1
|
Lee HK, Notario GR, Won SY, Kim JH, Lee SM, Kim HS, Cho SR. Elevated sclerostin levels contribute to reduced bone mineral density in non-ambulatory stroke patients. Bone Rep 2025; 25:101829. [PMID: 40225703 PMCID: PMC11986488 DOI: 10.1016/j.bonr.2025.101829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 04/15/2025] Open
Abstract
Osteoporosis following stroke is a significant impediment to patient recovery. Decreased mechanical loading and locomotion following the onset of paralysis in stroke patients, especially those who are non-ambulatory, contributes greatly to bone loss. Sclerostin, a protein encoded by the SOST gene, accumulates as a result of reduced mechanical loading and inhibits bone formation. This study explores the relationship between mechanical unloading, sclerostin levels, and bone mineral density (BMD) in stroke patients, utilizing three cohorts. Analysis of Cohort 1, consisting of patients with available sclerostin level measurements, found significantly elevated sclerostin levels in non-ambulatory patients compared to ambulatory patients, indicating the influence of ambulatory status on sclerostin regulation. Cohort 2, consisting of patients with BMD measurements, demonstrated that prolonged mechanical unloading in non-ambulatory patients resulted in a greater decline in BMD over time. Analysis in Cohort 3 patients, who had bilateral BMD measurements available, revealed that hemiplegic sides subjected to reduced mechanical loading exhibited lower BMD compared to non-hemiplegic sides. These findings collectively confirm the hypothesis that reduced mechanical loading elevates sclerostin levels and accelerates bone loss. By integrating data across the three cohorts, this study underscores the critical impact of mechanical unloading on bone health, particularly in chronic stroke patients with limited mobility. Our study provides clinical insights for treatments integrating ambulatory status, sclerostin levels, and BMD in chronic stroke patients and highlights an increased need for therapeutics targeting mechanical loading pathways and sclerostin accumulation which can be administered to treat chronic osteoporosis following stroke.
Collapse
Affiliation(s)
- Hye Kyoung Lee
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Geneva Rose Notario
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Young Won
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Hwan Kim
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su Min Lee
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Nursing, Yonsei University College of Nursing, Seoul, Republic of Korea
| | - Ha Seong Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Seosong Hospital, Incheon, Republic of Korea
| | - Sung-Rae Cho
- Department of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Yang S, Li K, Zhou Q, Zhang X, Guo D. Broad-Spectrum Antiviral Activity of the Orally Bioavailable Antiviral ATV014 Against Multiple Coronaviruses. MedComm (Beijing) 2025; 6:e70186. [PMID: 40242157 PMCID: PMC12000680 DOI: 10.1002/mco2.70186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/31/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Affiliation(s)
- Sidi Yang
- Guangzhou National LaboratoryGuangzhou International Bio IslandGuangzhouGuangdongChina
| | - Kun Li
- Guangzhou National LaboratoryGuangzhou International Bio IslandGuangzhouGuangdongChina
| | - Qifan Zhou
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and SynthesisDepartment of ChemistryCollege of Science, Academy for Advanced Interdisciplinary Studies and Medi‐X PingshanSouthern University of Science and TechnologyShenzhenGuangdongChina
| | - Xumu Zhang
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and SynthesisDepartment of ChemistryCollege of Science, Academy for Advanced Interdisciplinary Studies and Medi‐X PingshanSouthern University of Science and TechnologyShenzhenGuangdongChina
| | - Deyin Guo
- Guangzhou National LaboratoryGuangzhou International Bio IslandGuangzhouGuangdongChina
| |
Collapse
|
3
|
Simpson KL, Rothwell DG, Blackhall F, Dive C. Challenges of small cell lung cancer heterogeneity and phenotypic plasticity. Nat Rev Cancer 2025:10.1038/s41568-025-00803-0. [PMID: 40211072 DOI: 10.1038/s41568-025-00803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 04/12/2025]
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy with ~7% 5-year overall survival reflecting early metastasis and rapid acquired chemoresistance. Immunotherapy briefly extends overall survival in ~15% cases, yet predictive biomarkers are lacking. Targeted therapies are beginning to show promise, with a recently approved delta-like ligand 3 (DLL3)-targeted therapy impacting the treatment landscape. The increased availability of patient-faithful models, accumulating human tumour biobanks and numerous comprehensive molecular profiling studies have collectively facilitated the mapping and understanding of substantial intertumoural and intratumoural heterogeneity. Beyond the almost ubiquitous loss of wild-type p53 and RB1, SCLC is characterized by heterogeneously mis-regulated expression of MYC family members, yes-associated protein 1 (YAP1), NOTCH pathway signalling, anti-apoptotic BCL2 and epigenetic regulators. Molecular subtypes are based on the neurogenic transcription factors achaete-scute homologue 1 (ASCL1) and neurogenic differentiation factor 1 (NEUROD1), the rarer non-neuroendocrine transcription factor POU class 2 homeobox 3 (POU2F3), and immune- and inflammation-related signatures. Furthermore, SCLC shows phenotypic plasticity, including neuroendocrine-to-non-neuroendocrine transition driven by NOTCH signalling, which is associated with disease progression, chemoresistance and immune modulation and, in mouse models, with metastasis. Although these features pose substantial challenges, understanding the molecular vulnerabilities of transcription factor subtypes, the functional relevance of plasticity and cell cooperation offer opportunities for personalized therapies informed by liquid and tissue biomarkers.
Collapse
Affiliation(s)
- Kathryn L Simpson
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Dominic G Rothwell
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Fiona Blackhall
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Medical Oncology, Christie Hospital National Health Service, Foundation Trust, Manchester, UK
| | - Caroline Dive
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK.
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK.
- CRUK Lung Cancer Centre of Excellence, Manchester, UK.
| |
Collapse
|
4
|
Dreyer TJ, Keen JAC, Wells LM, Hopkinson M, Orriss IR, Holdsworth G, Pitsillides AA, Roberts SJ. Porcupine inhibition is a promising pharmacological treatment for severe sclerosteosis pathologies. Bone Res 2025; 13:44. [PMID: 40189599 PMCID: PMC11973224 DOI: 10.1038/s41413-025-00406-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 04/09/2025] Open
Abstract
Sclerosteosis, an ultra-rare disorder characterised by high bone mass (HBM) and skeletal overgrowth, leads to facial paralysis, hearing loss and raised intracranial pressure, which is currently managed only through high-risk surgery. Sclerosteosis is caused by SOST mutations and loss of functional sclerostin, a protein that suppresses osteogenesis by antagonising Wnt/β-catenin signalling. Herein, using in vitro and in vivo approaches, we explore whether LGK974, another potent Wnt inhibitor that targets porcupine (PORCN, Wnt-specific acyltransferase), is a promising sclerosteosis therapeutic. In vitro assays showed that 100 nmol/L LGK974 significantly reduced osteoblast alkaline phosphatase (ALP) activity/mineralisation, decreased Wnt/osteoblast marker (Axin2, Runx2 and Ocn) expression, and downregulated ossification and the Wnt signalling pathway, without affecting osteoclast numbers/resorption. To assess in vivo effects, 6-week-old male and female Sost deficient (Sost-/-) mice received LGK974 for 4 weeks and right hindlimbs were subjected to 20 N peak loading to assess mechanoadaptive interactions. µCT revealed significant reductions in vertebral trabecular number and lower cortical bone volume in loaded and non-loaded tibiae in male and female LGK974-treated Sost-/- mice. Interestingly, the target engagement biomarker Axin2 was only significantly reduced in male vertebrae, which may indicate differences in male and female response to LGK974. This study also shows that PORCN inhibition may effectively limit characteristic HBM and skeletal overgrowth in sclerosteosis patients at sites with severe pathology.
Collapse
Affiliation(s)
- Timothy J Dreyer
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Jacob A C Keen
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Leah M Wells
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Mark Hopkinson
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Isabel R Orriss
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | | | - Andrew A Pitsillides
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Scott J Roberts
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK.
| |
Collapse
|
5
|
Mohr-Allen SR, Gleghorn JP, Varner VD. Fluid secretion and luminal pressure control lateral branching morphogenesis in the embryonic avian lung. Dev Biol 2025; 520:251-263. [PMID: 39870322 DOI: 10.1016/j.ydbio.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
During lung development, the embryonic airway originates as a wishbone-shaped epithelial tube, which undergoes a series of branching events to build the bronchial tree. This process depends crucially on cell proliferation and is thought to involve distinct branching modes: lateral branching, wherein daughter branches emerge along the length of a parent branch, and bifurcations, wherein the tip of a parent branch splits to form two new daughter branches. The developing airway is fluid-filled, and previous studies have shown that altered luminal pressure can influence rates of branching morphogenesis. However, it is not clear if altered tissue mechanics influence patterns of proliferation along the embryonic airway epithelium nor if individual branching modes are affected differently by changes in luminal pressure. Here, we focused on mechanisms of lateral branching and used as a model system the embryonic avian lung, which forms exclusively via this branching mode during early development. We used microinjected fluid droplets or pharmacological modulators of fluid secretion to alter luminal fluid pressure either locally or globally within cultured embryonic lungs. Somewhat surprisingly, we found both local and global increases in luminal pressure to suppress the formation of new lateral branches while also promoting increased epithelial proliferation. In a consistent manner, decreased luminal pressure led to an increase in lateral branching morphogenesis. Morphometric analysis of airway branching patterns revealed that altered luminal pressure shifts the overall branching program, rather than simply changing rates of morphogenesis. Taken together, these results highlight the importance of mechanical forces during airway branching and suggest that different branching modes may be affected differently by luminal fluid pressure.
Collapse
Affiliation(s)
- Shelby R Mohr-Allen
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA; Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Victor D Varner
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA; Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
6
|
Brouns I, Adriaensen D, Timmermans JP. The pulmonary neuroepithelial body microenvironment represents an underestimated multimodal component in airway sensory pathways. Anat Rec (Hoboken) 2025; 308:1094-1117. [PMID: 36808710 DOI: 10.1002/ar.25171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 02/22/2023]
Abstract
Exciting new imaging and molecular tools, combined with state-of-the-art genetically modified mouse models, have recently boosted interest in pulmonary (vagal) sensory pathway investigations. In addition to the identification of diverse sensory neuronal subtypes, visualization of intrapulmonary projection patterns attracted renewed attention on morphologically identified sensory receptor end-organs, such as the pulmonary neuroepithelial bodies (NEBs) that have been our area of expertise for the past four decades. The current review aims at providing an overview of the cellular and neuronal components of the pulmonary NEB microenvironment (NEB ME) in mice, underpinning the role of these complexly organized structures in the mechano- and chemosensory potential of airways and lungs. Interestingly, the pulmonary NEB ME additionally harbors different types of stem cells, and emerging evidence suggests that the signal transduction pathways that are active in the NEB ME during lung development and repair also determine the origin of small cell lung carcinoma. Although documented for many years that NEBs appear to be affected in several pulmonary diseases, the current intriguing knowledge on the NEB ME seems to encourage researchers that are new to the field to explore the possibility that these versatile sensor-effector units may be involved in lung pathogenesis or pathobiology.
