AT. Establishment of an Immortalized Endometriotic Stromal Cell Line from Human Ovarian Endometrioma. Reprod Sci 2020; 27:2082-2091. [PMID: 32542539 PMCID: PMC7529860 DOI: 10.1007/s43032-020-00228-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/28/2020] [Indexed: 12/18/2022]
Abstract
Endometrial-like stromal cells, one of the main components of endometriotic lesions, are an important in vitro model for studying cellular and molecular mechanisms associated with lesion development in endometriosis. However, the short life span of primary endometriotic stromal cells (Ec-ESCs) limits their use. Human telomerase reverse transcriptase (hTERT) plasmids can be used to develop immortalized cell lines. Here we aimed to establish an endometriotic stromal cell line by hTERT immortalization. Primary Ec-ESCs were obtained from a human ovarian endometriotic cyst. The purity was assessed by morphology and the expression of vimentin, cytokeratin, and human interferon-inducible transmembrane protein 1 (hIFITM1). Cells were infected with hTERT lentiviral vector and selected with hygromycin. hTERT mRNA levels were confirmed by RT-qPCR. Immortalized Ec-ESCs (iEc-ESCs) were characterized by examining the expression of morphological markers and key genes of interest, TP53, estrogen receptor β (ERβ), progesterone receptor (PR), and steroidogenic factor-1 (SF-1). Karyotyping and in vitro decidualization studies were also performed. Ec-ESCs were positive for vimentin and hIFITM1 and negative for cytokeratin, indicating that they were representative of Ec-ESC. The fibroblast-like morphology, expression of TP53, ERβ, PR, and SF-1 did not change before and after hTERT immortalization. iEc-ESCs showed an impaired decidualization response like primary Ec-ESCs when compared to normal eutopic stromal cells. Karyotyping showed that 15/19 cells had normal female karyotype, while 4/19 cells had partial trisomy 11q. Collectively, we successfully established and characterized an immortalized endometriotic stromal cell line. It is potentially useful as an in vitro experimental model to investigate endometriosis biology.
Collapse
Affiliation(s)
- Yong Song
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Niraj R Joshi
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Erin Vegter
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Samantha Hrbek
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Bruce A Lessey
- Center for Fertility, Endocrinology and Menopause, Wake Forest University, Winston-Salem, NC, 27157, USA
| | - Asgerally T Fazleabas
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
14
|
Mao YQ, Han SF, Zhang SL, Zhang ZY, Kong CY, Chen HL, Li ZM, Cai PR, Han B, Wang LS. An approach using Caenorhabditis elegans screening novel targets to suppress tumour cell proliferation. Cell Prolif 2020; 53:e12832. [PMID: 32452127 PMCID: PMC7309951 DOI: 10.1111/cpr.12832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/07/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Tumour cell proliferation requires high metabolism to meet the bioenergetics and biosynthetic needs. Dauer in Caenorhabditis elegans is characterized by lower metabolism, and we established an approach with C elegans to find potential tumour therapy targets. MATERIALS AND METHODS RNAi screening was used to find dauer-related genes, and these genes were further analysed in glp-1(-) mutants for tumour-suppressing testing. The identified tumour-related genes were verified in clinical tumour tissues. RESULTS The lifespan of glp-1(-) mutants was found to be extended by classical dauer formation signalling. Then, 61 of 287 kinase-coding genes in Caenorhabditis elegans were identified as dauer-related genes, of which 27 were found to be homologous to human oncogenes. Furthermore, 12 dauer-related genes were randomly selected for tumour-suppressing test, and six genes significantly extended the lifespan of glp-1(-) mutants. Of these six genes, F47D12.9, W02B12.12 and gcy-21 were newly linked to dauer formation. These three new dauer-related genes significantly suppressed tumour cell proliferation and thus extended the lifespan of glp-1(-) mutants in a longevity- or dauer-independent manner. The mRNA expression profiles indicated that these dauer-related genes trigged similar low metabolism pattern in glp-1(-) mutants. Notably, the expression of homolog gene DCAF4L2/F47D12.9, TSSK6/W02B12.12 and NPR1/gcy-21 was found to be higher in glioma compared with adjacent normal tissue. In addition, the high expression of TSSK6/W02B12.12 and NPR1/gcy-21 correlated with a worse survival in glioma patients. CONCLUSIONS Dauer gene screening in combination with tumour-suppressing test in glp-1(-) mutants provided a useful approach to find potential targets for tumour therapy via suppressing tumour cell proliferation and rewiring tumour cell metabolism.
