1
|
Wei D, Liu J, Hu J, Zhang B, Pan Y, Xia Q, Wang F. An NF-κB-regulated cytokine enhances the antiviral resistance of silkworm, Bombyx mori. INSECT MOLECULAR BIOLOGY 2025; 34:426-439. [PMID: 39680673 DOI: 10.1111/imb.12980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024]
Abstract
Insect NF-κB-like factor, Relish, is activated by viral infection and induces the production of antiviral proteins. In this study, we performed a transcriptomic analysis of BmE cells expressing the active form of BmRelish (BmRelishact) and identified BmVago-like as the most strongly-induced secreted-protein. Expression of BmVago-like was specifically triggered by Bombyx mori Nucleo Polyhedro Virus (BmNPV) infection and regulated by BmSTING-BmRelish pathway. Incubating the fresh culture of cells with supernatant medium of BmVago-like expressing cells or recombinant BmVago-like protein (rBmVago-like) significantly increased antiviral resistance. On the contrary, reducing the expression of Bmvago-like by RNA interference (RNAi) in BmE cells as well as in silkworm larvae impaired antiviral response. Furthermore, we constructed transgenic silkworm line over-expressing BmVago-like (BmVago-likeOV) and found they had markedly lower viral load and higher survival rate after BmNPV infection compared with the wild-type control. Co-immunoprecipitation assay showed Bmintegrin β1 interacts with BmVago-like and it was involved in BmVago-like mediated antiviral response. Finally, we found the expression level of signalling molecules in the JAK-STAT pathway increased in rBmVago-like-treated cells and BmVago-likeOV silkworm larvae but decreased in RNAi-treated cells. In summary, our research uncovered an inducible antiviral response in silkworm mediated by cytokine BmVago-like, which is the downstream effector of BmSTING-BmRelish pathway and functions as an antiviral cytokine.
Collapse
Affiliation(s)
- Dongmei Wei
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| | - Jinming Liu
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| | - Jie Hu
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| | - Beilei Zhang
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| | - Yumeng Pan
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| | - Fei Wang
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| |
Collapse
|
2
|
Abulghasem EA, Price CA. The influence of CCN family proteins on ovarian physiology and pathology. Reprod Fertil Dev 2025; 37:RD24199. [PMID: 40359309 DOI: 10.1071/rd24199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The CCN family of proteins is comprised of six matricellular proteins known to regulate multiple cellular processes such as adhesion, proliferation, differentiation, and apoptosis. CCN proteins are known to function through the binding of integrin receptors and through the regulation of growth factors and cytokines in the context of cardiovascular and skeletal development, injury repair, fibrosis, inflammation and cancer. The expression and roles of several CCNs, particularly CCN1 and CCN2, have been investigated in the ovary as they are effectors of the Hippo signaling pathway, and their role in the development of ovarian fibrosis has been described. Here we review the patterns of expression of CCN1-6 in the ovarian follicle, and the role of CCN2 in follicle development and steroidogenesis, and the expression and potential actions of CCN1-6 in ovarian cancers. We highlight the roles CCNs may play in inflammatory processes, and put forth a case for CCN involvement in the process of ovulation.
Collapse
Affiliation(s)
- El Arbi Abulghasem
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, Canada
| | - Christopher A Price
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, Canada
| |
Collapse
|
3
|
Fan H, Zhao H, Gao L, Dong Y, Zhang P, Yu P, Ji Y, Chen ZS, Liang X, Chen Y. CCN1 Enhances Tumor Immunosuppression through Collagen-Mediated Chemokine Secretion in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500589. [PMID: 40287974 DOI: 10.1002/advs.202500589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/03/2025] [Indexed: 04/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense, immunosuppressive tumor microenvironment (TME) that limits therapeutic efficacy. This study investigates the role of cellular communication network factor 1 (CCN1, also known as Cyr61), an extracellular matrix-associated protein, in modulating the TME of PDAC. It is demonstrated that Ccn1 promotes PDAC progression by upregulating collagen and chemokine expression, thereby facilitating immune cell exclusion and enhancing tumor growth. Using a Ccn1-deficient PDAC model, decreased collagen and chemokine levels are observed, resulting in increased infiltration of cytotoxic immune cells and reduced myeloid-derived suppressor cells (MDSCs). Furthermore, Ccn1-deficient tumors exhibit heightened sensitivity to gemcitabine and show enhanced responsiveness to anti-programmed cell death 1 (anti-PD1) therapy. Mechanistically, Ccn1 regulates chemokine production through collagen expression, with chemokine levels remaining suppressed even upon interferon-gamma treatment in collagen-deficient cells. These findings highlight Ccn1 as a potential therapeutic target that reprograms the TME to enhance the efficacy of both chemotherapy and immunotherapy in PDAC, providing a novel approach for overcoming immune resistance in PDAC.
Collapse
Affiliation(s)
- Hongjie Fan
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Huzi Zhao
- Department of Pathology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, China
| | - Lili Gao
- Department of Pathology, Xinhua Hospital Affiliated to Medicine School of Shanghai Jiaotong University, Shanghai, 200082, China
| | - Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100006, China
| | - Pei Zhang
- Department of Mathematics, University of Maryland, College Park, Maryland, MD 20742, USA
| | - Pengfei Yu
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Yunfei Ji
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Xinmiao Liang
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Yang Chen
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| |
Collapse
|
4
|
Guo X, Li S. Bone metastases of prostate cancer: Molecular mechanisms, targeted diagnosis and targeted therapy (Review). Oncol Rep 2025; 53:46. [PMID: 39981932 PMCID: PMC11865881 DOI: 10.3892/or.2025.8879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/12/2024] [Indexed: 02/22/2025] Open
Abstract
Prostate cancer (PCa) is second only to lung cancer in terms of death among men worldwide. Advanced PCa frequently results in bone metastases, which occur in ~90% of patients and frequently result in severe skeleton‑related events. Currently, the treatment for this disease is limited to alleviating its clinical symptoms and cannot provide a complete cure. Therefore, the development of novel treatment strategies is particularly important. In recent years, numerous novel strategies for the diagnosis and treatment of PCa have emerged, resulting in good clinical efficacy. For example, strategies targeting prostate specific membrane antigen, poly ADP‑ribose polymerase and programmed cell death protein 1 have been applied to PCa‑induced bone metastasis, and have shown initial efficacy and great potential. Therefore, understanding the molecular mechanisms underlying the formation of bone metastases in patients with PCa is of importance for the effective management of this disease. The purpose of the present review is to comprehensively outline the roles of protein‑coding genes and non‑coding RNAs in the development of bone metastases of PCa to elucidate their significance in the management of PCa. The aim is to offer clinicians and researchers a comprehensive understanding of this topic.
Collapse
Affiliation(s)
- Xutang Guo
- Department of Urology, Gansu Province Maternity and Child Health Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Shaojun Li
- Department of Urology, Gansu Province Maternity and Child Health Hospital, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
5
|
Taheri M, Seirafianpour F, Fallahian A, Hosseinzadeh A, Reiter RJ, Mehrzadi S. Exploring melatonin's signalling pathways in the protection against age-related skin deterioration. Pharmacol Rep 2025; 77:375-391. [PMID: 39883394 DOI: 10.1007/s43440-025-00699-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/15/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
Melatonin, renowned for regulating sleep-wake cycles, also exhibits notable anti-aging properties for the skin. Synthesized in the pineal gland and various tissues including the skin, melatonin's efficacy arises from its capacity to combat oxidative stress and shield the skin from ultraviolet (UV)-induced damage. Moreover, it curbs melanin production, thereby potentially ameliorating hyperpigmentation. The presence of melatonin receptors in diverse skin cell types and its documented ability to enhance skin tone, hydration, and texture upon topical administration underscores its promise as an anti-aging agent. Melatonin's protective effects likely emanate from its multifaceted characteristics, encompassing antioxidant, anti-inflammatory, and immunomodulatory functions, as well as its influence on collagen synthesis and mitochondrial activity. Chronic inflammation and oxidative stress initiate a detrimental feedback loop. Reactive oxygen species (ROS), notorious for damaging cellular structures, provoke immune responses by oxidizing vital molecules and activating signaling proteins. This triggers heightened expression of inflammatory genes, perpetuating the cycle. Such dysregulation significantly compromises the body's resilience against infections and other health adversities. This study embarks on an exploration of the fundamental signaling pathways implicated in skin aging. Furthermore, it delves into the therapeutic potential of melatonin and its anti-aging attributes within the realm of skin health.
Collapse
Affiliation(s)
- Maryam Taheri
- Medical School, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Amirali Fallahian
- Department of Dermatology, School of Medicine, Rasool Akram Medical Complex, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Azam Hosseinzadeh
- Razi Drug Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX, United States
| | - Saeed Mehrzadi
- Razi Drug Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
6
|
Byrd CE, Schramm JE, Yang J, Barnes AE, Griffiths M, Ambade AS, Rosen DT, Cubero Salazar IM, Simpson CE, Tedford RJ, Hsu S, Vaidya D, Kolb TM, Pauciulo MW, Nichols WC, Ivy DD, Austin ED, Hassoun PM, Damico RL, Everett AD. Cellular Communication Network Protein 2 in the Right Ventricle of Pulmonary Arterial Hypertension. Pulm Circ 2025; 15:e70067. [PMID: 40177499 PMCID: PMC11961549 DOI: 10.1002/pul2.70067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025] Open
Abstract
Cellular communication network 2 (CCN2) is a secreted matricellular protein associated with pulmonary arterial hypertension (PAH) but has not been studied relative to PAH severity, outcomes, or right ventricle (RV) structure and function in a large human cohort and preclinical animal model. This study assessed the associations between CCN2 and PAH severity, survival, hemodynamic measurements, and cardiovascular dysfunction. Serum CCN2 levels were compared in 2548 adults with PAH and 216 controls. CCN2 levels in PAH patients were compared to functional and hemodynamic measurements, and survival outcomes. RV-pulmonary artery coupling and RV morphology were also assessed in a small subset of patients via pressure-volume loops and cardiac magnetic resonance imaging. In a preclinical PAH model, plasma CCN2 levels were compared between ventricles with PAH progression. CCN2 mRNA levels in both ventricles in the preclinical model were measured to compare with morphologic histologic variables. CCN2 serum levels were significantly higher in PAH compared to controls (p < 0.0001). Higher CCN2 levels were associated with reduced RV contractility (p = 0.003). Higher CCN2 levels were associated with worse 6MWD (p = 0.035), and higher risk of mortality or transplant (p = 0.025). In the preclinical model, prepulmonary CCN2 plasma levels increased with the progression of disease. CCN2 mRNA levels in the RV were associated with decreased RV capillary density (p = 0.015) and increased RV fibrosis (p = 0.045). Though more investigation is needed, it appears that CCN2 plays a role in the development of PAH and potentially in RV maladaptation in PAH.