Collapse
Affiliation(s)
- Inge Brouns
- Laboratory of Cell Biology and Histology (CBH), Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Dirk Adriaensen
- Laboratory of Cell Biology and Histology (CBH), Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology (CBH), Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
7
|
Hosseini F, Azadmehr A, Saleki K, Ahmadifard M, Oladnabi M, Shirzad M, Javanian M. Neuropilin-1 as a Neuroinflammatory Entry Factor for SARS-CoV-2 Is Attenuated in Vaccinated COVID-19 Patients: A Case-Control Study. Health Sci Rep 2025; 8:e70630. [PMID: 40196385 PMCID: PMC11973440 DOI: 10.1002/hsr2.70630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Background and Aim COVID-19 is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) as a pandemic infectious disease. So far, it has been known that this virus uses several receptors to enter the host cell, one of which is neuropilin-1 (NRP1). Also, one of the main causes of clinical manifestations, severity of disease, and mortality of patients is cytokine storm syndrome, one of these cytokines being interleukin (IL)-6. Our aim was to study the level of expression of NRP1 and IL-6 genes in COVID-19 patients by using peripheral blood mononuclear cells (PBMCs). Materials and Methods Our study population included the test group (80 patients with COVID-19) and the control group (30 healthy individuals). Venous blood was taken from all subjects. After isolating PBMCs from blood using Ficoll, RNA was extracted. Then, cDNA synthesis, the expression level of NRP1 and IL-6 compared to GAPDH housekeeping gene was measured by real-time PCR. Results The level of NRP1 gene expression was increased significantly in COVID-19 different groups compared to the control group. Surprisingly, it was observed that the amount of NRP1 gene decreased in the vaccinated group compared to nonvaccinated groups. IL-6 gene expression was also significantly increased in all groups except vaccinated patients compared to the control group. Also, the results indicated that there was a positive and statistically considerable relationship between IL-6 expression level and NRP1 expression level (p = 0.03). Conclusion The significant increase in the expression of NRP1 and IL-6 genes in COVID-19 patients, especially in moderate and severe cases, indicates their potential involvement in the progression of the disease, which may serve as biomarkers of disease severity. Also, since these genes play an important role in causing severe inflammation, cytokine storm, and immunopathological complications of COVID-19, further investigations maybe needed to achieve therapeutic goals to control COVID-19 and similar diseases.
Collapse
Affiliation(s)
- Faezeh Hosseini
- Student Research Committee, Babol University of Medical SciencesBabolIran
- Cellular and Molecular Biology Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| | - Abbas Azadmehr
- Cellular and Molecular Biology Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical SciencesBabolIran
- USERN OfficeBabol University of Medical SciencesBabolIran
- Department of E‐Learning in Medical SciencesFaculty of Medical Education and Learning Technologies, Shahid Beheshti University of Medical SciencesTehranIran
| | - Mohamadreza Ahmadifard
- Cellular and Molecular Biology Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| | - Morteza Oladnabi
- Ischemic Disorders Research Center, Golestan University of Medical SciencesGorganIran
| | - Moein Shirzad
- Cellular and Molecular Biology Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| | - Mostafa Javanian
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical SciencesBabolIran
| |
Collapse
|
8
|
Guo L, Peng Y, Yang C, Liu X, Xiong W, Liao W, Fan J. Mechanistic studies on the role of CHI3L1 in eosinophilic inflammation in chronic sinusitis. Front Immunol 2025; 16:1562546. [PMID: 40201175 PMCID: PMC11975569 DOI: 10.3389/fimmu.2025.1562546] [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: 01/17/2025] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
More than 10% of adults suffer from chronic rhinosinusitis (CRS), a chronic inflammatory condition that lowers quality of life, reduces productivity, and shortens work hours. Every year, more than 1 million surgeries are performed worldwide as a result of CRS. In recent years, targeted therapy for CRS has become a hotspot of research at home and abroad and has made significant progress, but CRS still has a high recurrence rate. Therefore CRS urgently needs precise targeted therapy. In the pathological process of CRS, the involvement of eosinophils is an important inflammatory mechanism. And excessive aggregation of eosinophils often leads to severe inflammatory responses. Studies have shown that chitinase 3-like protein 1 (CHI3L1) plays a key role in the activation and migration of eosinophils. This review will combine the latest research results to analyse in detail the biological properties of CHI3L1, its expression pattern in CRS, and the possible mechanisms by which it affects eosinophil aggregation by regulating immune responses and inflammatory processes, which will provide insights into the key role of CHI3L1 in the pathological process of CRS and offer a new target for the treatment of CRS.
Collapse
Affiliation(s)
- Ling Guo
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Peng
- Department of Otolaryngology Head and Neck Surgery, Chengdu Second People's Hospital, Chengdu, China
| | - Cheng Yang
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinghong Liu
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Weilan Xiong
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weijiang Liao
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiangang Fan
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
9
|
Jiang H, Zhu T, Chang Z, Liu Z, Ou W, Wang S. A Recurrent Small Cell Lung Carcinoma Harboring an EML4-ALK Fusion Mutation with Sustained Response to Ensartinib: A Case Report. Curr Oncol 2025; 32:163. [PMID: 40136367 PMCID: PMC11941435 DOI: 10.3390/curroncol32030163] [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: 01/05/2025] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine tumor. Lung cancer patients with ALK and EML4 fusions respond significantly to ALK inhibitors. The EML4-ALK fusion gene mutation is the result of an inversion of chromosome 2, which juxtaposes the 5 end of the EML4 gene with the 3 end of the ALK gene. In SCLC, the frequency of fusion genes is very low, and to the best of our knowledge, only four cases of ALK fusion gene mutations in SCLC have been reported. In this report, we describe the treatment of a 74-year-old female patient with SCLC who developed recurrence of hilar lymph node metastasis three years after surgical resection. Postoperative NGS showed that this patient is a SCLC patient harboring a rare EML4-ALK fusion mutation, and a satisfactory 43-month overall survival (OS) was achieved after treatment with ensartinib targeting the EML4-ALK fusion gene mutation. The ALK-TKI may be a new treatment option for these patients. This article provides a therapeutic reference.
Collapse
Affiliation(s)
| | | | | | | | | | - Siyu Wang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Yuexiu District, Guangzhou 510060, China; (H.J.); (T.Z.); (Z.C.); (Z.L.); (W.O.)
| |
Collapse
|
10
|
Su H, Chen L, Wu J, Cheng Z, Li J, Ren Y, Xu J, Dang Y, Zheng M, Cao Y, Gao J, Dai C, Hu X, Xie H, Chen J, Luo T, Zhu J, Wu C, Sha W, Chen C, Liu H. Proteogenomic characterization reveals tumorigenesis and progression of lung cancer manifested as subsolid nodules. Nat Commun 2025; 16:2414. [PMID: 40069142 PMCID: PMC11897189 DOI: 10.1038/s41467-025-57364-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Lung adenocarcinoma (LUAD) radiologically displayed as subsolid nodules (SSNs) is prevalent. Nevertheless, the precise clinical management of SSNs necessitates a profound understanding of their tumorigenesis and progression. Here, we analyze 66 LUAD displayed as SSNs covering 3 histological stages including adenocarcinoma in situ (AIS), minimally invasive adenocarcinoma (MIA) and invasive adenocarcinoma (IAC) by incorporating genomics, proteomics, phosphoproteomics and glycoproteomics. Intriguingly, cholesterol metabolism is aberrantly regulated in the preneoplastic AIS stage. Importantly, target ablation of proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes the initiation of LUAD. Furthermore, sustained endoplasmic reticulum stress is demonstrated to be a hallmark and a reliable biomarker of AIS progression to IAC. Consistently, target promotion of ER stress profoundly retards LUAD progression. Our study provides comprehensive proteogenomic landscape of SSNs, sheds lights on the tumorigenesis and progression of SSNs and suggests preventive and therapeutic strategies for LUAD.
Collapse
Affiliation(s)
- Hang Su
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Li Chen
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhongyi Cheng
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Jing Li
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Junfang Xu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Yifang Dang
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Mengge Zheng
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Yajuan Cao
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jiani Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Xuefei Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jianxia Chen
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Tao Luo
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Jun Zhu
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Wei Sha
- Department of tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Haipeng Liu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| |
Collapse
|
11
|
Prifti DK, Lauzier A, Garand C, Calvo E, Devillers R, Roy S, Dos Santos A, Descombes L, Trudel B, Laplante M, Bordeleau F, Elowe S. ARHGEF17/TEM4 regulates the cell cycle through control of G1 progression. J Cell Biol 2025; 224:e202311194. [PMID: 39903211 PMCID: PMC11792891 DOI: 10.1083/jcb.202311194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/17/2024] [Accepted: 12/10/2024] [Indexed: 02/06/2025] Open
Abstract
The Ras homolog (Rho) small GTPases coordinate diverse cellular functions including cell morphology, adhesion and motility, cell cycle progression, survival, and apoptosis via their role in regulating the actin cytoskeleton. The upstream regulators for many of these functions are unknown. ARHGEF17 (also known as TEM4) is a Rho family guanine nucleotide exchange factor (GEF) implicated in cell migration, cell-cell junction formation, and the mitotic checkpoint. In this study, we characterize the regulation of the cell cycle by TEM4. We demonstrate that TEM4-depleted cells exhibit multiple defects in mitotic entry and duration, spindle morphology, and spindle orientation. In addition, TEM4 insufficiency leads to excessive cortical actin polymerization and cell rounding defects. Mechanistically, we demonstrate that TEM4-depleted cells delay in G1 as a consequence of decreased expression of the proproliferative transcriptional co-activator YAP. TEM4-depleted cells that progress through to mitosis do so with decreased levels of cyclin B as a result of attenuated expression of CCNB1. Importantly, cyclin B overexpression in TEM4-depleted cells largely rescues mitotic progression and chromosome segregation defects in anaphase. Our study thus illustrates the consequences of Rho signaling imbalance on cell cycle progression and identifies TEM4 as the first GEF governing Rho GTPase-mediated regulation of G1/S.