Collapse
Affiliation(s)
- Yu-Qin Mao
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - San-Feng Han
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Shi-Long Zhang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Zheng-Yan Zhang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Chao-Yue Kong
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Hui-Ling Chen
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Zhan-Ming Li
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Pei-Ran Cai
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Bing Han
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Li-Shun Wang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Hu C, Huang Y, Chen Y. Targeted Modification of the Cationic Anticancer Peptide HPRP-A1 with iRGD To Improve Specificity, Penetration, and Tumor-Tissue Accumulation. Mol Pharm 2019; 16:561-572. [PMID: 30592418 DOI: 10.1021/acs.molpharmaceut.8b00854] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The chimeric peptide HPRP-A1-iRGD, composed of a chemically conjugated tumor-homing/penetration domain (iRGD) and a cationic anticancer peptide domain (HPRP-A1), was used to study the effect of targeted modification to enhance the peptide's specificity, penetration, and tumor accumulation ability. The iRGD domain exhibits tumor-targeting and tumor-penetrating activities by specifically binding to the neuropilin-1 receptor. Acting as a homing/penetration domain, iRGD contributed to enhancing the tumor selectivity, permeability, and targeting of HPRP-A1 by targeted receptor dependence. As the anticancer active domain, HPRP-A1 kills cancer cells by disrupting the cell membrane and inducing apoptosis. The in vitro membrane selectivity toward cancer cells, such as A549 and MDA-MB-23, and human umbilical vein endothelial cells (HUVECs), normal cells, the penetrability assessment in the A549 3D multiple cell sphere model, and the in vivo tumor-tissue accumulation test in the A549 xenograft model indicated that HPRP-A1-iRGD exhibited significant increases in the selectivity toward membranes that highly express NRP-1, the penetration distance in 3D multiple cell spheres, and the accumulation in tumor tissues after intravenous injection, compared with HPRP-A1 alone. The mechanism of the enhanced targeting ability of HPRP-A1-iRGD was demonstrated by the pull-down assay and biolayer interferometry test, which indicated that the chimeric peptide could specifically bind to the neuropilin-1 protein with high affinity. We believe that chemical conjugation with iRGD to increase the specificity, penetration, and tumor-tissue accumulation of HPRP-A1 is an effective and promising approach for the targeted modification of peptides as anticancer therapeutics.
Collapse
Affiliation(s)
- Cuihua Hu
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education , Jilin University , 2699 Qianjin Street , Changchun 130021 , China.,School of Life Sciences , Jilin University , Changchun 130021 , China
| | - Yibing Huang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education , Jilin University , 2699 Qianjin Street , Changchun 130021 , China.,School of Life Sciences , Jilin University , Changchun 130021 , China
| | - Yuxin Chen
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education , Jilin University , 2699 Qianjin Street , Changchun 130021 , China.,School of Life Sciences , Jilin University , Changchun 130021 , China
| |
Collapse
|
16
|
The development of activatable lytic peptides for targeting triple negative breast cancer. Cell Death Discov 2017; 3:17037. [PMID: 29263848 PMCID: PMC5629628 DOI: 10.1038/cddiscovery.2017.37] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/14/2017] [Accepted: 05/22/2017] [Indexed: 12/28/2022] Open
Abstract
Cytolytic peptides are an emerging class of promising cancer therapeutics shown to overcome drug resistance. They eliminate cancer cells via disruption of the phospholipid bilayer of cell membranes, a mechanism that differentiates it from traditional treatments. However, applications of lytic peptides via systematic administration are hampered by nonspecific toxicity. Here, we describe activatable, masked lytic peptides that are conjugated with anionic peptides via a cleavable linker sensitive to matrix metalloproteinases (Ac-w-βA-e8-XPLG*LAG-klUklUkklUklUk-NH2; lower case letters in the sequences represent D-amino-acids, U=Aib, α-aminoisobutyric acid, *cleavage site). The peptides were activated upon being introduced into the triple negative breast cancer cell line MDA-MB-231, which overexpresses secreted matrix metalloproteinases, to selectively cleave the peptide linker. Our results indicate that the activatable design could be applied to improve the targeting ability of lytic peptides.