Collapse
Affiliation(s)
- Carly E. Byrd
- Department of Pediatrics, Division of Neonatal‐Perinatal MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jennifer E. Schramm
- Department of Anesthesiology and Critical Care Medicine, Division of Pediatric Cardiac Critical CareJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jun Yang
- Department of Pediatrics, Division of Pediatric CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Allan E. Barnes
- Department of Pediatrics, Division of Pediatric CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Megan Griffiths
- Department of Pediatrics, UT Southwestern Medical CenterDivision of Pediatric CardiologyDallasTexasUSA
| | - Anjira S. Ambade
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Darin T. Rosen
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | | | - Catherine E. Simpson
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Ryan J. Tedford
- Department of Medicine, Division of CardiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Steven Hsu
- Department of Medicine, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Dhananjay Vaidya
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
- Johns Hopkins School of MedicineDivision of General Internal MedicineBaltimoreMarylandUSA
| | - Todd M. Kolb
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Michael W. Pauciulo
- Cincinnati Children's Hospital Medical CenterDivision of Human GeneticsCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - William C. Nichols
- Cincinnati Children's Hospital Medical CenterDivision of Human GeneticsCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - David D. Ivy
- Department of Pediatric CardiologyChildren's Hospital ColoradoDenverColoradoUSA
| | - Eric D. Austin
- Department of PediatricsVanderbilt University Medical Center, Division of Allergy, Immunology, and Pulmonary MedicineNashvilleTennesseeUSA
| | - Paul M. Hassoun
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Rachel L. Damico
- Division of Pulmonary, Critical Care and Sleep MedicineUniversity of MiamiMiamiFloridaUSA
| | - Allen D. Everett
- Department of Pediatrics, Division of Pediatric CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
7
|
Perbal B. The case of Connective Tissue Growth Factor (CTGF) and the pit of misleading and improper nomenclatures. J Cell Commun Signal 2025; 19:e12062. [PMID: 39712858 PMCID: PMC11656398 DOI: 10.1002/ccs3.12062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
|
8
|
Wang K, Mitoh Y, Horie K, Yoshida R. Exploring the Role of Ccn3 in Type III Cell of Mice Taste Buds. J Neurochem 2025; 169:e16291. [PMID: 39709613 DOI: 10.1111/jnc.16291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024]
Abstract
Different taste cells express unique cell-type markers, enabling researchers to distinguish them and study their functional differentiation. Using single-cell RNA-Seq of taste cells in mouse fungiform papillae, we found that Cellular Communication Network Factor 3 (Ccn3) was highly expressed in Type III taste cells but not in Type II taste cells. Ccn3 is a protein-coding gene involved in various biological processes, such as cell proliferation, angiogenesis, tumorigenesis, and wound healing. Therefore, in this study, we aimed to explore the expression and function of Ccn3 in mouse taste bud cells. Using reverse transcription polymerase chain reaction (RT-PCR), in situ hybridization, and immunohistochemistry (IHC), we confirmed that Ccn3 was predominantly expressed in Type III taste cells. Through IHC, quantitative real-time RT-PCR, gustatory nerve recordings, and short-term lick tests, we observed that Ccn3 knockout (Ccn3-KO) mice did not exhibit any significant differences in the expression of taste cell markers and taste responses compared to wild-type controls. To explore the function of Ccn3 in taste cells, bioinformatics analyses were conducted and predicted possible roles of Ccn3 in tissue regeneration, perception of pain, protein secretion, and immune response. Among them, an immune function is the most plausible based on our experimental results. In summary, our study indicates that although Ccn3 is strongly expressed in Type III taste cells, its knockout did not influence the basic taste response, but bioinformatics provided valuable insights into the possible role of Ccn3 in taste buds and shed light on future research directions.
Collapse
Affiliation(s)
- Kuanyu Wang
- Department of Oral Physiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yoshihiro Mitoh
- Department of Oral Physiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kengo Horie
- Department of Oral Physiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryusuke Yoshida
- Department of Oral Physiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
9
|
Abstract
The extracellular matrix (ECM) is the complex meshwork of proteins and glycans that forms the scaffold that surrounds and supports cells. It exerts key roles in all aspects of metazoan physiology, from conferring physical and mechanical properties on tissues and organs to modulating cellular processes such as proliferation, differentiation and migration. Understanding the mechanisms that orchestrate the assembly of the ECM scaffold is thus crucial to understand ECM functions in health and disease. This Review discusses novel insights into the compositional diversity of matrisome components and the mechanisms that lead to tissue-specific assemblies and architectures tailored to support specific functions. The Review then highlights recently discovered mechanisms, including post-translational modifications and metabolic pathways such as amino acid availability and the circadian clock, that modulate ECM secretion, assembly and remodelling in homeostasis and human diseases. Last, the Review explores the potential of 'matritherapies', that is, strategies to normalize ECM composition and architecture to achieve a therapeutic benefit.
Collapse
Affiliation(s)
- Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, USA.
- University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
10
|
Shorey-Kendrick LE, McEvoy CT, O'Sullivan SM, Milner K, Vuylsteke B, Tepper RS, Morgan TK, Roberts VHJ, Lo JO, Frias AE, Haas DM, Park B, Gao L, Vu A, Morris CD, Spindel ER. Vitamin C supplementation improves placental function and alters placental gene expression in smokers. Sci Rep 2024; 14:25486. [PMID: 39461975 PMCID: PMC11513119 DOI: 10.1038/s41598-024-73005-7] [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: 01/11/2024] [Accepted: 09/12/2024] [Indexed: 10/28/2024] Open
Abstract
Maternal smoking during pregnancy (MSDP), driven by nicotine crossing the placenta, causes lifelong decreases in offspring pulmonary function and vitamin C supplementation during pregnancy prevents some of those changes. We have also shown in animal models of prenatal nicotine exposure that vitamin C supplementation during pregnancy improves placental function. In this study we examined whether vitamin C supplementation mitigates the effects of MSDP on placental structure, function, and gene expression in pregnant human smokers. Doppler ultrasound was performed in a subset of 55 pregnant smokers participating in the "Vitamin C to Decrease the Effects of Smoking in Pregnancy on Infant Lung Function" (VCSIP) randomized clinical trial (NCT01723696) and in 33 pregnant nonsmokers. Doppler ultrasound measurements showed decreased umbilical vein Doppler velocity (Vmax) in placebo-treated smokers that was significantly improved in smokers randomized to vitamin C, restoring to levels comparable to nonsmokers. RNA-sequencing demonstrated that vitamin C supplementation to pregnant smokers was associated with changes in mRNA expression in genes highly relevant to vascular and cardiac development, suggesting a potential mechanism for vitamin C supplementation in pregnant smokers to improve some aspects of offspring health.
Collapse
Affiliation(s)
- Lyndsey E Shorey-Kendrick
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave, Beaverton, OR, 97006, USA
| | - Cindy T McEvoy
- Department of Pediatrics, Papé Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA
| | - Shannon M O'Sullivan
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave, Beaverton, OR, 97006, USA
| | - Kristin Milner
- Department of Pediatrics, Papé Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA
| | - Brittany Vuylsteke
- Department of Pediatrics, Papé Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA
| | - Robert S Tepper
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Terry K Morgan
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Beaverton, OR, USA
| | - Victoria H J Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Jamie O Lo
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Beaverton, OR, USA
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Antonio E Frias
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Beaverton, OR, USA
| | - David M Haas
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Byung Park
- Oregon Health & Science University-Portland State University School of Public Health, Knight Cancer Institute, Portland, USA
| | - Lina Gao
- Oregon Health & Science University-Portland State University School of Public Health, Knight Cancer Institute, Portland, USA
| | - Annette Vu
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Cynthia D Morris
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
- Oregon Clinical & Translational Research Institute, Oregon Health & Science University, Portland, OR, USA
| | - Eliot R Spindel
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave, Beaverton, OR, 97006, USA.
| |
Collapse
|
11
|
Christopoulou ME, Aletras AJ, Papakonstantinou E, Stolz D, Skandalis SS. WISP1 and Macrophage Migration Inhibitory Factor in Respiratory Inflammation: Novel Insights and Therapeutic Potentials for Asthma and COPD. Int J Mol Sci 2024; 25:10049. [PMID: 39337534 PMCID: PMC11432718 DOI: 10.3390/ijms251810049] [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: 07/08/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Recent advancements highlight the intricate interplay between the extracellular matrix (ECM) and immune responses, notably in respiratory diseases such as asthma and Chronic Obstructive Pulmonary Disease (COPD). The ECM, a dynamic structural framework within tissues, orches-trates a plethora of cellular processes, including immune cell behavior and tissue repair mecha-nisms. WNT1-inducible-signaling pathway protein 1 (WISP1), a key ECM regulator, controls immune cell behavior, cytokine production, and tissue repair by modulating integrins, PI3K, Akt, β-catenin, and mTOR signaling pathways. WISP1 also induces macrophage migration inhibitory factor (MIF) expression via Src kinases and epidermal growth factor receptor (EGFR) activation. MIF, through its wide range of activities, enhances inflammation and tissue restructuring. Rec-ognized for its versatile roles in regulating the immune system, MIF interacts with multiple immune components, such as the NLRP3 inflammasome, thereby sustaining inflammatory pro-cesses. The WISP1-MIF axis potentially unveils complex molecular mechanisms governing im-mune responses and inflammation. Understanding the intricate roles of WISP1 and MIF in the pathogenesis of chronic respiratory diseases such as asthma and COPD could lead to the identi-fication of novel targets for therapeutic intervention to alleviate disease severity and enhance patient outcomes.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexios J Aletras
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Eleni Papakonstantinou
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daiana Stolz
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Spyros S Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
12
|
Gu J, Tong W, Wang X, Gu L, Wang W, Zang T, Lou M, Liu Y. Multi-omics Analysis Revealed that the CCN Family Regulates Cell Crosstalk, Extracellular Matrix, and Immune Escape, Leading to a Poor Prognosis of Glioma. Cell Biochem Biophys 2024; 82:2157-2170. [PMID: 38837011 DOI: 10.1007/s12013-024-01323-8] [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] [Accepted: 05/17/2024] [Indexed: 06/06/2024]
Abstract
The CCN family is a group of matricellular proteins associated with the extracellular matrix. This study aims to explore the role of the CCN family in glioma development and its implications in the tumor microenvironment. Through analysis of bulk RNA-seq cohorts, correlations between CCN family expression and glioma subtypes, patient survival, and bioactive pathway enrichment were investigated. Additionally, single-cell datasets were employed to identify novel cell subgroups, followed by analyses of cell communication and transcription factors. Spatial transcriptomic analysis was utilized to validate the CCN family's involvement in glioma. Results indicate overexpression of CYR61,CTGF, and WISP1 in glioma, associated with unfavorable subtypes and reduced survival. Enrichment analyses revealed associations with oncogenic pathways, while CTGF and WISP1 expression correlated with increased infiltration of regulatory T cells and M2 macrophages. Single-cell analysis identified MES-like cells as the highest CCN expression. Moreover, intercellular signal transduction analysis demonstrated active pathways, including SPP1-CD44, in cell subgroups with elevated CYR61 and CTGF expression. Spatial transcriptomic analysis confirmed co-localization of CYR61,CTGF and SPP1-CD44 with high oncogenic pathway activity. These findings suggest that CCN family members may serve as potential prognostic biomarkers and therapeutic targets for glioma.
Collapse
Affiliation(s)
- Jingyan Gu
- Department of Neurosurgery, Shanghai General Hospital affiliated to Nanjing Medical University, Shanghai, China
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjie Tong
- Department of Neurosurgery, Shanghai General Hospital affiliated to Nanjing Medical University, Shanghai, China
- Department of Neurosurgery, Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xu Wang
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lianping Gu
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Zang
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meiqing Lou
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yaohua Liu
- Department of Neurosurgery, Shanghai General Hospital affiliated to Nanjing Medical University, Shanghai, China.
- Department of Neurosurgery, Shanghai General Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Wang H, Twumasi G, Xu Q, Xi Y, Qi J, Yang Z, Shen Z, Bai L, Li L, Liu H. Identification of candidate genes associated with primary feathers of tianfu nonghua ducks based on Genome-wide association studies. Poult Sci 2024; 103:103985. [PMID: 38968866 PMCID: PMC11269910 DOI: 10.1016/j.psj.2024.103985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024] Open
Abstract
The primary feathers of ducks have important economic value in the poultry industry. This study quantified the primary feather phenotype of Nonghua ducks, including the primary feathers' length, area, distribution of black spots, and feather symmetry. And genome-wide association analysis was used to screen candidate genes that affect the primary feather traits. The genome-wide association study (GWAS) results identified the genetic region related to feather length (FL) on chromosome 2. Through Linkage disequilibrium (LD) analysis, candidate regions (chr2: 115,246,393-116,501,448 bp) were identified and were further annotated to 5 genes: MRS2, GPLD1, ALDH5A1, KIAA0319, and ATP9B. Secondly, candidate regions related to feather black spots were identified on chromosome 21. Through LD analysis, the candidate regions (chr21: 163,552-2,183,853 bp) were screened and further annotated to 47 genes. Among them, STK4, CCN5, and YWHAB genes were related to melanin-related pathways or pigment deposition, which may be key genes affecting the distribution of black spots on feathers. In addition, we also screened 125 genes on multiple chromosomes that may be related to feather symmetry. Among them, significant SNPs on chromosome 1 were further identified as candidate regions (chr1: 142,118,209-142,223,605 bp) through LD analysis and annotated into 2 genes, TGFBRAP1 and LOC113839965. These results reported the genetic basis of the primary feather from multiple phenotypes, and offered valuable insights into the genetic basis for the growth and development of duck feathers and feather color pattern.