Collapse
Affiliation(s)
- Diogjena Katerina Prifti
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Annie Lauzier
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Chantal Garand
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Eva Calvo
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Romain Devillers
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
| | - Suparba Roy
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Alexsandro Dos Santos
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Laurence Descombes
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Cancer, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Benjamin Trudel
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Cancer, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval (LOEX), Québec, Canada
| | - Mathieu Laplante
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
| | - François Bordeleau
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Cancer, Québec, Canada
- Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec City, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval (LOEX), Québec, Canada
| | - Sabine Elowe
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Département de Pédiatrie, Faculté de Médicine, Université Laval, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| |
Collapse
|
12
|
Albendea-Gomez T, Mendoza-Tamajon S, Castro-Mecinas R, Escobar B, Ferreira Rocha S, Urra-Balduz S, Nicolas-Avila JA, Oliver E, Villalba-Orero M, Martin-Puig S. Vascular HIF2 Signaling Prevents Cardiomegaly, Alveolar Congestion, and Capillary Remodeling During Chronic Hypoxia. Arterioscler Thromb Vasc Biol 2025; 45:e78-e98. [PMID: 39846162 DOI: 10.1161/atvbaha.124.321780] [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: 08/29/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Hypoxia is associated with the onset of cardiovascular diseases including cardiac hypertrophy and pulmonary hypertension. HIF2 (hypoxia inducible factor 2) signaling in the endothelium mediates pulmonary arterial remodeling and subsequent elevation of the right ventricular systolic pressure during chronic hypoxia. Thus, novel therapeutic opportunities for pulmonary hypertension based on specific HIF2 inhibitors have been proposed. Nevertheless, HIF2 relevance beyond the pulmonary endothelium or in the cardiac adaptation to hypoxia remains elusive. Wt1 (Wilms tumor 1) lineage contributes to the heart and lung vascular compartments, including pericytes, endothelial cells, and smooth muscle cells. METHODS Here, we describe the response to chronic hypoxia of a novel HIF2 mutant mouse model in the Wt1 lineage (Hif2/Wt1 cKO [conditional knockout]), characterizing structural and functional aspects of the heart and lungs by means of classical histology, immunohistochemistry, flow cytometry, echocardiography, and lung ultrasound analysis. RESULTS Hif2/Wt1 cKO is protected against pulmonary remodeling and increased right ventricular systolic pressure induced by hypoxia, but displays alveolar congestion, inflammation, and hemorrhages associated with microvascular instability. Furthermore, lack of HIF2 in the Wt1 lineage leads to cardiomegaly, capillary remodeling, right and left ventricular hypertrophy, systolic dysfunction, and left ventricular dilation, suggesting pulmonary-independent cardiac direct roles of HIF2 in hypoxia. These structural defects are partially restored upon reoxygenation, while cardiac functional parameters remain altered. CONCLUSIONS Our results indicate that cardiopulmonary HIF2 signaling prevents excessive vascular proliferation during chronic hypoxia and define novel protective roles of HIF2 to warrant stable microvasculature and organ function.
Collapse
MESH Headings
- Animals
- Hypoxia/metabolism
- Hypoxia/physiopathology
- Vascular Remodeling
- Signal Transduction
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Mice, Knockout
- Disease Models, Animal
- Chronic Disease
- Cardiomegaly/metabolism
- Cardiomegaly/physiopathology
- Cardiomegaly/pathology
- Cardiomegaly/genetics
- Capillaries/metabolism
- Capillaries/pathology
- Capillaries/physiopathology
- Pulmonary Alveoli/metabolism
- Pulmonary Alveoli/pathology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/prevention & control
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Ventricular Function, Right
- Mice
- Male
- Mice, Inbred C57BL
- Ventricular Remodeling
- Ventricular Function, Left
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Pulmonary Artery/pathology
- Transcription Factors
Collapse
Affiliation(s)
- Teresa Albendea-Gomez
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- School of Medicine, Universidad Francisco de Vitoria, Madrid, Spain (T.A.-G., S.M.-P.)
| | - Susana Mendoza-Tamajon
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
| | - Rosana Castro-Mecinas
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
| | - Beatriz Escobar
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- Mouse Genome Editing Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain (B.E.)
| | - Susana Ferreira Rocha
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
| | - Sonia Urra-Balduz
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
| | - Jose Angel Nicolas-Avila
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- Cardiovascular Research Institute & Department of Microbiology and Immunology, University of California San Francisco (J.A.N.-A.)
| | - Eduardo Oliver
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB), Madrid, Spain (E.O.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (E.O.)
| | - Maria Villalba-Orero
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- Department of Animal Medicine and Surgery, Universidad Complutense de Madrid, Madrid, Spain (M.V.-O.)
| | - Silvia Martin-Puig
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- School of Medicine, Universidad Francisco de Vitoria, Madrid, Spain (T.A.-G., S.M.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Spain (S.M.-P.)
| |
Collapse
|
13
|
Zhang C, Wang K, Wang H. The emerging landscape and future perspective of SCLC transformation: From molecular mechanisms to therapeutic strategies. Crit Rev Oncol Hematol 2025; 207:104616. [PMID: 39805410 DOI: 10.1016/j.critrevonc.2025.104616] [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: 08/27/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Small-cell lung cancer (SCLC) is featured by high malignancy and undesirable prognosis. Transformed SCLC shares several common grounds but differ in biological behavior, molecular mechanism and therapeutic options from typical SCLC. SCLC transformation exerts indispensable role in drug resistance among patients with non-small cell lung cancer (NSCLC) upon various treatment modalities. Two hypotheses have been raised to account for SCLC transformation. It develops mostly in EGFR-mutant adenocarcinoma, and can also occur in ALK or ROS1 mutant patients, and EGFR-wildtype adenocarcinoma. Effective biomarkers for early detection, and therapeutic strategies are vital for improving survival for patients undergoing SCLC transformation. This review summarizes the emerging landscape in transformed SCLC, including its origin, molecular mechanisms, approaches for early detection and corresponding therapeutic options, in a bid to gain a comprehensive insight of this recalcitrant and tricky disease. More importantly, we also discuss challenges that lie ahead and future perspectives on this aggressive malignancy.
Collapse
Affiliation(s)
- Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Kai Wang
- Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
14
|
He H, Ma C, Wei W, Wang H, Lai Y, Liu M, Sun S, Ma Q, Lai J, Liu H, Liu H, Sun F, Lin X. Heparan sulfate regulates myofibroblast heterogeneity and function to mediate niche homeostasis during alveolar morphogenesis. Nat Commun 2025; 16:1834. [PMID: 39979343 PMCID: PMC11842828 DOI: 10.1038/s41467-025-57163-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
Postnatal respiration requires bulk formation of alveoli that produces extensive surface area for gas diffusion from epithelium to the circulatory system. Alveolar morphogenesis initiates at late gestation or postnatal stage during mammalian development and is mediated by coordination among multiple cell types. Here we show that fibroblast-derived Heparan Sulfate Glycosaminoglycan (HS-GAG) is essential for maintaining a niche that supports alveolar formation by modulating both biophysical and biochemical cues. Gli1-CreER mediated deletion of HS synthase gene Ext1 in lung fibroblasts results in enlarged and simplified alveolar structures. Ablation of HS results in loss of a subset of PDGFRαhi αSMA+ alveolar myofibroblasts residing in the distal alveolar region, which exhibit contractile properties and maintain WNT signaling activity to support normal proliferation and differentiation of alveolar epithelial cells. HS is essential for proliferation while preventing precocious apoptosis of alveolar myofibroblasts. We show that these processes are dependent upon FGF/MAPK signaling and forced activation of MAPK/ERK signaling partially corrected alveolar simplification and restored alveolar myofibroblast number and AT2 cell proliferation in HS deficient mice. These data reveal HS-dependent myofibroblast heterogeneity and function as an essential orchestrator for developing alveolar niche critical for the generation of gas exchange units.
Collapse
Affiliation(s)
- Hua He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
| | - Chong Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
| | - Wei Wei
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haonan Wang
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yutian Lai
- Department of Lung Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ming Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Shenfei Sun
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jiashuang Lai
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
| | - Hanxiang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
| | - Hanmin Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
| | - Fei Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
| | - Xinhua Lin
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Nair-Menon J, Kingsley C, Mesnaoui H, Lin P, Wilson K, Rohrer B, Kourtidis A. The subcellular topology of the RNAi machinery is multifaceted and reveals adherens junctions as an epithelial hub. RESEARCH SQUARE 2025:rs.3.rs-5837046. [PMID: 40034449 PMCID: PMC11875308 DOI: 10.21203/rs.3.rs-5837046/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The RNA interference (RNAi) machinery is a key cellular mechanism catalyzing biogenesis and function of miRNAs to post-transcriptionally regulate mRNA expression. The RNAi machinery includes a set of protein complexes with subcellular localization traditionally presented in a uniform fashion: the microprocessor processes miRNAs in the nucleus, whereas the DICER and the RNA-induced silencing complex (RISC) further process and enable activity of miRNAs in the cytoplasm. However, several studies have identified subcellular patterns of RNAi components that deviate from this model. We have particularly shown that RNAi complexes associate with the adherens junctions of well-differentiated epithelial cells, through the E-cadherin partner PLEKHA7. To assess the extent of these subcellular topological patterns, we examined subcellular localization of the microprocessor and RISC in a series of human cell lines and normal human tissues. Our results show that junctional localization of RNAi components is a broad characteristic of well-differentiated epithelia, but it is absent in transformed or mesenchymal cells and tissues. We also find extensive localization of the microprocessor in the cytoplasm, as well as of RISC in the nucleus. These findings expose a RNAi machinery with multifaceted subcellular topology that may inform its physiological role and calls for updating of the current models.
Collapse
Affiliation(s)
| | | | | | - Peter Lin
- Medical University of South Carolina (MUSC)
| | | | | | | |
Collapse
|
16
|
El Yousfi Y, Fernández-Farrán FJ, Oliver FJ, López-Rivas A, Yerbes R. Regulation of ER stress-induced apoptotic and inflammatory responses via YAP/TAZ-mediated control of the TRAIL-R2/DR5 signaling pathway. Cell Death Discov 2025; 11:42. [PMID: 39904986 PMCID: PMC11794427 DOI: 10.1038/s41420-025-02335-w] [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: 10/23/2024] [Revised: 01/10/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
In tumors, cancer cells are frequently exposed to adverse environmental conditions that result in endoplasmic reticulum (ER) stress. Mechanical signals emerging from extracellular matrix (ECM) rigidity and cell shape regulate the activity of transcriptional co-activators Yes-associated protein (YAP) and its paralog Transcriptional Coactivator with PDZ-binding motif (TAZ). However, the role of ECM rigidity and YAP/TAZ in tumor cell fate decisions under ER stress remains relatively unexplored. Our results suggest that the YAP/TAZ system plays an important role in the control of ER stress-induced cell death by mechanical signaling arising from ECM stiffness in tumor cells. Mechanistically, YAP/TAZ regulates apoptosis induced by ER stress in tumor cells by controlling the activation of the TRAIL-R2/DR5-mediated extrinsic apoptotic pathway through a dual mechanism. On the one hand, the YAP/TAZ system prevents intracellular TRAIL-R2/DR5 clustering in tumor cells. On the other hand, it inhibits cFLIP down-regulation in tumor cells experiencing ER stress. In addition, YAP/TAZ controls the expression of pro-inflammatory interleukin-8 (IL-8/CXCL8) in tumor cells undergoing ER stress by a TRAIL-R2/DR5/caspase-8-dependent mechanism. Although other mechanisms may also be involved in controlling cell death and inflammation in tumor cells facing environmental stress, our results support a model in which regulation of the subcellular localization and activity of the YAP/TAZ transcriptional co-activators could contribute to the microenvironmental control of cell fate decisions in tumor cells undergoing ER stress.