Collapse
|
17
|
Sakurai Y, Kajimoto K, Harashima H. Anti-angiogenic nanotherapy via active targeting systems to tumors and adipose tissue vasculature. Biomater Sci 2017; 3:1253-65. [PMID: 26261854 DOI: 10.1039/c5bm00113g] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Sophisticated drug delivery systems (DDS) are required for delivering drugs, especially macromolecules such as nucleic acids or proteins, to their sites of action. Therefore it is a prerequisite that future DDS are designed to selectively target a tissue. In this review, we focus on systems that actively target the vasculature in tumors or adipose tissues. For targeting tumor vasculatur, a new strategy referred to as dual-targeting is proposed that uses a combination of a receptor specific ligand and a cell penetrating peptide, which can induce the synergistic enhancement of tissue selectivity under in vivo conditions. A novel pH-sensitive cationic lipid was designed to enhance the endosomal release of encapsulated compounds such as siRNA as well as to improve the stability in blood circulation after intravenous administration. A cyclic RGD peptide is used as an active targeting ligand. For targeting adipose vasculature, prohibitin, which is expressed on the surface of adipose endothelial cells, was targeted with KGGRAKD peptides on the surface of PEGylated nanoparticles. Prohibitin targeted nanoparticles (PTNP) encapsulating Cytochrome c (CytC) can selectively target adipose vasculature by optimizing the lengths of the PEG linkers and can deliver CytC to adipose endothelial cells. PTNP can successfully induce anti-obese effects as well as apoptosis by delivering CytC to the cytosol in endothelial cells. Unexpectedly, the EPR (enhanced permeability and retention) effect, which is usually observed in tumor tissue, was also observed in the adipose vasculature, especially in obese mice, where PEGylated nanoparticles can pass through the endothelial barriers in adipose tissue. We believe that these achievements in active targeting will allow a greatly expanded use of DDS for nanomedicines.
Collapse
Affiliation(s)
- Yu Sakurai
- Faculty of Pharmaceutical Sciences, Hokkaido University, Japan.
| | | | | |
Collapse
|
18
|
Regulation of apoptotic pathways during endometriosis: from the molecular basis to the future perspectives. Arch Gynecol Obstet 2016; 294:897-904. [PMID: 27628753 DOI: 10.1007/s00404-016-4195-6] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 09/06/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE Endometriosis is defined as the presence of endometrial-like endometrial cells, glands and stroma outside the uterus, causing a strong inflammatory-like microenvironment in the affected tissue. This may provoke a breakdown in the peritoneal cavity homeostasis, with the consequent processes of immune alteration, documented by peripheral mononuclear cells recruitment and secretion of inflammatory cytokines in early phases and of angiogenic and fibrogenic cytokines in the late stages of the disease. Considering the pivotal role of interaction between immune and endometriotic cells, in this paper, we aim to shed light about the role of apoptosis pathways in modulating the fine-regulated peritoneal microenvironment during endometriosis. METHODS Narrative overview, synthesizing the findings of literature retrieved from searches of computerized databases. RESULTS In normal conditions, endometriotic cells, refluxed through the fallopian tubes into the peritoneal cavity, should be attacked and removed by phagocytes and NK cells. During endometriosis, the breakdown of peritoneal homeostasis causes the failure of scavenging mechanisms, allowing the survival of endometriotic cells. The consequent so-called "immunoescaping" of endometriotic cells could be due, at least in part, to the reduction of apoptotic-mediated pathways previously described. CONCLUSION Considering the large amount of evidence retrieved from in vitro as well as in vivo models, the reduced apoptosis of endometriotic cells together with the increased apoptosis of peritoneal fluid mononuclear cells may address the peritoneal homeostasis to a permissive environment for the progression of the disease.