Collapse
Affiliation(s)
- Huazhen Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Grace Twumasi
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Qian Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yang Xi
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Jingjing Qi
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Zhao Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Zhengyang Shen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Lili Bai
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Liang Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Hehe Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, PR China.
| |
Collapse
|
14
|
Bhattacharya R, Ward T, Kalejaiye TD, Mishra A, Leeman S, Arzaghi H, Seidman JG, Seidman CE, Musah S. Engineered human iPS cell models reveal altered podocytogenesis and glomerular capillary wall in CHD-associated SMAD2 mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606108. [PMID: 39211233 PMCID: PMC11360959 DOI: 10.1101/2024.08.02.606108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Early developmental programming involves extensive cell lineage diversification through shared molecular signaling networks. Clinical observations of congenital heart disease (CHD) patients carrying SMAD2 genetic variants revealed correlations with multi-organ impairments at the developmental and functional levels. For example, many CHD patients present with glomerulosclerosis, periglomerular fibrosis, and albuminuria. Still, it remains largely unknown whether SMAD2 variants associated with CHD can directly alter kidney cell fate, tissue patterning, and organ-level function. To address this question, we engineered human iPS cells (iPSCs) and organ-on-a-chip systems to uncover the role of pathogenic SMAD2 variants in kidney podocytogenesis. Our results show that abrogation of SMAD2 causes altered patterning of the mesoderm and intermediate mesoderm (IM) cell lineages, which give rise to nearly all kidney cell types. Upon further differentiation of IM cells, the mutant podocytes failed to develop arborizations and interdigitations. A reconstituted glomerulus-on-a-chip platform exhibited significant proteinuria as clinically observed in glomerulopathies. This study implicates CHD-associated SMAD2 mutations in kidney tissue malformation and provides opportunities for therapeutic discovery in the future.
Collapse
|
15
|
Chu CQ, Quan T. Fibroblast Yap/Taz Signaling in Extracellular Matrix Homeostasis and Tissue Fibrosis. J Clin Med 2024; 13:3358. [PMID: 38929890 PMCID: PMC11204269 DOI: 10.3390/jcm13123358] [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: 04/17/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Tissue fibrosis represents a complex pathological condition characterized by the excessive accumulation of collagenous extracellular matrix (ECM) components, resulting in impaired organ function. Fibroblasts are central to the fibrotic process and crucially involved in producing and depositing collagen-rich ECM. Apart from their primary function in ECM synthesis, fibroblasts engage in diverse activities such as inflammation and shaping the tissue microenvironment, which significantly influence cellular and tissue functions. This review explores the role of Yes-associated protein (Yap) and Transcriptional co-activator with PDZ-binding motif (Taz) in fibroblast signaling and their impact on tissue fibrosis. Gaining a comprehensive understanding of the intricate molecular mechanisms of Yap/Taz signaling in fibroblasts may reveal novel therapeutic targets for fibrotic diseases.
Collapse
Affiliation(s)
- Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR 97239, USA;
- Rheumatology Section, VA Portland Health Care System, Portland, OR 97239, USA
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Hu K, Xie L, Wu W, Zhang J, Li Y, He J, Zhang Y, Lu D, Koeffler HP, Lin L, Yin D. CYR61 Acts as an Intracellular Microtubule-Associated Protein and Coordinates Mitotic Progression via PLK1-FBW7 Pathway. Int J Biol Sci 2024; 20:3140-3155. [PMID: 38904029 PMCID: PMC11186368 DOI: 10.7150/ijbs.93335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Cysteine-rich angiogenic inducer 61 (CYR61), also called CCN1, has long been characterized as a secretory protein. Nevertheless, the intracellular function of CYR61 remains unclear. Here, we found that CYR61 is important for proper cell cycle progression. Specifically, CYR61 interacts with microtubules and promotes microtubule polymerization to ensure mitotic entry. Moreover, CYR61 interacts with PLK1 and accumulates during the mitotic process, followed by degradation as mitosis concludes. The proteolysis of CYR61 requires the PLK1 kinase activity, which directly phosphorylates two conserved motifs on CYR61, enhancing its interaction with the SCF E3 complex subunit FBW7 and mediating its degradation by the proteasome. Mutations of phosphorylation sites of Ser167 and Ser188 greatly increase CYR61's stability, while deletion of CYR61 extends prophase and metaphase and delays anaphase onset. In summary, our findings highlight the precise control of the intracellular CYR61 by the PLK1-FBW7 pathway, accentuating its significance as a microtubule-associated protein during mitotic progression.
Collapse
Affiliation(s)
- Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Limin Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Wenjing Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
- Department of Breast Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Jingyuan Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Yu Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen 518000, P.R. China
| | - Jiehua He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Yin Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Daning Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - H. Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lehang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| |
Collapse
|
17
|
Kim JY, Quan T. Emerging Perspectives of YAP/TAZ in Human Skin Epidermal and Dermal Aging. Ann Dermatol 2024; 36:135-144. [PMID: 38816974 PMCID: PMC11148314 DOI: 10.5021/ad.23.156] [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: 12/11/2023] [Revised: 01/25/2024] [Accepted: 02/18/2024] [Indexed: 06/01/2024] Open
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are key downstream effectors of the Hippo signaling pathway, which plays a central role in tissue homeostasis, organ development, and regeneration. While the dysregulation of YAP/TAZ has been linked to various human diseases, their involvement in the aging of human skin has only recently begun to manifest. In the skin, the YAP/TAZ effectors emerge as central regulators in maintaining homeostasis of epidermal stem cells and dermal extracellular matrix, and thus intimately linked to skin aging processes. This review underscores recent molecular breakthroughs highlighting how age-related decline of YAP/TAZ activity impacts human epidermal and dermal aging. Gaining insight into the evolving roles of YAP/TAZ in human skin aging presents a promising avenue for the development of innovative therapeutic approaches aimed at enhancing skin health and addressing age-related skin conditions.
Collapse
Affiliation(s)
- Jun Young Kim
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Liu YC, Chen SH, Kuan CH, Chen SH, Huang WY, Chen HX, Wang TW. Assembly of Interfacial Polyelectrolyte Complexation Fibers with Mineralization Gradient for Physiologically-Inspired Ligament Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314294. [PMID: 38572797 DOI: 10.1002/adma.202314294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/29/2024] [Indexed: 04/05/2024]
Abstract
Current synthetic grafts for ligament rupture repair often fail to integrate well with the surrounding biological tissue, leading to complications such as graft wear, fatigue, and subsequent re-rupture. To address this medical challenge, this study aims at advancing the development of a biological ligament through the integration of physiologically-inspired principles and tissue engineering strategies. In this study, interfacial polyelectrolyte complexation (IPC) spinning technique, along with a custom-designed collection system, to fabricate a hierarchical scaffold mimicking native ligament structure, is utilized. To emulate the bone-ligament interface and alleviate stress concentration, a hydroxyapatite (HAp) mineral gradient is strategically introduced near both ends of the scaffold to enhance interface integration and diminish the risk of avulsion rupture. Biomimetic viscoelasticity is successfully displayed to provide similar mechanical support to native ligamentous tissue under physiological conditions. By introducing the connective tissue growth factor (CTGF) and conducting mesenchymal stem cells transplantation, the regenerative potential of the synthetic ligament is significantly amplified. This pioneering study offers a multifaceted solution combining biomimetic materials, regenerative therapies, and advanced techniques to potentially transform ligament rupture treatment.
Collapse
Affiliation(s)
- Yu-Chung Liu
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30044, Taiwan
| | - Shih-Heng Chen
- Division of Trauma Plastic Surgery, Department of Plastic & Reconstructive Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan City, 33305, Taiwan
| | - Chen-Hsiang Kuan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, 100229, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Shih-Hsien Chen
- Division of Trauma Plastic Surgery, Department of Plastic & Reconstructive Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan City, 33305, Taiwan
| | - Wei-Yuan Huang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30044, Taiwan
| | - Hao-Xuan Chen
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30044, Taiwan
| | - Tzu-Wei Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30044, Taiwan
| |
Collapse
|
19
|
Nilay M, Rawool A, Mandal K. Progressive Pseudorheumatoid Dysplasia of Childhood (PPRD)-A Case Series with Recurrent c.740_741del Variant. J Pediatr Genet 2024; 13:62-68. [PMID: 38567170 PMCID: PMC10984709 DOI: 10.1055/s-0041-1736611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
Progressive pseudorheumatoid dysplasia (PPRD) is an autosomal recessive arthropathy, affecting school-aged children. It is characterized by progressive degeneration of the articular cartilage. The majority of the pathogenic variations are found in exon 2, exon 4, and exon 5 of the putative gene, CCN6 (WISP3). Three unrelated individuals with clinical diagnosis of PPD were included in this study. Detailed clinicoradiological evaluation was attempted with brief literature review. Exome sequencing was performed in all three cases. All the pathogenic variations detected in our cohort were located in exons 2 and 4 of WISP3 gene. Though the clinicoradiological features are already well described, this study in north India highlights the occurrence of a recurring pathogenic variant. The c.740_741del variant was a recurrent pathogenic variant seen in all three patients in this cohort. This may be a common pathogenic variant in the North Indian population; however, a larger cohort needs to be studied before drawing final conclusions. A proper molecular diagnosis is a must to end the diagnostic odyssey, safeguarding patients with PPRD from unnecessary use of drugs like corticosteroids.
Collapse
Affiliation(s)
- Mayank Nilay
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Anup Rawool
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
- Department of Clinical Genetics, Sahyadri Hospitals Limited, Pune, Maharashtra, India
| | - Kausik Mandal
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
20
|
Christopoulou ME, Skandalis SS, Papakonstantinou E, Stolz D, Aletras AJ. WISP1 induces the expression of macrophage migration inhibitory factor in human lung fibroblasts through Src kinases and EGFR-activated signaling pathways. Am J Physiol Cell Physiol 2024; 326:C850-C865. [PMID: 38145300 PMCID: PMC11193488 DOI: 10.1152/ajpcell.00410.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Wnt1-inducible signaling protein 1 (WISP1/CCN4) is a secreted matricellular protein that is implicated in lung and airway remodeling. The macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that has been associated with chronic lung diseases. In this study, we aimed to investigate the WISP1 signaling pathway and its ability to induce the expression of MIF in primary cultures of fibroblasts from normal human lungs (HLFs). Our results showed that WISP1 significantly stimulated the expression of MIF in a concentration- and time-dependent fashion. In WISP1-induced expression of MIF, αvβ5-integrin and chondroitin sulfate proteoglycans as well as Src tyrosine kinases, MAP kinases, phosphatidylinositol 3-kinase/Akt, PKC, and NF-κB were involved. WISP1-induced expression of MIF was attenuated in the presence of the Src kinase inhibitor PP2 or the MIF tautomerase activity inhibitor ISO-1. Moreover, WISP1 significantly increased the phosphorylation and activation of EGF receptor (EGFR) through transactivation by Src kinases. WISP1 also induced the expression of MIF receptor CD74 and coreceptor CD44, through which MIF exerts its effects on HLFs. In addition, it was found that MIF induced its own expression, as well as its receptors CD74/CD44, acting in an autocrine manner. Finally, WISP1-induced MIF promoted the expression of cyclooxygenase 2, prostaglandin E2, IL-6, and matrix metalloproteinase-2 demonstrating the regulatory role of WISP1-MIF axis in lung inflammation and remodeling involving mainly integrin αvβ5, Src kinases, PKC, NF-κB, and EGFR. The specific signaling pathways involved in WISP1-induced expression of MIF may prove to be excellent candidates for novel targets to control inflammation in chronic lung diseases.NEW & NOTEWORTHY The present study demonstrates for the first time that Wnt1-inducible signaling protein 1 (WISP1) regulates migration inhibitory factor (MIF) expression and activity and identifies the main signaling pathways involved. The newly discovered WISP1-MIF axis may drive lung inflammation and could result in the design of novel targeted therapies in inflammatory lung diseases.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Spyros S Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Eleni Papakonstantinou
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daiana Stolz
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexios J Aletras
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
21
|
Shi L, Zou H, Yi J. Construction of shared gene signature between rheumatoid arthritis and lung adenocarcinoma helps to predict the prognosis and tumor microenvironment of the LUAD patients. Front Mol Biosci 2024; 10:1314753. [PMID: 38268722 PMCID: PMC10806137 DOI: 10.3389/fmolb.2023.1314753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/26/2023] [Indexed: 01/26/2024] Open
Abstract
Introduction: Rheumatoid arthritis (RA) is a common chronic autoimmune disease with high incidence rate and high disability rate. One of the top complications is cancer, especially lung adenocarcinoma (LUAD). However, the molecular mechanisms linking RA and LUAD are still not clear. Therefore, in this study, we tried to identify the shared genetic signatures and local immune microenvironment between RA and LUAD and construct a clinical model for survival prediction. Methods: We obtained gene expression profiles and clinical information of patients with RA and LUAD from GEO and TCGA datasets. We performed differential analysis and Weighted Gene Co-expression Network Analysis (WGCNA) to discover the shared genes between RA and LUAD. Then, COX regression and LASSO analysis were employed to figure out genes significantly associated with survival. qRT-PCR and Western blot were utilized to validate the expression level of candidate genes. For clinical application, we constructed a nomogram, and also explored the value of RALUADS in characterizing immune infiltration features by CIBERSORT and xCell. Finally, responses to different drug therapy were predicted according to different RALUADS. Results: Our analysis identified two gene sets from differentially expressed genes and WGCNA gene modules of RA and LUAD. Filtered by survival analysis, three most significant shared genes were selected, CCN6, CDCA4 and ERLIN1, which were all upregulated in tumors and associated with poor prognosis. The three genes constituted RA and LUAD score (RALUADS). Our results demonstrated that RALUADS was higher in tumor patients and predicted poor prognosis in LUAD patients. Clinical nomogram combining RALUADS and other clinicopathological parameters had superior performance in survival prediction (AUC = 0.722). We further explored tumor immune microenvironment (TME) affected by RALUADS and observed RALUADS was closely related to the sensitivity of multiple immune blockades, chemotherapy and targeted drugs. Conclusion: Our findings suggest that there are shared physiopathologic processes and molecular profiles between RA and LUAD. RALUADS represents an excellent prognosis predictor and immune-related biomarker, which can be applied to select potential effective drugs and for LUAD patients with RA.