Collapse
Affiliation(s)
- Y El Yousfi
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain
| | - F J Fernández-Farrán
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain
| | - F J Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Centro de Investigación Biomédica en Red de Cáncer CIBERONC, Granada, Spain
| | - A López-Rivas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain
| | - R Yerbes
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain.
- Medical Physiology and Biophysics Department, Universidad de Sevilla and Instituto de Biomedicina de Sevilla (IBiS) (Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla), Seville, Spain.
| |
Collapse
|
17
|
Yang S, Wang X, Gao H, Yuan S. Motile cilia: Key developmental and functional roles in reproductive systems. Andrology 2025. [PMID: 39895399 DOI: 10.1111/andr.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Cilia are specialized microtubule-based organelles that extend from the cell surface and are classified into non-motile and motile types. The assembly and function of cilia are regulated by a complex molecular network that enables motile cilia to generate fluid flow across epithelial surfaces through coordinated beating. These motile cilia are found in the respiratory, nervous, and reproductive systems. In males, motile cilia are found in the efferent ducts and facilitate the transport of sperm from the testis to the epididymis. In females, they are mainly found in the oviducts, where they help to transport, nourish and fertilize eggs, and are also present in the endometrial epithelium. MATERIAL-METHODS This review compares the common factors that affect motile cilia in both male and female reproductive tracts, discusses the origin and development of multiciliated cell and cilia within the efferent ducts and oviducts, and enumerates the infertility or related reproductive diseases that may arise due to motile cilia defects. RESULTS-DISCUSSION In males, motile cilia in the efferent ducts create turbulence through their beating, which keeps semen suspended and prevents ductal obstruction. In females, motile cilia are distributed on the epithelia of the oviducts and the endometrium. Specifically, motile cilia in the infundibulum of the oviduct aid in capturing oocytes, while cilia in the isthmus region have been found to bind to sperm heads, facilitating the formation of the sperm reservoir. Several common factors, such as miR-34b/c and miR-449, TAp73, Gemc1, and estrogen, etc., have been shown to play crucial regulatory roles in motile cilia within the efferent ducts and oviducts, thereby further influencing fertility outcomes. CONCLUSIONS Pathogenic mutations that disrupt ciliary function can impair ciliogenesis or alter the structure of sperm flagella, potentially resulting in infertility. Consequently, motile cilia in both the male and female reproductive tracts are crucial for fertility. There are still numerous unresolved mysteries surrounding these cilia that merit further investigation by researchers, as they hold great significance for the clinical diagnosis and treatment of infertility and related reproductive disorders.
Collapse
Affiliation(s)
- Shiyu Yang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoli Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huihui Gao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Laboratory of Animal Center, Huazhong University of Science and Technology, Wuhan, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
| |
Collapse
|
18
|
Yu XH, Guo XN, Li K, Li JW, Wang K, Wang D, Liu BC. The Role of Wnt5a in Inflammatory Diseases. Immunology 2025; 174:203-212. [PMID: 39668514 DOI: 10.1111/imm.13882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/08/2024] [Accepted: 11/22/2024] [Indexed: 12/14/2024] Open
Abstract
Wnt5a plays an important role in cell development and maturation and is closely associated with various diseases, such as malignant tumours, metabolic disorders, fibrosis, growth and development. Recent studies have shown that Wnt5a expression and signal transduction are strongly involved in the inflammatory response. This study comprehensively reviewed the latest research progress on the association between Wnt5a and several inflammatory diseases, such as sepsis, asthma, chronic obstructive pulmonary disease, tuberculosis, rheumatoid arthritis, atherosclerosis and psoriasis vulgare. We elucidated the mechanism by which the Wnt5a protein is involved in the pathogenesis of these diseases, providing a basis for the prevention and treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xin-Hua Yu
- Department of Pediatrics, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Xin-Ning Guo
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kui Li
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Jia-Wei Li
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Kaijin Wang
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bi-Cui Liu
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Batool S, Chokkakula S, Jeong JH, Baek YH, Song MS. SARS-CoV-2 drug resistance and therapeutic approaches. Heliyon 2025; 11:e41980. [PMID: 39897928 PMCID: PMC11786845 DOI: 10.1016/j.heliyon.2025.e41980] [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: 05/02/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
In light of the transition of COVID-19 from a pandemic to an endemic phase, there is still a dire need to address challenges associated with drug resistance, particularly among immunocompromised and high-risk populations. This review explores the current state of research on SARS-CoV-2 drug resistance and underscores the ongoing need for effective therapeutic strategies. It critically evaluates existing knowledge on resistance mechanisms and therapeutic options, aiming to consolidate information and highlight areas for future research. By examining the complex interactions between the virus and its host, the review advocates for a multifaceted approach, including combination therapies, targeted drug development, and continuous surveillance of viral mutations. It also emphasizes the impact of evolving viral variants on antiviral efficacy and suggests adaptive treatment protocols. This review aims to enhance our understanding of SARS-CoV-2 drug resistance and contribute to more effective management of COVID-19 through a discussion of promising strategies such as drug repurposing and combination therapies.
Collapse
Affiliation(s)
- Sania Batool
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Santosh Chokkakula
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Ju Hwan Jeong
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Yun Hee Baek
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Min-Suk Song
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| |
Collapse
|
20
|
Bai J, Asakawa T, Yuan W, Lin Y, Ju H, Xu D, Yang M, Li S, Li G, Guo D, Lu H, Zhang X. Efficacy and safety of SHEN26, a novel oral small molecular RdRp inhibitor for COVID-19 treatment: a multicenter, randomized, double-blinded, placebo-controlled, phase II clinical trial. Virol J 2025; 22:16. [PMID: 39863888 PMCID: PMC11762885 DOI: 10.1186/s12985-025-02631-y] [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: 12/06/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND SHEN26 (ATV014) is an oral RNA-dependent RNA polymerase (RdRp) inhibitor with potential anti-SARS-CoV-2 activity. Safety, tolerability, and pharmacokinetic characteristics were verified in a Phase I study. This phase II study aimed to verify the efficacy and safety of SHEN26 in COVID-19 patients. METHODS This was a multicenter randomized double-blind placebo-controlled study. Mild-to-moderate adult patients with COVID-19 were recruited and randomly assigned to the high-dose (400 mg), low-dose (200 mg), or placebo groups (1:1:1). The primary outcome measure was "changes in RNA levels on Day seven (D7)". The second outcome measures were "changes of RNA levels on D3, D5, D10, D28," "Time of clearance of virus." RESULTS A total of 91 patients were recruited in this study between December 08, 2022, and January 27, 2023. Twelve patients dropped out due to a lack of examination results. Finally, the data of 79 patients (24 in the placebo group, 31 in the 200 mg group, and 24 in the 400 mg group) were analyzed. No significant differences in the baseline data were observed between the groups. The changes of viral load were significantly higher on D3 (P = 0.0119), and D5 (P = 0.0120) in 400 mg group (vs. placebo group), and the difference value achieved 1.06 log10 copies/mL on D3 and 1.21 log10 copies/mL on D5. No significant difference was found in the viral clearance time between SHEN26 administrating groups and placebo groups. Administration of SHEN26 did not enhance drug-related ADEs and did not induce ADEs, and ADE inducing drug withdrawal, dose reduction, or death. Moreover, SHEN26 did not worsen the renal function. CONCLUSIONS Our findings indicate a better efficacy of a high dose (400 mg) for COVID-19 treatment. These preliminary data on the efficacy and safety provide useful information and a working basis for further verification and development of SHEN26 as a novel oral small-molecule antiviral drug for treating COVID-19.
Collapse
Affiliation(s)
- Jiangtao Bai
- Shenzhen Kexing Pharmaceutical Co., Ltd, Shenzhen, Guangdong, 518057, China
| | - Tetsuya Asakawa
- Institute of Neurology, Shenzhen Third People's Hospital, Second Hospital, Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong, 518112, China.
- National Clinical Research Center for Infectious Disease, Shenzhen, 518112, China.
| | - Wenfang Yuan
- Department of Infectious Diseases, Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei, 050021, China
| | - Yuanlong Lin
- Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, 518112, China
- National Clinical Research Center for Infectious Disease, Shenzhen, 518112, China
| | - Hao Ju
- Department of Endocrinology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
| | - Dandan Xu
- Shenzhen Kexing Pharmaceutical Co., Ltd, Shenzhen, Guangdong, 518057, China
| | - Mingming Yang
- Shenzhen Kexing Pharmaceutical Co., Ltd, Shenzhen, Guangdong, 518057, China
| | - Shuo Li
- Medi-X Pingshan, Southern University of Science and Technology, Shenzhen, Guangdong, 518118, China
- Shenzhen AntiV Pharma Co. Ltd., Shenzhen, Guangdong, 518118, China
| | - Guanguan Li
- Shenzhen AntiV Pharma Co. Ltd., Shenzhen, Guangdong, 518118, China
| | - Deyin Guo
- Guangzhou National Laboratory, Guangzhou International BioIsland, Guangzhou, Guangdong, 510320, China.
| | - Hongzhou Lu
- Institute of Neurology, Shenzhen Third People's Hospital, Second Hospital, Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong, 518112, China.
- Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, 518112, China.
- National Clinical Research Center for Infectious Disease, Shenzhen, 518112, China.
| | - Xumu Zhang
- Medi-X Pingshan, Southern University of Science and Technology, Shenzhen, Guangdong, 518118, China.