Collapse
|
19
|
Song Y, Fu J, Zhou M, Xiao L, Feng X, Chen H, Huang W. Activated Hippo/Yes-Associated Protein Pathway Promotes Cell Proliferation and Anti-apoptosis in Endometrial Stromal Cells of Endometriosis. J Clin Endocrinol Metab 2016; 101:1552-61. [PMID: 26977530 PMCID: PMC4880175 DOI: 10.1210/jc.2016-1120] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CONTEXT The imbalance in cell proliferation and apoptosis is considered an important role in the pathogenesis of endometriosis, but the exact mechanisms remains unclear. A newly established signaling pathway–Hippo/Yes-associated protein (YAP) pathway plays a critical role in the proliferation and apoptosis processes. However, studies focusing on Hippo/YAP pathway and endometriosis are lacking. OBJECTIVE The objective was to explore the function of the Hippo/YAP pathway in endometriosis. SETTING AND DESIGN The expression of YAP was first investigated in endometrium of women with or without endometriosis. The role of YAP in cell proliferation and apoptosis is identified by transfection of endometrial stromal cells (ESCs) in vitro, subsequent Verteporfin treatments in eutopic ESCs in vitro, and endometriosis animal model of nude mice in vivo. RESULTS Our results revealed that increased expression of YAP and decreased expression of p-YAP in ectopic and eutopic endometrium compared with normal endometrium. YAP knockdown in eutopic ESCs decreased cell proliferation and enhanced cell apoptosis companied with decreased expression of TEAD1, CTGF, and B-cell lymphoma/leukemia (BCL)-2; whereas overexpression of YAP resulted in increased proliferation and decreased apoptosis of normal ESCs with increased expression of TEAD1, CTGF, and BCL-2. By chromatin immunoprecipitation qPCR CTGF and BCL-2 were identified as directly downstream target genes of YAP-TEAD1 active complex. Eutopic ESCs treated with Verteporfin revealed decreased proliferation and enhanced apoptosis whereas in endometriosis animal models of nude mice treated with Verteporfin, the size of endometriotic lesions was significantly reduced. CONCLUSIONS Our study suggests that the Hippo/YAP-signaling pathway plays a critical role in the pathogenesis of endometriosis and should present a novel therapeutic method against endometriosis.
Collapse
Affiliation(s)
- Yong Song
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu Sichuan 610041, People's Republic of China
| | - Jing Fu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu Sichuan 610041, People's Republic of China
| | - Min Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu Sichuan 610041, People's Republic of China
| | - Li Xiao
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu Sichuan 610041, People's Republic of China
| | - Xue Feng
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu Sichuan 610041, People's Republic of China
| | - Hengxi Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu Sichuan 610041, People's Republic of China
| | - Wei Huang
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu Sichuan 610041, People's Republic of China
| |
Collapse
|
20
|
Abstract
Endometriosis is a disease characterized by the growth of endometrial tissue outside the uterus and is associated with chronic pelvic pain. Peritoneal fluid (PF) of women with endometriosis is a dynamic milieu and is rich in inflammatory markers, pain-inducing prostaglandins prostaglandin E2 and prostaglandin F2α, and lipid peroxides; and the endometriotic tissue is innervated with nociceptors. Our clinical study showed that the abundance of oxidatively modified lipoproteins in the PF of women with endometriosis and the ability of antioxidant supplementation to alleviate endometriosis-associated pain. We hypothesized that oxidatively modified lipoproteins present in the PF are the major source of nociceptive molecules that play a key role in endometriosis-associated pain. In this study, PF obtained from women with endometriosis or control women were used for (1) the detection of lipoprotein-derived oxidation-sensitive pain molecules, (2) the ability of such molecules to induce nociception, and (3) the ability of antioxidants to suppress this nociception. LC-MS/MS showed the generation of eicosanoids by oxidized-lipoproteins to be similar to that seen in the PF. Oxidatively modified lipoproteins induced hypothermia (intracerebroventricular) in CD-1 mice and nociception in the Hargreaves paw withdrawal latency assay in Sprague-Dawley rats. Antioxidants, vitamin E and N-acetylcysteine, and the nonsteroidal anti-inflammatory drug indomethacin suppressed the pain-inducing ability of oxidatively modified lipoproteins. Treatment of human endometrial cells with oxidatively modified lipoproteins or PF from women with endometriosis showed upregulation of similar genes belonging to opioid and inflammatory pathways. Our finding that oxidatively modified lipoproteins can induce nociception has a broader impact not only on the treatment of endometriosis-associated pain but also on other diseases associated with chronic pain.