Collapse
Affiliation(s)
- Liping Shi
- Department of Pharmacology, Gannan Healthcare Vocational College, Ganzhou, China
| | - Houwen Zou
- Department of Pharmacology, Dermatology Hospital of Ganzhou, Ganzhou, China
| | - Jian Yi
- Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
22
|
Mutsaers SE, Miles T, Prêle CM, Hoyne GF. Emerging role of immune cells as drivers of pulmonary fibrosis. Pharmacol Ther 2023; 252:108562. [PMID: 37952904 DOI: 10.1016/j.pharmthera.2023.108562] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
The pathogenesis of pulmonary fibrosis, including idiopathic pulmonary fibrosis (IPF) and other forms of interstitial lung disease, involves a complex interplay of various factors including host genetics, environmental pollutants, infection, aberrant repair and dysregulated immune responses. Highly variable clinical outcomes of some ILDs, in particular IPF, have made it difficult to identify the precise mechanisms involved in disease pathogenesis and thus the development of a specific cure or treatment to halt and reverse the decline in patient health. With the advent of in-depth molecular diagnostics, it is becoming evident that the pathogenesis of IPF is unlikely to be the same for all patients and therefore will likely require different treatment approaches. Chronic inflammation is a cardinal feature of IPF and is driven by both innate and adaptive immune responses. Inflammatory cells and activated fibroblasts secrete various pro-inflammatory cytokines and chemokines that perpetuate the inflammatory response and contribute to the recruitment and activation of more immune cells and fibroblasts. The balance between pro-inflammatory and regulatory immune cell subsets, as well as the interactions between immune cell types and resident cells within the lung microenvironment, ultimately determines the extent of fibrosis and the potential for resolution. This review examines the role of the innate and adaptive immune responses in pulmonary fibrosis, with an emphasis on IPF. The role of different immune cell types is discussed as well as novel anti-inflammatory and immunotherapy approaches currently in clinical trial or in preclinical development.
Collapse
Affiliation(s)
- Steven E Mutsaers
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia.
| | - Tylah Miles
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia
| | - Cecilia M Prêle
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia; School of Medical, Molecular and Forensic Sciences, Murdoch University, WA, Australia
| | - Gerard F Hoyne
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia; The School of Health Sciences and Physiotherapy, University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
23
|
Li C, Alemany-Ribes M, Raftery RM, Nwoko U, Warman ML, Craft AM. Directed differentiation of human pluripotent stem cells into articular cartilage reveals effects caused by absence of WISP3, the gene responsible for progressive pseudorheumatoid arthropathy of childhood. Ann Rheum Dis 2023; 82:1547-1557. [PMID: 37679035 DOI: 10.1136/ard-2023-224304] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023]
Abstract
OBJECTIVES Progressive pseudorheumatoid arthropathy of childhood (PPAC), caused by deficiency of WNT1 inducible signalling pathway protein 3 (WISP3), has been challenging to study because no animal model of the disease exists and cartilage recovered from affected patients is indistinguishable from common end-stage osteoarthritis. Therefore, to gain insights into why precocious articular cartilage failure occurs in this disease, we made in vitro derived articular cartilage using isogenic WISP3-deficient and WISP3-sufficient human pluripotent stem cells (hPSCs). METHODS We generated articular cartilage-like tissues from induced-(i) PSCs from two patients with PPAC and one wild-type human embryonic stem cell line in which we knocked out WISP3. We compared these tissues to in vitro-derived articular cartilage tissues from two isogenic WISP3-sufficient control lines using histology, bulk RNA sequencing, single cell RNA sequencing and in situ hybridisation. RESULTS WISP3-deficient and WISP3-sufficient hPSCs both differentiated into articular cartilage-like tissues that appeared histologically similar. However, the transcriptomes of WISP3-deficient tissues differed significantly from WISP3-sufficient tissues and pointed to increased TGFβ, TNFα/NFκB, and IL-2/STAT5 signalling and decreased oxidative phosphorylation. Single cell sequencing and in situ hybridisation revealed that WISP3-deficient cartilage contained a significantly higher fraction (~4 fold increase, p<0.001) of superficial zone chondrocytes compared with deeper zone chondrocytes than did WISP3-sufficient cartilage. CONCLUSIONS WISP3-deficient and WISP3-sufficient hPSCs can be differentiated into articular cartilage-like tissues, but these tissues differ in their transcriptomes and in the relative abundances of chondrocyte subtypes they contain. These findings provide important starting points for in vivo studies when an animal model of PPAC or presymptomatic patient-derived articular cartilage becomes available.
Collapse
Affiliation(s)
- Chaochang Li
- Department of Orthopedic Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Mireia Alemany-Ribes
- Department of Orthopedic Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Rosanne M Raftery
- Department of Orthopedic Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Uzochi Nwoko
- Department of Orthopedic Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Matthew L Warman
- Department of Orthopedic Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - April M Craft
- Department of Orthopedic Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Department of Orthopedic Surgery, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Liu J, Lv S, Ma W, Yang D, Zhang X. Effect of WISP1 on paraquat-induced EMT. Toxicol In Vitro 2023; 93:105693. [PMID: 37689312 DOI: 10.1016/j.tiv.2023.105693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND Paraquat (PQ) can induce pulmonary fibrosis (PF) by modulating epithelial-mesenchymal transition (EMT) of alveolar epithelial cells, but the molecular mechanism is unknown. In this paper, the role of Wnt-inducible signaling protein-1 (WISP1) in PQ-induced EMT was inspected. METHODS The morphology, apoptosis, and mortality of A549 cells were observed through a microscope. The mRNA and protein levels of WISP1, E-cadherin, and Vimentin were confirmed by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot. RESULTS With the increase of PQ concentration, the morphology of A549 cells was apparently changed, cell apoptosis and mortality were enhanced. Besides, the E-cadherin abundance was reduced (p < 0.01), however, WISP1 and Vimentin contents were boosted after PQ treatment (p < 0.01). With the increase of PQ treatment time, the epithelial index of cells first increased and then decreased. The expression of WISP1 gene increased significantly with the increase of PQ treatment time (p < 0.01). Silence of WISP1 abolished the effect of PQ treatment on E-cadherin and Vimentin levels (p < 0.01). Downregulation of WISP1 curbed morphology change and PQ-induced EMT in A549 cells. CONCLUSION Knockdown of WISP1 inhibited PQ-induced EMT in A549 cells. This conclusion might provide a new therapeutic target for PQ poisoning treatment.
Collapse
Affiliation(s)
- Jingyan Liu
- Department of Emergency, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Longgang District People's Hospital of Shenzhen, Shenzhen, Guangdong 518172, PR China
| | - Shengnan Lv
- Department of Out-patient, Linyi People's Hospital, Linyi 276000, Shandong, China
| | - Wanling Ma
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Longgang District People's Hospital of Shenzhen, Shenzhen, Guangdong 518172, PR China
| | - Dong Yang
- Department of Vascular Surgery, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Longgang District People's Hospital of Shenzhen, Shenzhen, Guangdong 518172, PR China
| | - Xuchang Zhang
- Department of Geriatrics Emergency, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Longgang District People's Hospital of Shenzhen, Shenzhen, Guangdong 518172, PR China.
| |
Collapse
|
25
|
Quan T. Human Skin Aging and the Anti-Aging Properties of Retinol. Biomolecules 2023; 13:1614. [PMID: 38002296 PMCID: PMC10669284 DOI: 10.3390/biom13111614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
The skin is the most-extensive and -abundant tissue in the human body. Like many organs, as we age, human skin experiences gradual atrophy in both the epidermis and dermis. This can be primarily attributed to the diminishing population of epidermal stem cells and the reduction in collagen, which is the primary structural protein in the human body. The alterations occurring in the epidermis and dermis due to the aging process result in disruptions to the structure and functionality of the skin. This creates a microenvironment conducive to age-related skin conditions such as a compromised skin barrier, slowed wound healing, and the onset of skin cancer. This review emphasizes the recent molecular discoveries related to skin aging and evaluates preventive approaches, such as the use of topical retinoids. Topical retinoids have demonstrated promise in enhancing skin texture, diminishing fine lines, and augmenting the thickness of both the epidermal and dermal layers.
Collapse
Affiliation(s)
- Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
26
|
Yu L, Wen H, Liu C, Wang C, Yu H, Zhang K, Han Q, Liu Y, Han Z, Li Z, Liu N. Embryonic stem cell-derived extracellular vesicles rejuvenate senescent cells and antagonize aging in mice. Bioact Mater 2023; 29:85-97. [PMID: 37449253 PMCID: PMC10336196 DOI: 10.1016/j.bioactmat.2023.06.011] [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: 12/13/2022] [Revised: 05/11/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Aging is a degenerative process that leads to tissue dysfunction and death. Embryonic stem cells (ESCs) have great therapeutic potential for age-related diseases due to their capacity for self-renewal and plasticity. However, the use of ESCs in clinical treatment is limited by immune rejection, tumourigenicity and ethical issues. ESC-derived extracellular vesicles (EVs) may provide therapeutic effects that are comparable to those of ESCs while avoiding unwanted effects. Here, we fully evaluate the role of ESC-EVs in rejuvenation in vitro and in vivo. Using RNA sequencing (RNA-Seq) and microRNA sequencing (miRNA-Seq) screening, we found that miR-15b-5p and miR-290a-5p were highly enriched in ESC-EVs, and induced rejuvenation by silencing the Ccn2-mediated AKT/mTOR pathway. These results demonstrate that miR-15b-5p and miR-290a-5p function as potent activators of rejuvenation mediated by ESC-EVs. The rejuvenating effect of ESC-EVs was further investigated in vivo by injection into aged mice. The results showed that ESC-EVs successfully ameliorated the pathological age-related phenotypes and rescued the transcriptome profile of aged mice. Our findings demonstrate that ESC-EVs treatment can rejuvenate senescence both in vitro and in vivo and suggest the therapeutic potential of ESC-EVs as a novel cell-free alternative to ESCs for age-related diseases.