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
21
|
Jiang H, Xia H, Wang Z, Xiong F. Discovery of Severe Acute Respiratory Syndrome Coronavirus 2 Main Protease Inhibitors through Rational Design of Novel Fluorinated 1,3,4-oxadiazole Amide Derivatives: An In-Silico Study. Chem Biodivers 2025:e202403179. [PMID: 39853882 DOI: 10.1002/cbdv.202403179] [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: 12/01/2024] [Revised: 12/23/2024] [Accepted: 01/24/2025] [Indexed: 01/26/2025]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) variants continue to emerge, there is an urgent need to develop more effective antiviral drugs capable of combating the COVID-19 pandemic. The main protease (Mpro) of SARS-CoV-2 is an evolutionarily conserved drug discovery target. The present study mainly focused on chemoinformatics computational methods to investigate the efficacy of our newly designed trifluoromethyl-1,3,4-oxadiazole amide derivatives as SARS-CoV-2 Mpro inhibitors. Drug-likeness absorption, distribution, metabolism, excretion, and toxicity analysis, molecular docking simulation, density functional theory (DFT), and molecular dynamics simulation methods were included. A comprehensive drug-likeness analysis was performed on the 14 newly designed compounds (1a-1n), and this series of small molecule inhibitors showed potential anti-SARS-CoV-2 activity. In order to reveal the mechanism of drug interaction, these novel compounds were classified by structure, and molecular docking simulations were performed. The results showed good interactions and identified the key amino acid residue GLY-143. Further DFT analysis using B3LYP-D3BJ functional and 6-311 + + G (d, p) basis set was performed to optimize the optimal configuration of the Mpro inhibitors, and the infrared spectrum of the vibration frequency was analyzed to clearly understand the structure and stability of the drug. The electrostatic potential map was analyzed to predict the reactivity of functional groups and protein-substrate interactions. The frontier molecular orbital analysis and density of states map showed the reactivity level and stability of the drug itself, among which 1i had the smallest energy gap difference (ΔEgap = 3.64 ev), showing good reactivity. The analysis of global reactivity descriptors such as electrophilic index (ω) and chemical potential (μ) also showed that our newly designed Mpro inhibitors had stronger interactions. Molecular dynamics simulation further revealed the stable binding of the Mpro inhibitors in a solvent environment. The binding free energy results calculated by Molecular Mechanics / Poisson Boltzmann Surface Area (MM/PBSA) all exceeded the Food and Drug Administration-approved standard reference drug (Nirmatrelvir), and the free energy landscape and principal component analysis also further described the energy sites formed during the binding process between the drug molecule and the ligand-protein and the changes in conformation. These new series of small molecule inhibitors studied in this work will provide the necessary theoretical basis for the synthesis and activity evaluation of novel SARS-CoV-2 Mpro inhibitors.
Collapse
Affiliation(s)
- Huiying Jiang
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, P. R. China
| | - Heping Xia
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, P. R. China
| | - Zhonghua Wang
- Shanghai Engineering Research Center of Green Fluoropharmaceutical Technology, School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, P. R. China
| | - Fei Xiong
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, P. R. China
| |
Collapse
|
22
|
Yabut OR, Arela J, Gomez HG, Castillo JG, Ngo T, Pleasure SJ. Aberrant FGF signaling promotes granule neuron precursor expansion in SHH subgroup infantile medulloblastoma. eLife 2025; 13:RP100767. [PMID: 39835775 PMCID: PMC11750132 DOI: 10.7554/elife.100767] [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] [Indexed: 01/30/2025] Open
Abstract
Mutations in Sonic Hedgehog (SHH) signaling pathway genes, for example, Suppressor of Fused (SUFU), drive granule neuron precursors (GNP) to form medulloblastomas (MBSHH). However, how different molecular lesions in the Shh pathway drive transformation is frequently unclear, and SUFU mutations in the cerebellum seem distinct. In this study, we show that fibroblast growth factor 5 (FGF5) signaling is integral for many infantile MBSHH cases and that FGF5 expression is uniquely upregulated in infantile MBSHH tumors. Similarly, mice lacking SUFU (Sufu-cKO) ectopically express Fgf5 specifically along the secondary fissure where GNPs harbor preneoplastic lesions and show that FGFR signaling is also ectopically activated in this region. Treatment with an FGFR antagonist rescues the severe GNP hyperplasia and restores cerebellar architecture. Thus, direct inhibition of FGF signaling may be a promising and novel therapeutic candidate for infantile MBSHH.
Collapse
Affiliation(s)
- Odessa R Yabut
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Jessica Arela
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Hector G Gomez
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Jesse Garcia Castillo
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Thomas Ngo
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Samuel J Pleasure
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| |
Collapse
|
23
|
Daniele T, Cury J, Morin MC, Ahier A, Isaia D, Jarriault S. Essential and dual effects of Notch activity on a natural transdifferentiation event. Nat Commun 2025; 16:75. [PMID: 39746948 PMCID: PMC11697417 DOI: 10.1038/s41467-024-55286-8] [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] [Received: 04/02/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Cell identity can be reprogrammed, naturally or experimentally, albeit with low frequency. Why some cells, but not their neighbours, undergo a cell identity conversion remains unclear. We find that Notch signalling plays a key role to promote natural transdifferentiation in C. elegans hermaphrodites. Endogenous Notch signalling endows a cell with the competence to transdifferentiate by promoting plasticity factors expression (hlh-16/Olig and sem-4/Sall). Strikingly, ectopic Notch can trigger additional transdifferentiation in vivo. However, Notch signalling can both promote and block transdifferentiation depending on its activation timing. Notch only promotes transdifferentiation during an early precise window of opportunity and signal duration must be tightly controlled in time. Our findings emphasise the importance of temporality and dynamics of the underlying molecular events preceding the initiation of natural cell reprogramming. Finally, our results support a model where both an extrinsic signal and the intrinsic cellular context combine to empower a cell with the competence to transdifferentiate.
Collapse
Affiliation(s)
- Thomas Daniele
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
- Vertex Pharmaceuticals (CH) GmbH, Zug, Switzerland
| | - Jeanne Cury
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
| | - Marie-Charlotte Morin
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
| | - Arnaud Ahier
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Davide Isaia
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France
- Skyhawk Therapeutics, Basel, Switzerland
| | - Sophie Jarriault
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM U1298, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
24
|
Chen PJ, Wang K, Lin MH, Sharma R, Kalajzic Z, O'Brien M, Yadav S. Alendronate partially rescues the periodontal defects in OIM mouse model of osteogenesis imperfecta. Sci Rep 2025; 15:88. [PMID: 39747677 PMCID: PMC11695738 DOI: 10.1038/s41598-024-84756-8] [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] [Received: 07/19/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025] Open
Abstract
Osteogenesis imperfecta (OI) is a fairly common generalized connective disorder characterized by low bone mass, bone deformities and impaired bone quality that predisposes affected individuals to musculoskeletal fragility. Periodontal ligament (PDL)-alveolar bone and PDL-cementum entheses' roles under OI conditions during physiological loading and orthodontic forces remain largely unknown. In addition, bisphosphonates (e.g., alendronate) are commonly used therapeutics for the treatment of OI. Our knowledge, in terms of the affects of alendronate treatment on the PDL entheses in OI is also far from complete. In this study, we identified craniofacial skeletal defects in an osteogenesis imperfecta (oim) murine model of OI. Relative to wild-type littermates, oim mice were found to have decreased skull length, cranial height/width/length, nose length, nasal length, and frontal length. Next, we discovered that oim mice exhibited defects in several dental structures, including short roots and decreased volumes of the alveolar bone, dentin, and cellular cementum. Further, we specifically investigated periodontal defects in the oim mice. Alveolar bone loss in oim mice was primarily associated with elevated bone resorption due to an increased osteoclast number, along with reduced bone formation related to increased sclerostin (SOST) expression. PDL fibers in oim mice were disrupted and discontinuous, while Sharpey's fibers at the PDL-bone entheses were reduced. Mechanism-based studies showed that catabolism of the PDL was elevated in oim mice, as revealed by an increase in MMP13 and CTSK expression. Meanwhile, the quality of the collagen fibers were impaired in oim mice due to a large accumulation of uncleaved collagen I fibers. With alendronate treatment, however, we could partially rescue these phenotypes. This study, for the first time, characterized periodontal defects in oim mice, detailed craniofacial defects and demonstrated the effectiveness of alendronate in partially restoring these defects.
Collapse
Affiliation(s)
- Po-Jung Chen
- Department of Growth and Development, University of Nebraska Medical Center, 4000 East Campus Loop South, 68583-0740, Lincoln, NE, US.
| | - Ke Wang
- Division of Orthodontics, University of Connecticut Health Center, Farmington, US
| | - Meng-Hsuan Lin
- Department of Adult Restorative Dentistry, University of Nebraska Medical Center, Lincoln, US
| | | | - Zana Kalajzic
- University of Connecticut Health Center, Farmington, US
| | - Mara O'Brien
- University of Connecticut Health Center, Farmington, US
| | - Sumit Yadav
- Department of Growth and Development, University of Nebraska Medical Center, 4000 East Campus Loop South, 68583-0740, Lincoln, NE, US
| |
Collapse
|
25
|
Bondeelle L, Clément S, Bergeron A, Tapparel C. Lung stem cells and respiratory epithelial chimerism in transplantation. Eur Respir Rev 2025; 34:240146. [PMID: 39971397 PMCID: PMC11836672 DOI: 10.1183/16000617.0146-2024] [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: 06/25/2024] [Accepted: 11/21/2024] [Indexed: 02/21/2025] Open
Abstract
Stem cells are capable of self-renewal and differentiation into specialised types. They range from totipotent cells to multipotent or somatic stem cells and ultimately to unipotent cells. Some adult multipotent stem cells can have the potential to regenerate and colonise diverse tissues. The respiratory airways and lung mucosa, exposed to ambient air, perform vital roles for all human tissues and organs. They serve as barriers against airborne threats and are essential for tissue oxygenation. Despite low steady-state turnover, lungs are vulnerable to injuries and diseases from environmental exposure. Lung stem cells are crucial due to their regenerative potential and ability to replace damaged cells. Lung repair with extrapulmonary stem cells can occur, leading to the coexistence of respiratory cells with different genetic origins, a phenomenon known as airway epithelial chimerism. The impact of such chimerism in lung repair and disease is actively studied. This review explores different stem cell types, focusing on pulmonary stem cells. It discusses airway epithelium models derived from stem cells for studying lung diseases and examines lung chimerism, particularly in lung transplantation and haematopoietic stem cell transplantation, highlighting its significance in understanding tissue repair and chimerism-mediated repair processes in lung pathology.