Collapse
|
21
|
Borrelli GM, Kaufmann AM, Abrão MS, Mechsner S. Addition of MCP-1 and MIP-3β to the IL-8 appraisal in peritoneal fluid enhances the probability of identifying women with endometriosis. J Reprod Immunol 2015; 109:66-73. [PMID: 25704572 DOI: 10.1016/j.jri.2015.01.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 12/22/2014] [Accepted: 01/08/2015] [Indexed: 02/07/2023]
Abstract
Chemokines have been associated with endometriosis. Our study was aimed at evaluating the levels of six chemokines--CXCL8 (IL-8), CXCL12 (SDF-1), CCL2 (MCP-1), CCL5 (RANTES), CCL19 (MIP-3β), and CCL21 (6-Ckine)--in the peritoneal fluid (PF) of patients with and controls without endometriosis by multiplexed cytokine assay. In this retrospective case-control study conducted at the Charité University Hospital, patients (n = 36) and controls (n = 27) were enrolled. The patients were separated into groups according to stage of the disease: I-II (n = 21), III-IV (n = 1 5), and according to clinical findings: peritoneal endometriosis (PE; n = 7), deep-infiltrating endometriosis (DIE) affecting the retrocervical area (n = 13) or the bowel/rectovaginal site (n = 14). The subjects were also separated according to the cycle phase: follicular (n = 14) or luteal (n = 8) and the previous use (n = 25) or not (n = 38) of hormones. PF was collected from all subjects (n = 63) consecutively during laparoscopy. The concentration of chemokines in the PF was assessed using Luminex(®) x-MAP(®) technology. Sensitivity and specificity were calculated. A model of multiple logistic regressions estimated the odds of endometriosis for each combination of the chemokines detected. We observed significantly higher concentrations of IL-8 (p < 0.001), MCP-1 (p = 0.014), and MIP-3β (p = 0.022) in the PF of women with endometriosis than in the controls. A joint evaluation revealed that elevated levels of the three chemokines had a positive endometriosis prediction value of 89.1%. The combined assessment of MCP-1, MIP-3β, and IL-8 concentration in PF improved the likelihood of identifying patients with endometriosis. Future studies should investigate this panel in peripheral blood samples.
Collapse
Affiliation(s)
- G M Borrelli
- Charité Universitätsmedizin Berlin - Gynecology Clinic, Campus Benjamin Franklin, Berlin, Germany; University of Sao Paulo, Faculty of Medicine - Department of Obstetrics and Gynecology, Brazil.
| | - A M Kaufmann
- Charité Universitätsmedizin Berlin - Gynecology Clinic, Campus Benjamin Franklin, Berlin, Germany
| | - M S Abrão
- University of Sao Paulo, Faculty of Medicine - Department of Obstetrics and Gynecology, Brazil
| | - S Mechsner
- Charité Universitätsmedizin Berlin - Gynecology Clinic, Campus Benjamin Franklin, Berlin, Germany.
| |
Collapse
|