Collapse
Affiliation(s)
- Lu Yu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Hang Wen
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chang Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chen Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Huaxin Yu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Kaiyue Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Qingsheng Han
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yue Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhongchao Han
- Institute of Stem Cells, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Na Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences Nankai University, Tianjin, 300071, China
| |
Collapse
|
27
|
Schumacher N, Thomsen I, Brundert F, Hejret V, Düsterhöft S, Tichý B, Schmidt-Arras D, Voss M, Rose-John S. EGFR stimulation enables IL-6 trans-signalling via iRhom2-dependent ADAM17 activation in mammary epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119489. [PMID: 37271223 DOI: 10.1016/j.bbamcr.2023.119489] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/14/2023] [Accepted: 05/05/2023] [Indexed: 06/06/2023]
Abstract
The cytokine interleukin-6 (IL-6) has considerable pro-inflammatory properties and is a driver of many physiological and pathophysiological processes. Cellular responses to IL-6 are mediated by membrane-bound or soluble forms of the IL-6 receptor (IL-6R) complexed with the signal-transducing subunit gp130. While expression of the membrane-bound IL-6R is restricted to selected cell types, soluble IL-6R (sIL-6R) enables gp130 engagement on all cells, a process termed IL-6 trans-signalling and considered to be pro-inflammatory. sIL-6R is predominantly generated through proteolytic processing by the metalloproteinase ADAM17. ADAM17 also liberates ligands of the epidermal growth factor receptor (EGFR), which is a prerequisite for EGFR activation and results in stimulation of proliferative signals. Hyperactivation of EGFR mostly due to activating mutations drives cancer development. Here, we reveal an important link between overshooting EGFR signalling and the IL-6 trans-signalling pathway. In epithelial cells, EGFR activity induces not only IL-6 expression but also the proteolytic release of sIL-6R from the cell membrane by increasing ADAM17 surface activity. We find that this derives from the transcriptional upregulation of iRhom2, a crucial regulator of ADAM17 trafficking and activation, upon EGFR engagement, which results in increased surface localization of ADAM17. Also, phosphorylation of the EGFR-downstream mediator ERK mediates ADAM17 activity via interaction with iRhom2. In sum, our study reveals an unforeseen interplay between EGFR activation and IL-6 trans-signalling, which has been shown to be fundamental in inflammation and cancer.
Collapse
Affiliation(s)
- Neele Schumacher
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany.
| | - Ilka Thomsen
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| | - Florian Brundert
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| | - Vaclav Hejret
- CEITEC-Central European Institute of Technology, Masaryk University, Czech Republic
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, University Hospital Aachen/RWTH, Aachen, Germany
| | - Boris Tichý
- CEITEC-Central European Institute of Technology, Masaryk University, Czech Republic
| | | | - Matthias Voss
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| |
Collapse
|
28
|
Baxter RC. Signaling Pathways of the Insulin-like Growth Factor Binding Proteins. Endocr Rev 2023; 44:753-778. [PMID: 36974712 PMCID: PMC10502586 DOI: 10.1210/endrev/bnad008] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/25/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
The 6 high-affinity insulin-like growth factor binding proteins (IGFBPs) are multifunctional proteins that modulate cell signaling through multiple pathways. Their canonical function at the cellular level is to impede access of insulin-like growth factor (IGF)-1 and IGF-2 to their principal receptor IGF1R, but IGFBPs can also inhibit, or sometimes enhance, IGF1R signaling either through their own post-translational modifications, such as phosphorylation or limited proteolysis, or by their interactions with other regulatory proteins. Beyond the regulation of IGF1R activity, IGFBPs have been shown to modulate cell survival, migration, metabolism, and other functions through mechanisms that do not appear to involve the IGF-IGF1R system. This is achieved by interacting directly or functionally with integrins, transforming growth factor β family receptors, and other cell-surface proteins as well as intracellular ligands that are intermediates in a wide range of pathways. Within the nucleus, IGFBPs can regulate the diverse range of functions of class II nuclear hormone receptors and have roles in both cell senescence and DNA damage repair by the nonhomologous end-joining pathway, thus potentially modifying the efficacy of certain cancer therapeutics. They also modulate some immune functions and may have a role in autoimmune conditions such as rheumatoid arthritis. IGFBPs have been proposed as attractive therapeutic targets, but their ubiquity in the circulation and at the cellular level raises many challenges. By understanding the diversity of regulatory pathways with which IGFBPs interact, there may still be therapeutic opportunities based on modulation of IGFBP-dependent signaling.
Collapse
Affiliation(s)
- Robert C Baxter
- Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital,St Leonards, NSW 2065, Australia
| |
Collapse
|
29
|
El Dessouki D, Amr K, Kholoussi N, Rady HM, Temtamy SA, Abdou MMS, Aglan M. Clinical and molecular characterization in a cohort of patients with progressive pseudorheumatoid dysplasia. Am J Med Genet A 2023; 191:2329-2336. [PMID: 37377052 DOI: 10.1002/ajmg.a.63339] [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: 03/21/2023] [Revised: 06/05/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023]
Abstract
Progressive pseudorheumatoid dysplasia (PPRD), a rare autosomal recessive syndrome, is a type of skeletal dysplasia associated with pain, stiffness, swelling of multiple joints, and the absence of destructive changes. PPRD occurs due to loss of function pathogenic variants in WISP3 (CCN6) gene, located on chromosome 6q22. In this study, 23 unrelated Egyptian PPRD patients were clinically diagnosed based on medical history, physical and radiological examinations, and laboratory investigations. Sequencing of the whole WISP3 (CCN6) exons and introns boundaries was carried out for all patients. A total of 11 different sequence variations were identified in the WISP3 (CCN6) gene, five of them were new pathogenic variants: the NM_003880.3: c.80T>A (p.L27*), c.161delG (p.C54fs*12), c.737T>C (p.Leu246Pro), c.347-1G>A (IVS3-1G>A), and c.376C>T (p.Q126*). The results of this study expand the spectrum of WISP3 (CCN6) pathogenic variants associated with PPRD. Clinical and genetic analysis is important for proper genetic counseling to curb this rare disorder in the families.
Collapse
Affiliation(s)
- Dina El Dessouki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Khalda Amr
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Naglaa Kholoussi
- Immunogenetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Hanaa M Rady
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Samia Ali Temtamy
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Manal M S Abdou
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona Aglan
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
30
|
Hassan MDS, Razali N, Abu Bakar AS, Abu Hanipah NF, Agarwal R. Connective tissue growth factor: Role in trabecular meshwork remodeling and intraocular pressure lowering. Exp Biol Med (Maywood) 2023; 248:1425-1436. [PMID: 37873757 PMCID: PMC10657592 DOI: 10.1177/15353702231199466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023] Open
Abstract
Connective tissue growth factor (CTGF) is a distinct signaling molecule modulating many physiological and pathophysiological processes. This protein is upregulated in numerous fibrotic diseases that involve extracellular matrix (ECM) remodeling. It mediates the downstream effects of transforming growth factor beta (TGF-β) and is regulated via TGF-β SMAD-dependent and SMAD-independent signaling routes. Targeting CTGF instead of its upstream regulator TGF-β avoids the consequences of interfering with the pleotropic effects of TGF-β. Both CTGF and its upstream mediator, TGF-β, have been linked with the pathophysiology of glaucomatous optic neuropathy due to their involvement in the regulation of ECM homeostasis. The excessive expression of these growth factors is associated with glaucoma pathogenesis via elevation of the intraocular pressure (IOP), the most important risk factor for glaucoma. The raised in the IOP is due to dysregulation of ECM turnover resulting in excessive ECM deposition at the site of aqueous humor outflow. It is therefore believed that CTGF could be a potential therapeutic target in glaucoma therapy. This review highlights the CTGF biology and structure, its regulation and signaling, its association with the pathophysiology of glaucoma, and its potential role as a therapeutic target in glaucoma management.
Collapse
Affiliation(s)
| | - Norhafiza Razali
- Institute of Medical Molecular Biotechnology (IMMB), Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
- Center for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
| | - Amy Suzana Abu Bakar
- Institute of Medical Molecular Biotechnology (IMMB), Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
- Center for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
| | - Noor Fahitah Abu Hanipah
- Institute of Medical Molecular Biotechnology (IMMB), Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University (IMU), 57000 Kuala Lumpur, Malaysia
| |
Collapse
|
31
|
Zhou F, Sun J, Ye L, Jiang T, Li W, Su C, Ren S, Wu F, Zhou C, Gao G. Fibronectin promotes tumor angiogenesis and progression of non-small-cell lung cancer by elevating WISP3 expression via FAK/MAPK/ HIF-1α axis and activating wnt signaling pathway. Exp Hematol Oncol 2023; 12:61. [PMID: 37468964 DOI: 10.1186/s40164-023-00419-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 06/02/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Fibronectin, an extracellular matrix protein, has been reported to be associated with heterogeneous cancer stemness, angiogenesis and progression in multiple cancer types. However, the roles and the underlying mechanism of fibronectin on the progression NSCLC need to be further elucidated. METHODS Public dataset such as Kaplan-Meier Plotter was used to determine the prognostic significance of genes. The correlation of different protein expression in clinical and xenograft tissues was tested by immunohistochemistry experiment. Both in vitro and in vivo experiments were performed to determine the role of fibronectin on the tumor growth, metastasis, and angiogenesis in NSCLC. The activation of key signaling pathway under fibronectin was examined by WB assay. RNA-seq was applicated to screening the target gene of fibronectin. Rescue experiment was performed to confirm the role of target gene in fibronectin-mediated function in NSCLC. Finally, luciferase and CHIP assays were used to elucidate the mechanism by which fibronectin regulated the target gene. RESULTS Our results revealed that fibronectin was up-regulated in cancer tissues compared with the normal ones in NSCLC patients. Dish- coated fibronectin enhanced the tumor growth, metastasis, and angiogenesis of NSCLC in vitro and in vivo by promoting EMT and maintaining stemness of NSCLC cells. As expected, fibronectin activated FAK and its downstream MAPK/ERK signaling pathway. WISP3 was screened as a potential target gene of fibronectin. Interestingly, WISP3 effectively activated Wnt signaling pathway, and knockdown of WISP3 effectively blocked the influence of fibronectin on the migration, invasion and vascular structure formation potential of NSCLC cells. Our data also manifested that fibronectin elevated the transcription of WISP3 gene by promoting the binding of HIF-1α to the promoter region of WISP3 in NSCLC cells. CONCLUSIONS Our findings sketched the outline of the route for fibronectin exert its role in NSCLC, in which fibronectin activated downstream FAK and MAPK/ERK signaling pathways, and mediated the accumulation of HIF-1α. Then, HIF-1α enabled the transcription of WISP3, and subsequently promoted the activation of Wnt signaling pathway, and finally enhanced the tumor growth, metastasis, and angiogenesis in NSCLC.
Collapse
Affiliation(s)
- Fei Zhou
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Jianguo Sun
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, P R China
| | - Lingyun Ye
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Tao Jiang
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wei Li
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Chunxia Su
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shengxiang Ren
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Fengying Wu
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| | - Caicun Zhou
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| | - Guanghui Gao
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
32
|
Ascencio G, de Cruz MA, Abuel J, Alvarado S, Arriaga Y, Conrad E, Castro A, Eichelberger K, Galvan L, Gundy G, Garcia JAI, Jimenez A, Lu NT, Lugar C, Marania R, Mendsaikhan T, Ortega J, Nand N, Rodrigues NS, Shabazz K, Tam C, Valenciano E, Hayzelden C, Eritano AS, Riggs B. A deficiency screen of the 3rd chromosome for dominant modifiers of the Drosophila ER integral membrane protein, Jagunal. G3 (BETHESDA, MD.) 2023; 13:jkad059. [PMID: 36932646 PMCID: PMC10320142 DOI: 10.1093/g3journal/jkad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/19/2023]
Abstract
The mechanism surrounding chromosome inheritance during cell division has been well documented, however, organelle inheritance during mitosis is less understood. Recently, the endoplasmic reticulum (ER) has been shown to reorganize during mitosis, dividing asymmetrically in proneuronal cells prior to cell fate selection, indicating a programmed mechanism of inheritance. ER asymmetric partitioning in proneural cells relies on the highly conserved ER integral membrane protein, Jagunal (Jagn). Knockdown of Jagn in the compound Drosophila eye displays a pleotropic rough eye phenotype in 48% of the progeny. To identify genes involved in Jagn dependent ER partitioning pathway, we performed a dominant modifier screen of the 3rd chromosome for enhancers and suppressors of this Jagn-RNAi-induced rough eye phenotype. We screened through 181 deficiency lines covering the 3L and 3R chromosomes and identified 12 suppressors and 10 enhancers of the Jagn-RNAi phenotype. Based on the functions of the genes covered by the deficiencies, we identified genes that displayed a suppression or enhancement of the Jagn-RNAi phenotype. These include Division Abnormally Delayed (Dally), a heparan sulfate proteoglycan, the γ-secretase subunit Presenilin, and the ER resident protein Sec63. Based on our understanding of the function of these targets, there is a connection between Jagn and the Notch signaling pathway. Further studies will elucidate the role of Jagn and identified interactors within the mechanisms of ER partitioning during mitosis.