Collapse
Affiliation(s)
- Louise Bondeelle
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Sophie Clément
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Anne Bergeron
- Pneumology Department, Geneva University Hospitals, Geneva, Switzerland
- Co-last author
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
- Co-last author
| |
Collapse
|
26
|
Yoffe L, Bhinder B, Kang SW, Zhang H, Singh A, Ravichandran H, Markowitz G, Martin M, Kim J, Zhang C, Elemento O, Tansey W, Bates S, McGraw TE, Borczuk A, Lee HS, Altorki NK, Mittal V. Acquisition of discrete immune suppressive barriers contributes to the initiation and progression of preinvasive to invasive human lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630523. [PMID: 39803458 PMCID: PMC11722343 DOI: 10.1101/2024.12.31.630523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Computerized chest tomography (CT)-guided screening in populations at risk for lung cancer has increased the detection of preinvasive subsolid nodules, which progress to solid invasive adenocarcinoma. Despite the clinical significance, there is a lack of effective therapies for intercepting the progression of preinvasive to invasive adenocarcinoma. To uncover determinants of early disease emergence and progression, we used integrated single-cell approaches, including scRNA-seq, multiplexed imaging mass cytometry and spatial transcriptomics, to construct the first high-resolution map of the composition, lineage/functional states, developmental trajectories and multicellular crosstalk networks from microdissected non-solid (preinvasive) and solid compartments (invasive) of individual part-solid nodules. We found that early disease initiation and subsequent progression are associated with the evolution of immune-suppressive cellular phenotypes characterized by decreased cytotoxic CD8 T and NK cells, increased T cell exhaustion and accumulation of immunosuppressive regulatory T cells (Tregs) and M2-like macrophages expressing TREM2. Within Tregs, we identified a unique population of 4-1BB+ Treg subset enriched for the IL2-STAT5 suppressive pathway with transcription profiles supporting discrete metabolic alterations. Spatial analysis showed increased density of suppressive immune cells around tumor cells, increased exhaustion phenotype of both CD4 and CD8 T cells expressing chemokine CXCL13, and spatial microcomplex of endothelial and lymphocyte interactions within tertiary lymphoid structures. The single-cell architecture identifies determinants of early disease emergence and progression, which may be developed not only as diagnostic/prognostic biomarkers but also as targets for disease interception. Additionally, our dataset constitutes a valuable resource for the preinvasive lung cancer research community.
Collapse
Affiliation(s)
- Liron Yoffe
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Bhavneet Bhinder
- Department of Physiology and Biophysics, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Sung Wook Kang
- David Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Haoran Zhang
- Department of Computer Science, University of Texas at Austin, TX 78712, USA
| | - Arshdeep Singh
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Hiranmayi Ravichandran
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Geoffrey Markowitz
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Mitchell Martin
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Junbum Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Chen Zhang
- Department of Pathology, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Wesley Tansey
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stewart Bates
- Interventional Oncology, Johnson and Johnson, 50-100 Holmers Farm Way, High Wycombe, UK, HP12 4DP
| | - Timothy E. McGraw
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Department of Biochemistry, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Alain Borczuk
- Department of Pathology, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Hyun-Sung Lee
- David Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Nasser K. Altorki
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| | - Vivek Mittal
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 525 East 68th Street, New York, New York 10065, USA
| |
Collapse
|
27
|
Chen X, Chen R, Wu Y, Yu A, Wang F, Ying C, Yin Y, Chen X, Ma L, Fu Y. FABP5+ macrophages contribute to lipid metabolism dysregulation in type A aortic dissection. Int Immunopharmacol 2024; 143:113438. [PMID: 39447410 DOI: 10.1016/j.intimp.2024.113438] [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: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Type A aortic dissection (TAAD) is an acute onset disease with a high mortality rate. TAAD is caused by a tear in the aortic intima and subsequent blood infiltration. The most prominent characteristics of TAAD are aortic media degeneration and inflammatory cell infiltration, which disturb the structural integrity and function of nonimmune and immune cells in the aortic wall. However, to date, there is no systematic evaluation of the interactions between nonimmune cells and immune cells and their effects on metabolism in the context of aortic dissection. Here, multiomics, including bulk RNA-seq, single-cell RNA-seq, and lipid metabolomics, was applied to elucidate the comprehensive TAAD lipid metabolism landscape. Normally, monocytes in the stress response state secrete a variety of cytokines. Injured fibroblasts lack the ability to degrade lipids, which is suspected to contribute to a high lipid environment. Macrophages differentiate into fatty acid binding protein 5-positive (FABP5+) macrophages under the stimulation of metabolic substrates. Moreover, the upregulation of Fabp5+ macrophages were retrospectively validated in TAAD model mice and TAAD patients. Finally, Fabp5+ macrophages can generate a wide range of proinflammatory cytokines, which possibly contribute to TAAD pathogenesis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Ruoshi Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yuefeng Wu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; The Lab of Biomed-X, Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining 310000, China
| | - Anfeng Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Fei Wang
- GeneChem Technology Co. Ltd., Shanghai 201203, China
| | - Chenxi Ying
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yifei Yin
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Xiaofan Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Liang Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.
| | - Yufei Fu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou 310006, China.
| |
Collapse
|
28
|
Nakagaki R, Mukaibo T, Monir A, Gao X, Munemasa T, Nodai T, Tamura A, Obikane YH, Kondo Y, Masaki C, Hosokawa R. Simulated microgravity environment inhibits matrix mineralization during the osteoblast to osteocyte differentiation. Biochem Biophys Res Commun 2024; 739:150963. [PMID: 39550861 DOI: 10.1016/j.bbrc.2024.150963] [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: 08/21/2024] [Revised: 10/16/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
This study investigates the effects of microgravity on the differentiation and mineralization of IDG-SW3 osteocyte-like cells to understand the response of bone cells to microgravity and develop strategies to mitigate bone loss in astronauts. IDG-SW3 cells were cultured in collagen-coated dishes and subjected to a 3D clinostat to simulate microgravity 14 days after initiating differentiation. The static group remained under normal gravity. Cells were analyzed on days 14, 18, 22, and 26. Alizarin red staining demonstrated a substantial and time-dependent increase in mineralization in the static group, whereas the microgravity group exhibited little detectable mineralization throughout the experimental period. Quantitative RT-PCR revealed significant upregulation of Rankl, Alpl, Dmp1, and Fgf23 and downregulation of Sost and Phex in the microgravity group. RNA sequencing on day 26 showed distinct gene expression profiles between conditions. Heatmaps highlighted upregulated genes (Ptgs2, Alpl, Comp, Atf4, Lox) and downregulated genes (Rspo2, Ank, Ptn, Mmp13, Aspn, Spp1) under microgravity. Gene ontology (GO) enrichment analysis indicated that upregulated genes were associated with cytoskeletal organization and receptor activities, while downregulated genes were linked to extracellular matrix components and immune response. These findings provide insights into the molecular mechanisms of bone loss in space and emphasize the importance of gravity in bone remodeling. Future studies should validate these genes' functions in osteocyte biology under microgravity.
Collapse
Affiliation(s)
- Ryutaro Nakagaki
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Taro Mukaibo
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan.
| | - Ahmed Monir
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan; Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Zagazig University, Sharkia, Egypt
| | - Xin Gao
- Lister Hill National Center for Biomedical Communication, National Library of Medicine, NIH, Bethesda, MD, USA
| | - Takashi Munemasa
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Tomotaka Nodai
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Akiko Tamura
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Yui Hirata Obikane
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Yusuke Kondo
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Chihiro Masaki
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Ryuji Hosokawa
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
29
|
Li Y, Luo Y, Huang D, Peng L. Sclerostin as a new target of diabetes-induced osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1491066. [PMID: 39720253 PMCID: PMC11666367 DOI: 10.3389/fendo.2024.1491066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Sclerostin, a protein synthesized by bone cells, is a product of the SOST gene. Sclerostin is a potent soluble inhibitor of the WNT signaling pathway, and is known to inhibit bone formation by inhibiting osteocyte differentiation and function. Currently, sclerostin has been the subject of numerous animal experiments and clinical investigations. By conducting a literature review, we have gained insights into the most recent advancements in research. Patients with both type 1 diabetes and type 2 diabetes have high levels of serum sclerostin. Patients with type 1 diabetes and type 2 diabetes are both more likely to suffer from osteoporosis, and serum sclerostin levels are elevated in osteoporosis. Many studies have confirmed that sclerostin has been implicated in the pathogenesis of osteoporosis, so we speculate that sclerostin plays an important role in osteoporosis through the glucose metabolism pathway, which may promote the osteoporosis of morbidity in type 1 diabetes and type 2 diabetes. Based on this, we propose whether serum sclerostin can predict type 1 diabetes and type 2 diabetes-induced osteoporosis, and whether it can be a new target for the prevention and treatment of type 1 diabetes and type 2 diabetes-induced osteoporosis, providing new ideas for clinicians and researchers.
Collapse
Affiliation(s)
- Yanhua Li
- Department of Endocrinology and Metabolism, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Yaheng Luo
- Department of Endocrinology and Metabolism, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Debin Huang
- Department of Endocrinology and Metabolism, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Lele Peng
- Department of Endocrinology and Metabolism, Want Want Hospital, Changsha, Hunan, China
| |
Collapse
|
30
|
Zhao D, Tu C, Zhang L, Guda T, Gu S, Jiang JX. Activation of connexin hemichannels enhances mechanosensitivity and anabolism in disused and aged bone. JCI Insight 2024; 9:e177557. [PMID: 39641271 PMCID: PMC11623949 DOI: 10.1172/jci.insight.177557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 10/11/2024] [Indexed: 12/07/2024] Open
Abstract
Mechanical loading, essential for bone health, promotes bone formation and remodeling. However, the positive response diminishes in cases of disuse and aging, leading to bone loss and an increased fracture risk. This study demonstrates that activating hemichannels (HCs) using a connexin 43 (Cx43) antibody, Cx43(M2), in bone osteocytes revitalizes aging and disused bones. Using a hindlimb suspension (HLS) disuse model and a tibial mechanical loading model, we found that Cx43(M2) inhibited bone loss and osteocyte apoptosis induced by unloading in 16-week-old adult mice. Additionally, it enhanced bone mass in response to tibial loading in 22-month-old aged mice. The HC opening released bone anabolic factor prostaglandin E2 (PGE2) and suppressed catabolic factor sclerostin (SOST). This suppressed the increase of cortical bone formation and reduction of bone resorption during unloading and promoted trabecular and cortical bone formation during loading. Cx43(M2)-induced HC opening, coupled with PGE2 release, effectively rescued unloading-induced bone loss and restored the diminished anabolic response of aged bones to mechanical loading. Activating HCs with the Cx43 antibody holds promise as a de novo therapeutic approach, as it can overcome the limitations of existing treatment regimens for treating bone loss and osteoporosis associated with aging and disuse.