Collapse
Affiliation(s)
- Gerson Ascencio
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Matthew A de Cruz
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Judy Abuel
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Sydney Alvarado
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Yuma Arriaga
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Emily Conrad
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Alonso Castro
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Katharine Eichelberger
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Laura Galvan
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Grace Gundy
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | | | - Alyssa Jimenez
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Nhien Tuyet Lu
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Catharine Lugar
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Ronald Marania
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Tserendavaa Mendsaikhan
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Jose Ortega
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Natasha Nand
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Nicole S Rodrigues
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Khayla Shabazz
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Cynnie Tam
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Emmanuel Valenciano
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Clive Hayzelden
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Anthony S Eritano
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| | - Blake Riggs
- Department of Biology, San Francisco State University, 1600 Holloway Ave., San Francisco, CA 4132, USA
| |
Collapse
|
33
|
Monsen VT, Attramadal H. Structural insights into regulation of CCN protein activities and functions. J Cell Commun Signal 2023:10.1007/s12079-023-00768-5. [PMID: 37245184 DOI: 10.1007/s12079-023-00768-5] [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: 02/15/2023] [Accepted: 05/07/2023] [Indexed: 05/29/2023] Open
Abstract
CCN proteins play important functions during development, in repair mechanisms following tissue injury, as well as in pathophysiologic mechanisms of metastasis of cancer. CCNs are secreted proteins that have a multimodular structure and are categorized as matricellular proteins. Although the prevailing view is that CCN proteins regulate biologic processes by interacting with a wide array of other proteins in the microenvironment of the extracellular matrix, the molecular mechanisms of action of CCN proteins are still poorly understood. Not dissuading the current view, however, the recent appreciation that these proteins are signaling proteins in their own right and may even be considered preproproteins controlled by endopeptidases to release a C-terminal bioactive peptide has opened new avenues of research. Also, the recent resolution of the crystal structure of two of the domains of CCN3 have provided new knowledge with implications for the entire CCN family. These resolved structures in combination with structural predictions based upon the AlphaFold artificial intelligence tool provide means to shed new light on CCN functions in context of the notable literature in the field. CCN proteins have emerged as important therapeutic targets in several disease conditions, and clinical trials are currently ongoing. Thus, a review that critically discusses structure - function relationship of CCN proteins, in particular as it relates to interactions with other proteins in the extracellular milieu and on the cell surface, as well as to cell signaling activities of these proteins, is very timely. Suggested mechanism for activation and inhibition of signaling by the CCN protein family (graphics generated with BioRender.com ).
Collapse
Affiliation(s)
- Vivi Talstad Monsen
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
34
|
Li C, Ribes MA, Raftery R, Nwoko U, Warman ML, Craft AM. Directed differentiation of human pluripotent stem cells into articular cartilage reveals effects caused by absence of WISP3 , the gene responsible for Progressive Pseudorheumatoid Arthropathy of Childhood. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.01.535214. [PMID: 37066225 PMCID: PMC10103998 DOI: 10.1101/2023.04.01.535214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Objectives Progressive Pseudorheumatoid Arthropathy of Childhood (PPAC), caused by deficiency of WNT1 inducible signaling pathway protein 3 ( WISP3 ), has been challenging to study because no animal model of the disease exists and cartilage recovered from affected patients is indistinguishable from common end-stage osteoarthritis. Therefore, to gain insights into why precocious articular cartilage failure occurs in this disease, we made in vitro derived articular cartilage using isogenic WISP3 -deficient and WISP3 -sufficient human pluripotent stem cells (hPSCs). Methods We generated articular cartilage-like tissues from induced-(i)PSCs from 2 patients with PPAC and 1 wild-type human embryonic stem cell line in which we knocked out WISP3. We compared these tissues to in vitro -derived articular cartilage tissues from 2 isogenic WISP3 -sufficient control lines using histology, bulk RNA sequencing, single cell RNA sequencing, and in situ hybridization. Results WISP3 -deficient and WISP3 -sufficient hPSCs both differentiated into articular cartilage-like tissues that appeared histologically similar. However, the transcriptomes of WISP3 -deficient tissues differed significantly from WISP3 -sufficient tissues and pointed to increased TGFβ, TNFα/NFkB, and IL-2/STAT5 signaling and decreased oxidative phosphorylation. Single cell sequencing and in situ hybridization revealed that WISP3 -deficient cartilage contained a significantly higher fraction (∼ 4-fold increase, p < 0.001) of superficial zone chondrocytes compared to deeper zone chondrocytes than did WISP3 -sufficient cartilage. Conclusions WISP3 -deficient and WISP3 -sufficient hPSCs can be differentiated into articular cartilage-like tissues, but these tissues differ in their transcriptomes and in the relative abundances of chondrocyte sub-types they contain. These findings provide important starting points for in vivo studies when an animal model of PPAC or presymptomtic patient-derived articular cartilage becomes available. KEY MESSAGES What is already known on this topic: Loss-of-function mutations in WISP3 cause Progressive Pseudorheumatoid Arthropathy of Childhood (PPAC), yet the precise function of WISP3 in cartilage is unknown due to the absence of cartilage disease Wisp3 knockout mice and the lack of available PPAC patient cartilage that is not end-stage. Thus, most functional studies of WISP3 have been performed in vitro using WISP3 over-expressing cell lines (i.e., not wild-type) and WISP3 -deficient chondrocytes. What this study adds: We describe 3 new WISP3 -deficient human pluripotent stem cell (hPSC) lines and show they can be differentiated into articular cartilage-like tissue. We compare in vitro -derived articular cartilage made from WISP3 -deficient and isogenic WISP3 - sufficient hPSCs using bulk RNA sequencing, single cell RNA sequencing, and in situ hybridization. We observe significant differences in the expression of genes previously associated with cartilage formation and homeostasis in the TGFβ, TNFα/NFkB, and IL-2/STAT5 signaling pathways. We also observe that WISP3-deficient cartilage-like tissues contain significantly higher fractions of chondrocytes that express superficial zone transcripts. These data suggest precocious cartilage failure in PPAC is the result of abnormal articular cartilage formation, dysregulated homeostatic signaling, or both.How this study might affect research, practice or policy: This study uses in vitro -derived articular cartilage to generate hypotheses for why cartilage fails in children with PPAC. This work prioritizes downstream studies to be performed when pre-symptomatic patient-derived cartilage samples or animal model of PPAC becomes available. It is essential to know how WISP3 functions in cartilage to develop therapies that benefit patients with PPAC and other degenerative joint diseases.
Collapse
|
35
|
Garrett EC, Bielawski AM, Ruchti E, Sherer LM, Waghmare I, Hess-Homeier D, McCabe BD, Stowers RS, Certel SJ. The matricellular protein Drosophila Cellular Communication Network Factor is required for synaptic transmission and female fertility. Genetics 2023; 223:iyac190. [PMID: 36602539 PMCID: PMC9991515 DOI: 10.1093/genetics/iyac190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 01/06/2023] Open
Abstract
Within the extracellular matrix, matricellular proteins are dynamically expressed nonstructural proteins that interact with cell surface receptors, growth factors, and proteases, as well as with structural matrix proteins. The cellular communication network factors family of matricellular proteins serve regulatory roles to regulate cell function and are defined by their conserved multimodular organization. Here, we characterize the expression and neuronal requirement for the Drosophila cellular communication network factor family member. Drosophila cellular communication network factor is expressed in the nervous system throughout development including in subsets of monoamine-expressing neurons. Drosophila cellular communication network factor-expressing abdominal ganglion neurons innervate the ovaries and uterus and the loss of Drosophila cellular communication network factor results in reduced female fertility. In addition, Drosophila cellular communication network factor accumulates at the synaptic cleft and is required for neurotransmission at the larval neuromuscular junction. Analyzing the function of the single Drosophila cellular communication network factor family member will enhance our potential to understand how the microenvironment impacts neurotransmitter release in distinct cellular contexts and in response to activity.
Collapse
Affiliation(s)
| | - Ashley M Bielawski
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Evelyne Ruchti
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Lewis M Sherer
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Indrayani Waghmare
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - David Hess-Homeier
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Brian D McCabe
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - R Steven Stowers
- Department of Cell Biology and Microbiology, Montana State University, Bozeman, MT 59717, USA
| | - Sarah J Certel
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
36
|
Ganguly A, Padhan DK, Sengupta A, Chakraborty P, Sen M. CCN6 influences transcription and controls mitochondrial mass and muscle organization. FASEB J 2023; 37:e22815. [PMID: 36794678 DOI: 10.1096/fj.202201533r] [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: 09/22/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
Mutations in Cellular Communication Network Factor 6 (CCN6) are linked to the debilitating musculoskeletal disease Progressive Pseudo Rheumatoid Dysplasia (PPRD), which disrupts mobility. Yet, much remains unknown about CCN6 function at the molecular level. In this study, we revealed a new function of CCN6 in transcriptional regulation. We demonstrated that CCN6 localizes to chromatin and associates with RNA Polymerase II in human chondrocyte lines. Using zebrafish as a model organism we validated the nuclear presence of CCN6 and its association with RNA Polymerase II in different developmental stages from 10 hpf embryo to adult fish muscle. In concurrence with these findings, we confirmed the requirement of CCN6 in the transcription of several genes encoding mitochondrial electron transport complex proteins in the zebrafish, both in the embryonic stages and in the adult muscle. Reduction in the expression of these genes upon morpholino-mediated knockdown of CCN6 protein expression led to reduced mitochondrial mass, which correlated with defective myotome organization during zebrafish muscle development. Overall, this study suggests that the developmental musculoskeletal abnormalities linked with PPRD could be contributed at least partly by impaired expression of genes encoding mitochondrial electron transport complexes due to defects in CCN6 associated transcriptional regulation.
Collapse
Affiliation(s)
- Ananya Ganguly
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Deepesh Kumar Padhan
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Archya Sengupta
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Pritam Chakraborty
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Biochemistry and Molecular Biology, Southern Illinois University, USA
| | - Malini Sen
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
37
|
Song Y, Li C, Luo Y, Guo J, Kang Y, Yin F, Ye L, Sun D, Yu J, Zhang X. CCN6 improves hepatic steatosis, inflammation, and fibrosis in non-alcoholic steatohepatitis. Liver Int 2023; 43:357-369. [PMID: 36156376 DOI: 10.1111/liv.15430] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/20/2022] [Accepted: 09/20/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND AIMS CCN6 is a secretory protein with functions of maintaining mitochondrial homeostasis and anti-oxidative stress; and yet, whether it is involved in the pathogenesis of non-alcoholic steatohepatitis (NASH) is still obscure. We investigated the role and mechanism of CCN6 in the development of NASH. METHODS Human liver tissue samples were collected to detect the expression profile of CCN6. High-fat-high-cholesterol (HFHC) and methionine choline-deficient (MCD) diet were applied to mice to establish NASH animal models. Liver-specific overexpression of CCN6 was induced in mice by tail vein injection of adeno-associated virus (AAV), and then the effect of CCN6 on the course of NASH was observed. Free fatty acid (FFA) was applied to HepG2 cells to construct the cell model of steatosis, and the effect of CCN6 was investigated by knocking down the expression of CCN6 through small interfering RNA (siRNA) transfection. RESULTS We found that CCN6 expression was significantly downregulated in the liver of NASH. We confirmed that liver-specific overexpression of CCN6 significantly attenuated hepatic steatosis, inflammation response and fibrosis in NASH mice. Based on RNA-seq analysis, we revealed that CCN6 significantly affected the MAPK pathway. Then, by interfering with apoptosis signal-regulating kinase 1 (ASK1), we identified the ASK1/MAPK pathway pairs as the targets of CCN6 action. CONCLUSIONS CCN6 protects against hepatic steatosis, inflammation response and fibrosis by inhibiting the activation of ASK1 along with its downstream MAPK signalling. CCN6 may be a potential therapeutic target for the treatment of NASH.
Collapse
Affiliation(s)
- Yiran Song
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chenyang Li
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuxin Luo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jinbo Guo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yaxing Kang
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fengrong Yin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lihong Ye
- Department of Pathology, Shijiazhuang Fifth Hospital, Shijiazhuang, China
| | - Donglei Sun
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jun Yu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaolan Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
38
|
Novel Anti-Cancer Products Targeting AMPK: Natural Herbal Medicine against Breast Cancer. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020740. [PMID: 36677797 PMCID: PMC9863744 DOI: 10.3390/molecules28020740] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/15/2023]
Abstract
Breast cancer is a common cancer in women worldwide. The existing clinical treatment strategies have been able to limit the progression of breast cancer and cancer metastasis, but abnormal metabolism, immunosuppression, and multidrug resistance involving multiple regulators remain the major challenges for the treatment of breast cancer. Adenosine 5'-monophosphate (AMP)-Activated Protein Kinase (AMPK) can regulate metabolic reprogramming and reverse the "Warburg effect" via multiple metabolic signaling pathways in breast cancer. Previous studies suggest that the activation of AMPK suppresses the growth and metastasis of breast cancer cells, as well as stimulating the responses of immune cells. However, some other reports claim that the development and poor prognosis of breast cancer are related to the overexpression and aberrant activation of AMPK. Thus, the role of AMPK in the progression of breast cancer is still controversial. In this review, we summarize the current understanding of AMPK, particularly the comprehensive bidirectional functions of AMPK in cancer progression; discuss the pharmacological activators of AMPK and some specific molecules, including the natural products (including berberine, curcumin, (-)-epigallocatechin-3-gallate, ginsenosides, and paclitaxel) that influence the efficacy of these activators in cancer therapy; and elaborate the role of AMPK as a potential therapeutic target for the treatment of breast cancer.
Collapse
|
39
|
Kablar B. Skeletal Muscle's Role in Prenatal Inter-organ Communication: A Phenogenomic Study with Qualitative Citation Analysis. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:1-19. [PMID: 37955769 DOI: 10.1007/978-3-031-38215-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Gene targeting in mice allows for a complete elimination of skeletal (striated or voluntary) musculature in the body, from the beginning of its development, resulting in our ability to study the consequences of this ablation on other organs. Here I focus on the relationship between the muscle and lung, motor neurons, skeleton, and special senses. Since the inception of my independent laboratory, in 2000, with my team, we published more than 30 papers (and a book chapter), nearly 400 pages of data, on these specific relationships. Here I trace, using Web of Science, nearly 600 citations of this work, to understand its impact. The current report contains a summary of our work and its impact, NCBI's Gene Expression Omnibus accession numbers of all our microarray data, and three clear future directions doable by anyone using our publicly available data. Together, this effort furthers our understanding of inter-organ communication during prenatal development.
Collapse
Affiliation(s)
- Boris Kablar
- Department of Medical Neuroscience, Anatomy and Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
40
|
Yuan Z, Wang J, Zhang H, Chai Y, Xu Y, Miao Y, Yuan Z, Zhang L, Jiang Z, Yu Q. Glycocholic acid aggravates liver fibrosis by promoting the up-regulation of connective tissue growth factor in hepatocytes. Cell Signal 2023; 101:110508. [PMID: 36341984 DOI: 10.1016/j.cellsig.2022.110508] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022]
Abstract
AIMS The precise role of bile acid in the progression of liver fibrosis has yet to be elucidated. In this study, common bile duct ligation was used as an in vivo mouse model for the evaluation of bile acids that promote liver connective tissue growth factor expression. MAIN METHODS Primary rat and mice hepatocytes, as well as primary rat hepatic stellate and HepaRG cells were evaluated as in vitro models for promoting the expression of connective tissue growth factor by bile acids. KEY FINDINGS Compared with taurochenodeoxycholic acid, glycochenodeoxycholic acid, and taurocholic acid, glycocholic acid (GCA) most strongly promoted the secretion of connective tissue growth factor in mouse primary hepatocytes, rat primary hepatocytes and HepaRGs. GCA did not directly promote the activation of hepatic stellate cells. The administration of GCA in mice with ligated bile ducts promotes the progression of liver fibrosis, which may promote the yes-associated protein of hepatocytes into the nucleus, resulting in the hepatocytes secreting more connective tissue growth factor for hepatic stellate cell activation. In conclusion, our data showed that GCA can induce the expression of connective tissue growth factor in hepatocytes by promoting the nuclear translocation of yes-associated protein, thereby activating hepatic stellate cells. SIGNIFICANCE Our findings help to elucidate the contribution of GCA to the progression of hepatic fibrosis in cholestatic disease and aid the clinical monitoring of cholestatic liver fibrosis development.
Collapse
Affiliation(s)
- Zihang Yuan
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Jie Wang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Haoran Zhang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanyuan Chai
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Yunxia Xu
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Yingying Miao
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Ziqiao Yuan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Luyong Zhang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhenzhou Jiang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Qinwei Yu
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
41
|
Burt MA, Kalejaiye TD, Bhattacharya R, Dimitrakakis N, Musah S. Adriamycin-Induced Podocyte Injury Disrupts the YAP-TEAD1 Axis and Downregulates Cyr61 and CTGF Expression. ACS Chem Biol 2022; 17:3341-3351. [PMID: 34890187 DOI: 10.1021/acschembio.1c00678] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The most severe forms of kidney diseases are often associated with irreversible damage to the glomerular podocytes, the highly specialized epithelial cells that encase glomerular capillaries and regulate the removal of toxins and waste from the blood. Several studies revealed significant changes to podocyte cytoskeletal structure during disease onset, suggesting possible roles of cellular mechanosensing in podocyte responses to injury. Still, this topic remains underexplored partly due to the lack of appropriate in vitro models that closely recapitulate human podocyte biology. Here, we leveraged our previously established method for the derivation of mature podocytes from human induced pluripotent stem cells (hiPSCs) to help uncover the roles of yes-associated protein (YAP), a transcriptional coactivator and mechanosensor, in podocyte injury response. We found that while the total expression levels of YAP remain relatively unchanged during Adriamycin (ADR)-induced podocyte injury, the YAP target genes connective tissue growth factor (CTGF) and cysteine-rich angiogenic inducer 61 (Cyr61) are significantly downregulated. Intriguingly, TEAD1 is significantly downregulated in podocytes injured with ADR. By examining multiple independent modes of cellular injury, we found that CTGF and Cyr61 expression are downregulated only when podocytes were exposed to molecules known to disrupt the cell's mechanical integrity or cytoskeletal structure. To our knowledge, this is the first report that the YAP-TEAD1 signaling axis is disrupted when stem cell-derived human podocytes experience biomechanical injury. Together, these results could help improve the understanding of kidney disease mechanisms and highlight CTGF and Cyr61 as potential therapeutic targets or biomarkers for patient stratification.
Collapse
Affiliation(s)
- Morgan A Burt
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Titilola D Kalejaiye
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Rohan Bhattacharya
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - Samira Musah
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, North Carolina 27708, United States
- Department of Medicine, Division of Nephrology, Duke University School of Medicine, Durham, North Carolina 27710, United States
- Department of Cell Biology, Duke University, Durham, North Carolina 27710, United States
| |
Collapse
|
42
|
Zolfaghari S, Kaasbøll OJ, Monsen VT, Sredic B, Hagelin EMV, Attramadal H. The carboxyl-terminal TSP1-homology domain is the biologically active effector peptide of matricellular protein CCN5 that counteracts profibrotic CCN2. J Biol Chem 2022; 299:102803. [PMID: 36529291 PMCID: PMC9860493 DOI: 10.1016/j.jbc.2022.102803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Cellular Communication Network (CCN) proteins have multimodular structures important for their roles in cellular responses associated with organ development and tissue homeostasis. CCN2 has previously been reported to be secreted as a preproprotein that requires proteolytic activation to release its bioactive carboxyl-terminal fragment. Here, our goal was to resolve whether CCN5, a divergent member of the CCN family with converse functions relative to CCN2, releases the TSP1 homology domain as its bioactive signaling entity. The recombinant CCN5 or CCN3 TSP1 homology domains were produced in ExpiCHO-S or DG44 CHO cells as secretory fusion proteins appended to the carboxyl-terminal end of His-Halo-Sumo or amino-terminal end of human albumin and purified from the cell culture medium. We tested these fusion proteins in various phosphokinase signaling pathways or cell physiologic assays. Fusion proteins with the CCN5 TSP1 domain inhibited key signaling pathways previously reported to be stimulated by CCN2, irrespective of fusion partner. The fusion proteins also efficiently inhibited CCN1/2-stimulated cell migration and gap closure following scratch wound of fibroblasts. Fusion protein with the CCN3 TSP1 domain inhibited these functions with similar efficacy and potency as that of the CCN5 TSP1 domain. The CCN5 TSP1 domain also recapitulated a positive regulatory function previously assigned to full-length CCN5, that is, induction of estrogen receptor-α mRNA expression in triple negative MDA-MB-231 mammary adenocarcinoma cells and inhibited epithelial-to-mesenchymal transition and CCN2-induced mammosphere formation of MCF-7 adenocarcinoma cells. In conclusion, the CCN5 TSP1 domain is the bioactive entity that confers the biologic functions of unprocessed CCN5.
Collapse
Affiliation(s)
- Sima Zolfaghari
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Vivi T. Monsen
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bojana Sredic
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
| | | | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway,For correspondence: Håvard Attramadal
| |
Collapse
|
43
|
Broeders M, van Rooij J, Oussoren E, van Gestel T, Smith C, Kimber S, Verdijk R, Wagenmakers M, van den Hout J, van der Ploeg A, Narcisi R, Pijnappel W. Modeling cartilage pathology in mucopolysaccharidosis VI using iPSCs reveals early dysregulation of chondrogenic and metabolic gene expression. Front Bioeng Biotechnol 2022; 10:949063. [PMID: 36561048 PMCID: PMC9763729 DOI: 10.3389/fbioe.2022.949063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022] Open
Abstract
Mucopolysaccharidosis type VI (MPS VI) is a metabolic disorder caused by disease-associated variants in the Arylsulfatase B (ARSB) gene, resulting in ARSB enzyme deficiency, lysosomal glycosaminoglycan accumulation, and cartilage and bone pathology. The molecular response to MPS VI that results in cartilage pathology in human patients is largely unknown. Here, we generated a disease model to study the early stages of cartilage pathology in MPS VI. We generated iPSCs from four patients and isogenic controls by inserting the ARSB cDNA in the AAVS1 safe harbor locus using CRISPR/Cas9. Using an optimized chondrogenic differentiation protocol, we found Periodic acid-Schiff positive inclusions in hiPSC-derived chondrogenic cells with MPS VI. Genome-wide mRNA expression analysis showed that hiPSC-derived chondrogenic cells with MPS VI downregulated expression of genes involved in TGF-β/BMP signalling, and upregulated expression of inhibitors of the Wnt/β-catenin signalling pathway. Expression of genes involved in apoptosis and growth was upregulated, while expression of genes involved in glycosaminoglycan metabolism was dysregulated in hiPSC-derived chondrogenic cells with MPS VI. These results suggest that human ARSB deficiency in MPS VI causes changes in the transcriptional program underlying the early stages of chondrogenic differentiation and metabolism.
Collapse
Affiliation(s)
- M. Broeders
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Jgj van Rooij
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, Netherlands
| | - E. Oussoren
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Tjm van Gestel
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Ca Smith
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Sj Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rm Verdijk
- Department of Pathology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Maem Wagenmakers
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, Netherlands
| | - Jmp van den Hout
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - At van der Ploeg
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - R. Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Wwmp Pijnappel
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
44
|
Hirose K, Kuwahara M, Nakata E, Tetsunaga T, Yamada K, Saiga K, Takigawa M, Ozaki T, Kubota S, Hattori T. Elevated Expression of CCN3 in Articular Cartilage Induces Osteoarthritis in Hip Joints Irrespective of Age and Weight Bearing. Int J Mol Sci 2022; 23:15311. [PMID: 36499638 PMCID: PMC9738275 DOI: 10.3390/ijms232315311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) occurs not only in the knee but also in peripheral joints throughout the whole body. Previously, we have shown that the expression of cellular communication network factor 3 (CCN3), a matricellular protein, increases with age in knee articular cartilage, and the misexpression of CCN3 in cartilage induces senescence-associated secretory phenotype (SASP) factors, indicating that CCN3 promotes cartilage senescence. Here, we investigated the correlation between CCN3 expression and OA degenerative changes, principally in human femoral head cartilage. Human femoral heads obtained from patients who received total hip arthroplasty were categorized into OA and femoral neck fracture (normal) groups without significant age differences. Gene expression analysis of RNA obtained from femoral head cartilage revealed that CCN3 and MMP-13 expression in the non-weight-bearing part was significantly higher in the OA group than in the normal group, whereas the weight-bearing OA parts and normal cartilage showed no significant differences in the expression of these genes. The expression of COL10A1, however, was significantly higher in weight-bearing OA parts compared with normal weight-bearing parts, and was also higher in weight-bearing parts compared with non-weight-bearing parts in the OA group. In contrast, OA primary chondrocytes from weight-bearing parts showed higher expression of CCN3, p16, ADAMTS4, and IL-1β than chondrocytes from the corresponding normal group, and higher ADAMTS4 and IL-1β in the non-weight-bearing part compared with the corresponding normal group. Acan expression was significantly lower in the non-weight-bearing group in OA primary chondrocytes than in the corresponding normal chondrocytes. The expression level of CCN3 did not show significant differences between the weight-bearing part and non-weight-bearing part in both OA and normal primary chondrocytes. Immunohistochemical analysis showed accumulated CCN3 and aggrecan neoepitope staining in both the weight-bearing part and non-weight-bearing part in the OA group compared with the normal group. The CCN3 expression level in cartilage had a positive correlation with the Mankin score. X-ray analysis of cartilage-specific CCN3 overexpression mice (Tg) revealed deformation of the femoral and humeral head in the early stage, and immunohistochemical analysis showed accumulated aggrecan neoepitope staining as well as CCN3 staining and the roughening of the joint surface in Tg femoral and humeral heads. Primary chondrocytes from the Tg femoral head showed enhanced expression of Ccn3, Adamts5, p16, Il-6, and Tnfα, and decreased expression of Col2a1 and -an. These findings indicate a correlation between OA degenerative changes and the expression of CCN3, irrespective of age and mechanical loading. Furthermore, the Mankin score indicates that the expression level of Ccn3 correlates with the progression of OA.