Collapse
Affiliation(s)
- Dezhi Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
- School of Medicine, Northwest University, Xi’an, China
| | - Chao Tu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lidan Zhang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
| | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
| |
Collapse
|
31
|
Masciale V, Banchelli F, Grisendi G, Samarelli AV, Raineri G, Rossi T, Zanoni M, Cortesi M, Bandini S, Ulivi P, Martinelli G, Stella F, Dominici M, Aramini B. The molecular features of lung cancer stem cells in dedifferentiation process-driven epigenetic alterations. J Biol Chem 2024; 300:107994. [PMID: 39547513 PMCID: PMC11714729 DOI: 10.1016/j.jbc.2024.107994] [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: 03/23/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Cancer stem cells (CSCs) may be dedifferentiated somatic cells following oncogenic processes, representing a subpopulation of cells able to promote tumor growth with their capacities for proliferation and self-renewal, inducing lineage heterogeneity, which may be a main cause of resistance to therapies. It has been shown that the "less differentiated process" may have an impact on tumor plasticity, particularly when non-CSCs may dedifferentiate and become CSC-like. Bidirectional interconversion between CSCs and non-CSCs has been reported in other solid tumors, where the inflammatory stroma promotes cell reprogramming by enhancing Wnt signaling through nuclear factor kappa B activation in association with intracellular signaling, which may induce cells' pluripotency, the oncogenic transformation can be considered another important aspect in the acquisition of "new" development programs with oncogenic features. During cell reprogramming, mutations represent an initial step toward dedifferentiation, in which tumor cells switch from a partially or terminally differentiated stage to a less differentiated stage that is mainly manifested by re-entry into the cell cycle, acquisition of a stem cell-like phenotype, and expression of stem cell markers. This phenomenon typically shows up as a change in the form, function, and pattern of gene and protein expression, and more specifically, in CSCs. This review would highlight the main epigenetic alterations, major signaling pathways and driver mutations in which CSCs, in tumors and specifically, in lung cancer, could be involved, acting as key elements in the differentiation/dedifferentiation process. This would highlight the main molecular mechanisms which need to be considered for more tailored therapies.
Collapse
Affiliation(s)
- Valentina Masciale
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Federico Banchelli
- Department of Statistical Sciences "Paolo Fortunati", Alma Mater Studiorum- University of Bologna, Bologna, Italy
| | - Giulia Grisendi
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Anna Valeria Samarelli
- Laboratory of and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Giulia Raineri
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michele Zanoni
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michela Cortesi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Martinelli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Franco Stella
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy; Division of Oncology, University Hospital of Modena and Reggio Emilia, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy.
| |
Collapse
|
32
|
Cooper AH, Barry AM, Chrysostomidou P, Lolignier R, Wang J, Redondo Canales M, Titterton HF, Bennett DL, Weir GA. Peripheral nerve injury results in a biased loss of sensory neuron subpopulations. Pain 2024; 165:2863-2876. [PMID: 39158319 PMCID: PMC11562755 DOI: 10.1097/j.pain.0000000000003321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/11/2024] [Accepted: 05/25/2024] [Indexed: 08/20/2024]
Abstract
ABSTRACT There is a rich literature describing the loss of dorsal root ganglion (DRG) neurons following peripheral axotomy, but the vulnerability of discrete subpopulations has not yet been characterised. Furthermore, the extent or even presence of neuron loss following injury has recently been challenged. In this study, we have used a range of transgenic recombinase driver mouse lines to genetically label molecularly defined subpopulations of DRG neurons and track their survival following traumatic nerve injury. We find that spared nerve injury leads to a marked loss of cells containing DRG volume and a concomitant loss of small-diameter DRG neurons. Neuron loss occurs unequally across subpopulations and is particularly prevalent in nonpeptidergic nociceptors, marked by expression of Mrgprd. We show that this subpopulation is almost entirely lost following spared nerve injury and severely depleted (by roughly 50%) following sciatic nerve crush. Finally, we used an in vitro model of DRG neuron survival to demonstrate that nonpeptidergic nociceptor loss is likely dependent on the absence of neurotrophic support. Together, these results profile the extent to which DRG neuron subpopulations can survive axotomy, with implications for our understanding of nerve injury-induced plasticity and pain.
Collapse
Affiliation(s)
- Andrew H. Cooper
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | - Allison M. Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | - Romane Lolignier
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | - Jinyi Wang
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | | | - Heather F. Titterton
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Greg A. Weir
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
33
|
Xu Y, Li M, Bai L. Pulmonary Epithelium Cell Fate Determination: Chronic Obstructive Pulmonary Disease, Lung Cancer, or Both. Am J Respir Cell Mol Biol 2024; 71:632-645. [PMID: 39078237 DOI: 10.1165/rcmb.2023-0448tr] [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: 12/18/2023] [Accepted: 07/30/2024] [Indexed: 07/31/2024] Open
Abstract
The concurrence of chronic obstructive pulmonary disease (COPD) and lung cancer has been widely reported and extensively addressed by pulmonologists and oncologists. However, most studies have focused on shared risk factors, DNA damage pathways, immune microenvironments, inflammation, and imbalanced proteases/antiproteases. In the present review, we explore the association between COPD and lung cancer in terms of airway pluripotent cell fate determination and discuss the various cell types and signaling pathways involved in the maintenance of lung epithelium homeostasis and their involvement in the pathogenesis of co-occurring COPD and lung cancer.
Collapse
Affiliation(s)
- Yu Xu
- Department of Clinical Oncology, Army Medical Center, and
| | - Mengxia Li
- Department of Clinical Oncology, Army Medical Center, and
| | - Li Bai
- Department of Respiratory and Critical Medicine, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
34
|
Fitzsimons LA, Staurengo-Ferrari L, Khomula EV, Bogen O, Araldi D, Bonet IJM, Green PG, Jordan EE, Sclafani F, Nowak CE, Moulton JK, Ganter GK, Levine JD, Tucker KL. The Nociceptor Primary Cilium Contributes to Mechanical Nociceptive Threshold and Inflammatory and Neuropathic Pain. J Neurosci 2024; 44:e1265242024. [PMID: 39349056 PMCID: PMC11580782 DOI: 10.1523/jneurosci.1265-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/16/2024] [Accepted: 09/20/2024] [Indexed: 10/02/2024] Open
Abstract
The primary cilium, a single microtubule-based organelle protruding from the cell surface and critical for neural development, also functions in adult neurons. While some dorsal root ganglion neurons elaborate a primary cilium, whether it is expressed by and functional in nociceptors is unknown. Recent studies have shown the role of Hedgehog, whose canonical signaling is primary cilium dependent, in nociceptor sensitization. We establish the presence of primary cilia in soma of rat nociceptors, where they contribute to mechanical threshold, prostaglandin E2 (PGE2)-induced hyperalgesia, and chemotherapy-induced neuropathic pain (CIPN). Intrathecal administration of siRNA targeting Ift88, a primary cilium-specific intraflagellar transport (IFT) protein required for ciliary integrity, resulted in attenuation of Ift88 mRNA and nociceptor primary cilia. Attenuation of primary cilia was associated with an increase in mechanical nociceptive threshold in vivo and decrease in nociceptor excitability in vitro, abrogation of hyperalgesia, and nociceptor sensitization induced by both a prototypical pronociceptive inflammatory mediator PGE2 and paclitaxel CIPN, in a sex-specific fashion. siRNA targeting Ift52, another IFT protein, and knockdown of NompB, the Drosophila Ift88 ortholog, also abrogated CIPN and reduced baseline mechanosensitivity, respectively, providing independent confirmation for primary cilia control of nociceptor function. Hedgehog-induced hyperalgesia is attenuated by Ift88 siRNA, supporting the role for primary cilia in Hedgehog-induced hyperalgesia. Attenuation of CIPN by cyclopamine (intradermal and intraganglion), which inhibits Hedgehog signaling, supports the role of Hedgehog in CIPN. Our findings support the role of the nociceptor primary cilium in control of mechanical nociceptive threshold and inflammatory and neuropathic pain, the latter Hedgehog-dependent.
Collapse
Affiliation(s)
- Lindsey A Fitzsimons
- Deparment of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
| | - Larissa Staurengo-Ferrari
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Eugen V Khomula
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Oliver Bogen
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Dionéia Araldi
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Ivan J M Bonet
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Paul G Green
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
- Department of Preventative and Restorative Dental Sciences, University of California San Francisco, San Francisco 94115
| | - Ethan E Jordan
- Deparment of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
| | - Finn Sclafani
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
- School of Biological Sciences, College of Arts and Sciences, University of New England, Biddeford, Maine 04005
| | - Connor E Nowak
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
- School of Biological Sciences, College of Arts and Sciences, University of New England, Biddeford, Maine 04005
| | - Julie K Moulton
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
- School of Biological Sciences, College of Arts and Sciences, University of New England, Biddeford, Maine 04005
| | - Geoffrey K Ganter
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
- School of Biological Sciences, College of Arts and Sciences, University of New England, Biddeford, Maine 04005
| | - Jon D Levine
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
- Department of Medicine, Division of Neuroscience, University of California San Francisco, San Francisco 94115
| | - Kerry L Tucker
- Deparment of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
| |
Collapse
|
35
|
Shen S, Wang P, Wu P, Huang P, Chi T, Xu W, Xi Y. CasRx-based Wnt activation promotes alveolar regeneration while ameliorating pulmonary fibrosis in a mouse model of lung injury. Mol Ther 2024; 32:3974-3989. [PMID: 39245939 PMCID: PMC11573616 DOI: 10.1016/j.ymthe.2024.09.008] [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: 03/06/2024] [Revised: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024] Open
Abstract
Wnt/β-catenin signaling is an attractive target for regenerative medicine. A powerful driver of stem cell activity and hence tissue regeneration, Wnt signaling can promote fibroblast proliferation and activation, leading to fibrosis, while prolonged Wnt signaling is potentially carcinogenic. Thus, to harness its therapeutic potential, the activation of Wnt signaling must be transient, reversible, and tissue specific. In the lung, Wnt signaling is essential for alveolar stem cell activity and alveolar regeneration, which is impaired in lung fibrosis. Activation of Wnt/β-catenin signaling in lung epithelium may have anti-fibrotic effects. Here, we used intratracheal adeno-associated virus 6 injection to selectively deliver CasRx into the lung epithelium, where it reversibly activates Wnt signaling by simultaneously degrading mRNAs encoding Axin1 and Axin2, negative regulators of Wnt/β-catenin signaling. Interestingly, CasRx-mediated Wnt activation specifically in lung epithelium not only promotes alveolar type II cell proliferation and alveolar regeneration but also inhibits lung fibrosis resulted from bleomycin-induced injury, relevant in both preventive and therapeutic settings. Our study offers an attractive strategy for treating pulmonary fibrosis, with general implications for regenerative medicine.