Collapse
Affiliation(s)
- Kazuki Hirose
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Miho Kuwahara
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Eiji Nakata
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Tomonori Tetsunaga
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Kazuki Yamada
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Kenta Saiga
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School/Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Satoshi Kubota
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Takako Hattori
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
45
|
Cárdenas-León CG, Mäemets-Allas K, Klaas M, Lagus H, Kankuri E, Jaks V. Matricellular proteins in cutaneous wound healing. Front Cell Dev Biol 2022; 10:1073320. [PMID: 36506087 PMCID: PMC9730256 DOI: 10.3389/fcell.2022.1073320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Cutaneous wound healing is a complex process that encompasses alterations in all aspects of the skin including the extracellular matrix (ECM). ECM consist of large structural proteins such as collagens and elastin as well as smaller proteins with mainly regulative properties called matricellular proteins. Matricellular proteins bind to structural proteins and their functions include but are not limited to interaction with cell surface receptors, cytokines, or protease and evoking a cellular response. The signaling initiated by matricellular proteins modulates differentiation and proliferation of cells having an impact on the tissue regeneration. In this review we give an overview of the matricellular proteins that have been found to be involved in cutaneous wound healing and summarize the information known to date about their functions in this process.
Collapse
Affiliation(s)
| | - Kristina Mäemets-Allas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Mariliis Klaas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Heli Lagus
- Department of Plastic Surgery and Wound Healing Centre, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Viljar Jaks
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia,Dermatology Clinic, Tartu University Clinics, Tartu, Estonia,*Correspondence: Viljar Jaks,
| |
Collapse
|
46
|
Waters JA, Urbano I, Robinson M, House CD. Insulin-like growth factor binding protein 5: Diverse roles in cancer. Front Oncol 2022; 12:1052457. [PMID: 36465383 PMCID: PMC9714447 DOI: 10.3389/fonc.2022.1052457] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022] Open
Abstract
Insulin-like growth factor binding proteins (IGFBPs) and the associated signaling components in the insulin-like growth factor (IGF) pathway regulate cell differentiation, proliferation, apoptosis, and adhesion. Of the IGFBPs, insulin-like growth factor binding protein 5 (IGFBP5) is the most evolutionarily conserved with a dynamic range of IGF-dependent and -independent functions, and studies on the actions of IGFBP5 in cancer have been somewhat paradoxical. In cancer, the IGFBPs respond to external stimuli to modulate disease progression and therapeutic responsiveness in a context specific manner. This review discusses the different roles of IGF signaling and IGFBP5 in disease with an emphasis on discoveries within the last twenty years, which underscore a need to clarify the IGF-independent actions of IGFBP5, the impact of its subcellular localization, the differential activities of each of the subdomains, and the response to elements of the tumor microenvironment (TME). Additionally, recent advances addressing the role of IGFBP5 in resistance to cancer therapeutics will be discussed. A better understanding of the contexts in which IGFBP5 functions will facilitate the discovery of new mechanisms of cancer progression that may lead to novel therapeutic opportunities.
Collapse
Affiliation(s)
- Jennifer A. Waters
- Biology Department, San Diego State University, San Diego, CA, United States
| | - Ixchel Urbano
- Biology Department, San Diego State University, San Diego, CA, United States
| | - Mikella Robinson
- Biology Department, San Diego State University, San Diego, CA, United States
| | - Carrie D. House
- Biology Department, San Diego State University, San Diego, CA, United States,Moore’s Cancer Center, University of California, San Diego, San Diego, CA, United States,*Correspondence: Carrie D. House,
| |
Collapse
|
47
|
Wei JR, Hao ZZ, Xu C, Huang M, Tang L, Xu N, Liu R, Shen Y, Teichmann SA, Miao Z, Liu S. Identification of visual cortex cell types and species differences using single-cell RNA sequencing. Nat Commun 2022; 13:6902. [PMID: 36371428 PMCID: PMC9653448 DOI: 10.1038/s41467-022-34590-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022] Open
Abstract
The primate neocortex exerts high cognitive ability and strong information processing capacity. Here, we establish a single-cell RNA sequencing dataset of 133,454 macaque visual cortical cells. It covers major cortical cell classes including 25 excitatory neuron types, 37 inhibitory neuron types and all glial cell types. We identified layer-specific markers including HPCAL1 and NXPH4, and also identified two cell types, an NPY-expressing excitatory neuron type that expresses the dopamine receptor D3 gene; and a primate specific activity-dependent OSTN + sensory neuron type. Comparisons of our dataset with humans and mice show that the gene expression profiles differ between species in relation to genes that are implicated in the synaptic plasticity and neuromodulation of excitatory neurons. The comparisons also revealed that glutamatergic neurons may be more diverse across species than GABAergic neurons and non-neuronal cells. These findings pave the way for understanding how the primary cortex fulfills the high-cognitive functions.
Collapse
Affiliation(s)
- Jia-Ru Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Mengyao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lei Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Nana Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ruifeng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuhui Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| | - Zhichao Miao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China.
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Genome Campus, Cambridge, UK.
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China.
| |
Collapse
|
48
|
An Update on In Vitro Folliculogenesis: A New Technique for Post-Cancer Fertility. Biomedicines 2022; 10:biomedicines10092217. [PMID: 36140316 PMCID: PMC9496077 DOI: 10.3390/biomedicines10092217] [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: 07/20/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: Obtaining in vitro mature oocytes from ovarian tissue to preserve women’s fertility is still a challenge. At present, there is a therapeutic deadlock for girls and women who need emergency fertility preservation in case of a high risk of ovary invasion by malignant cells. In such a case, ovarian tissue cannot be engrafted; an alternative could be in vitro folliculogenesis. Methods: This review focuses on the progress of in vitro folliculogenesis in humans. PubMed and Embase databases were used to search for original English-language articles. Results: The first phase of in vitro folliculogenesis is carried out in the original ovarian tissue. The addition of one (or more) initiation activator(s) is not essential but allows better yields and the use of a 3D culture system at this stage provides no added value. The second stage requires a mechanical and/or enzymatic isolation of the secondary follicles. The use of an activator and/or a 3D culture system is then necessary. Conclusion: The current results are promising but there is still a long way to go. Obtaining live births in large animals is an essential step in validating this in vitro folliculogenesis technique.
Collapse
|
49
|
Xi Y, LaCanna R, Ma HY, N'Diaye EN, Gierke S, Caplazi P, Sagolla M, Huang Z, Lucio L, Arlantico A, Jeet S, Brightbill H, Emson C, Wong A, Morshead KB, DePianto DJ, Roose-Girma M, Yu C, Tam L, Jia G, Ramalingam TR, Marsters S, Ashkenazi A, Kim SH, Kelly R, Wu S, Wolters PJ, Feldstein AE, Vander Heiden JA, Ding N. A WISP1 antibody inhibits MRTF signaling to prevent the progression of established liver fibrosis. Cell Metab 2022; 34:1377-1393.e8. [PMID: 35987202 DOI: 10.1016/j.cmet.2022.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 06/06/2022] [Accepted: 07/19/2022] [Indexed: 01/18/2023]
Abstract
Fibrosis is the major risk factor associated with morbidity and mortality in patients with non-alcoholic steatohepatitis (NASH)-driven chronic liver disease. Although numerous efforts have been made to identify the mediators of the initiation of liver fibrosis, the molecular underpinnings of fibrosis progression remain poorly understood, and therapies to arrest liver fibrosis progression are elusive. Here, we identify a pathway involving WNT1-inducible signaling pathway protein 1 (WISP1) and myocardin-related transcription factor (MRTF) as a central mechanism driving liver fibrosis progression through the integrin-dependent transcriptional reprogramming of myofibroblast cytoskeleton and motility. In mice, WISP1 deficiency protects against fibrosis progression, but not fibrosis onset. Moreover, the therapeutic administration of a novel antibody blocking WISP1 halted the progression of existing liver fibrosis in NASH models. These findings implicate the WISP1-MRTF axis as a crucial determinant of liver fibrosis progression and support targeting this pathway by antibody-based therapy for the treatment of NASH fibrosis.
Collapse
Affiliation(s)
- Ying Xi
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Ryan LaCanna
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Hsiao-Yen Ma
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Elsa-Noah N'Diaye
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Sarah Gierke
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Patrick Caplazi
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Meredith Sagolla
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Zhiyu Huang
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Laura Lucio
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Alexander Arlantico
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Surinder Jeet
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Hans Brightbill
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Claire Emson
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Aaron Wong
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Katrina B Morshead
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Daryle J DePianto
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Merone Roose-Girma
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Charles Yu
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Lucinda Tam
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Guiquan Jia
- Department of Biomarker Discovery, Genentech, South San Francisco, CA, USA
| | | | - Scot Marsters
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Avi Ashkenazi
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Si Hyun Kim
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Ryan Kelly
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Shuang Wu
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Paul J Wolters
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ariel E Feldstein
- Department of Pediatrics, University of California, San Diego, San Diego, CA, USA
| | | | - Ning Ding
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
50
|
Nguyen MT, Lee MA, Kim YK, Kook H, Jeong D, Jang SP, Kwak TH, Park WJ. The matricellular protein CCN5 induces apoptosis in myofibroblasts through SMAD7-mediated inhibition of NFκB. PLoS One 2022; 17:e0269735. [PMID: 35917315 PMCID: PMC9345366 DOI: 10.1371/journal.pone.0269735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
We previously showed that the matricellular protein CCN5 reverses established cardiac fibrosis (CF) through inducing apoptosis in myofibroblasts (MyoFBs) but not in cardiomyocytes or fibroblasts (FBs). In this study, we set out to elucidate the molecular mechanisms underlying CCN5-mediated selective apoptosis of MyoFBs. We first observed that the apoptotic protein p53 and the anti-apoptotic protein NFκB are simultaneously induced in MyoFBs. When the expression level of p53 was suppressed using a siRNA, CCN5 did not induce apoptosis in MyoFBs. By contrast, when NFκB signaling was inhibited using IKK VII, an IκB inhibitor, MyoFBs underwent apoptosis even in the absence of CCN5. SMAD7 is one of the downstream targets of CCN5 and it was previously shown to potentiate apoptosis in epithelial cells through inhibition of NFκB. In accordance with these reports, when the expression of SMAD7 was suppressed using a siRNA, NFκB signaling was enhanced, and CCN5 did not induce apoptosis. Lastly, we used a luciferase reporter construct to show that CCN5 positively regulated SMAD7 expression at the transcriptional level. Collectively, our data suggest that a delicate balance between the two mutually antagonistic proteins p53 and NFκB is maintained for MyoFBs to survive, and CCN5 tips the balance in favor of the apoptotic protein p53. This study provides insight into the anti-fibrotic activity of CCN5 during the regression of CF.
Collapse
Affiliation(s)
- Mai Tuyet Nguyen
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Min-Ah Lee
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Jeollanam-do, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanam-do, Republic of Korea
| | - Dongtak Jeong
- Department of Molecular & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, Gyeonggi-do, Republic of Korea
| | - Seung Pil Jang
- BethphaGen, S3-203, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Tae Hwan Kwak
- BethphaGen, S3-203, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- * E-mail:
| |
Collapse
|