Collapse
Affiliation(s)
- Shengxi Shen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Ping Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Pei Wu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Pengyu Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Engineering Research Center of Pulmonary and Critical Care Medicine Technology and Device (Ministry of Education), Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Tian Chi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Department of Immunobiology, Yale University Medical School, New Haven, CT 06520, USA
| | - Wenqing Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ying Xi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
36
|
Choi J, Chudziak J, Lee JH. Bi-directional regulation between inflammation and stem cells in the respiratory tract. J Cell Sci 2024; 137:jcs263413. [PMID: 39508347 PMCID: PMC11574357 DOI: 10.1242/jcs.263413] [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] [Indexed: 11/15/2024] Open
Abstract
Inflammation plays a crucial role in tissue injury, repair and disease, orchestrating a complex interplay of immune responses and cellular processes. Recent studies have uncovered the intricate connection between inflammation and stem cell dynamics, shedding light on the central role of stem cells in tissue regeneration. This Review highlights the significance of inflammation in shaping epithelial stem cell dynamics and its implications for tissue repair, regeneration and aging. We explore the multifaceted interactions between inflammation and stem cells, focusing on how inflammatory signals affect stem cell behavior and fate as well as the remodeling of their niche in the respiratory tract. We also discuss the concept of 'inflammatory memory' in epithelial stem cells, where prior inflammatory stimuli endow these cells with enhanced regenerative potential and confer long-lasting protective mechanisms for maintaining tissue integrity and function. Furthermore, we review the impact of cell senescence induced by inflammation on tissue regeneration and aging, delving into the molecular mechanisms underlying the modulation of signaling pathways, epigenetic modifications and cellular crosstalk. Understanding these dynamic processes not only deepens our knowledge of tissue homeostasis and repair but also holds profound implications for regenerative medicine strategies aimed at preventing pulmonary diseases.
Collapse
Affiliation(s)
- Jinwook Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jakub Chudziak
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB1 0AW, UK
| | - Joo-Hyeon Lee
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB1 0AW, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EL, UK
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York NY 10065, USA
| |
Collapse
|
37
|
Iwaya C, Suzuki A, Iwata J. Loss of Sc5d results in micrognathia due to a failure in osteoblast differentiation. J Adv Res 2024; 65:153-165. [PMID: 38086515 PMCID: PMC11519736 DOI: 10.1016/j.jare.2023.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/30/2023] [Accepted: 12/09/2023] [Indexed: 01/01/2024] Open
Abstract
INTRODUCTION Mutations in genes related to cholesterol metabolism, or maternal diet and health status, affect craniofacial bone formation. However, the precise role of intracellular cholesterol metabolism in craniofacial bone development remains unclear. OBJECTIVE The aim of this study is to determine how cholesterol metabolism aberrations affect craniofacial bone development. METHODS Mice with a deficiency in Sc5d, which encodes an enzyme involved in cholesterol synthesis, were analyzed with histology, micro computed tomography (microCT), and cellular and molecular biological methods. RESULTS Sc5d null mice exhibited mandible hypoplasia resulting from defects in osteoblast differentiation. The activation of the hedgehog and WNT/β-catenin signaling pathways, which induce expression of osteogenic genes Col1a1 and Spp1, was compromised in the mandible of Sc5d null mice due to a failure in the formation of the primary cilium, a cell surface structure that senses extracellular cues. Treatments with an inducer of hedgehog or WNT/β-catenin signaling or with simvastatin, a drug that restores abnormal cholesterol production, partially rescued the defects in osteoblast differentiation seen in Sc5d mutant cells. CONCLUSION Our results indicate that loss of Sc5d results in mandibular hypoplasia through defective primary cilia-mediated hedgehog and WNT/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Chihiro Iwaya
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
| | - Akiko Suzuki
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
| | - Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA; MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
38
|
Lo CH, Chen S, Zhao J, Liu Z, Wang B, Wang Q, Kowal TJ, Sun Y. Defective Neurogenesis in Lowe Syndrome is Caused by Mitochondria Loss and Cilia-related Sonic Hedgehog Defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621496. [PMID: 39553960 PMCID: PMC11565974 DOI: 10.1101/2024.11.01.621496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Human brain development is a complex process that requires intricate coordination of multiple cellular and developmental events. Dysfunction of lipid metabolism can lead to neurodevelopmental disorders. Lowe syndrome (LS) is a recessive X-linked disorder associated with proximal tubular renal disease, congenital cataracts and glaucoma, and central nervous system developmental delays. Mutations in OCRL, which encodes an inositol polyphosphate 5-phosphatase, lead to the development of LS. The cellular mechanism responsible for neuronal dysfunction in LS is unknown. Here we show depletion of mitochondrial DNA and decrease in mitochondrial activities result in neuronal differentiation defects. Increased astrocytes, which are secondary responders to neurodegeneration, are observed in neuronal (iN) cells differentiated from Lowe patient-derived iPSCs and an LS mouse model. Inactivation of cilia-related sonic hedgehog signaling, which organizes the pattern of cellular neuronal differentiation, is observed in an OCRL knockout, iN cells differentiated from Lowe patient-derived iPSCs, and an LS mouse model. Taken together, our findings indicate that mitochondrial dysfunction and impairment of the ciliary sonic hedgehog signaling pathway represent a novel pathogenic mechanism underlying the disrupted neuronal differentiation observed in LS.
Collapse
Affiliation(s)
- Chien-Hui Lo
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Siyu Chen
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jingyu Zhao
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Zhiquan Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Biao Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Qing Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tia J. Kowal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
- Palo Alto Veterans Administration, Palo Alto, CA, USA
- Maternal Children Health Research Institute at Stanford, Stanford University School of Medicine, Palo Alto, CA, USA
- BioX, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
39
|
Wang J, Zhu F, Luo R, Cui Y, Zhang Z, Xu M, Zhao Y, He Y, Yang W, Li N, Zhu Z, Chen Y, Wang T, Jiang X, Lin C. YAP Alleviates Pulmonary Fibrosis Through Promoting Alveolar Regeneration via Modulating the Stemness of Alveolar Type 2 Cells. Stem Cells Dev 2024; 33:586-594. [PMID: 39119800 DOI: 10.1089/scd.2024.0101] [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] [Indexed: 08/10/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with no cure except transplantation. Abnormal alveolar epithelial regeneration is a key driver of IPF development. The function of Yes1 Associated Transcriptional Regulator (YAP) in alveolar regeneration and IPF pathogenesis remains elusive. Here, we first revealed the activation of YAP in alveolar epithelium 2 cells (AEC2s) from human IPF lungs and fibrotic mouse lungs. Notably, conditional deletion of YAP in mouse AEC2s exacerbated bleomycin-induced pulmonary fibrosis. Intriguingly, we showed in both conditional knockout mice and alveolar organoids that YAP deficiency impaired AEC2 proliferation and differentiation into alveolar epithelium 1 cells (AEC1s). Mechanistically, YAP regulated expression levels of genes associated with cell cycle progression and AEC1 differentiation. Furthermore, overexpression of YAP in vitro promoted cell proliferation. These results indicate the critical role of YAP in alveolar regeneration and IPF pathogenesis. Our findings provide new insights into the regulation of alveolar regeneration and IPF pathogenesis, paving the road for developing novel treatment strategies.
Collapse
Affiliation(s)
- Jingyu Wang
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Fengqing Zhu
- Department of Pulmonary and Critical Care Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Renru Luo
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Yingyin Cui
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Ziyu Zhang
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Mengling Xu
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Yuanyuan Zhao
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Yonghui He
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Wenqing Yang
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Nianle Li
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Zhu Zhu
- GuangDong GemPharmatech Co., Ltd., Foshan, China
| | | | - Tao Wang
- GuangDong GemPharmatech Co., Ltd., Foshan, China
| | - Xuan Jiang
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Chuwen Lin
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
40
|
Cui X, Fu J. Reinitiating lung development: a novel approach in the management of bronchopulmonary dysplasia. Respir Res 2024; 25:384. [PMID: 39449014 PMCID: PMC11515458 DOI: 10.1186/s12931-024-02996-8] [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] [Received: 06/12/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the predominant chronic lung disease in preterm infants, linked with various adverse long-term outcomes. Multiple prenatal and postnatal risk factors can impede lung development, leading to BPD. Current management of BPD relies heavily on pharmacotherapies and alterations in ventilatory strategies. However, these interventions only mitigate BPD symptoms without addressing underlying alveolar, vascular, structural, and functional deficiencies. Given the retarded lung development in infants with BPD and the limitations of existing modalities, new therapeutic approaches are imperative. The induced differentiation of stem/progenitor cells and the spatiotemporal expression patterns of growth factors associated with lung developmental processes are critical for lung development reactivation in BPD, which focuses on stimulating pulmonary vasculogenesis and alveolarization. This review summarizes the process of lung development and offers a comprehensive overview of advancements in therapies designed to reinitiate lung development in BPD. Furthermore, we assessed the potential of these therapies for maintaining lung homeostasis and effectively restoring pulmonary structure and function through stem/progenitor cells and growth factors, which have been widely researched.
Collapse
Affiliation(s)
- Xuewei Cui
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
41
|
Kohon AI, Man K, Hessami A, Mathis K, Webb J, Fang J, Radfar P, Yang Y, Meckes B. Targeting nanoparticles to lung cancer-derived A549 cells based on changes on interstitial stiffness in biomimetic models. iScience 2024; 27:111015. [PMID: 39435151 PMCID: PMC11492096 DOI: 10.1016/j.isci.2024.111015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/27/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
The mechanical properties and forces of the extracellular environment modulate alveolar epithelial cell behavior. To model cancer/fibrosis associated stiffening and dynamic stretch, a biomimetic device was developed that imitates the active forces in the alveolus, while allowing control over the interstitial matrix stiffness. Alveolar epithelial A549 cancer cells were cultured on the devices and their transcriptome was profiled with RNA sequencing. Pathway analysis showed soft materials upregulated the expression of proteoglycans associated with cancer. Consequently, liposomes were modified with peptides targeting heparan sulfate and chondroitin sulfates of the cell surface glycocalyx. Chondroitin sulfate A targeting improved uptake in cells seeded on stiff biomimetic devices, which is attributed to increased chondroitin sulfate proteoglycan localization on cell surfaces in comparison to cells grown on soft devices. These results demonstrate the critical role that mechanical stiffness and stretch play in the alveolus and the importance of including these properties in nanotherapeutic design.
Collapse
Affiliation(s)
- Afia Ibnat Kohon
- Department of Biomedical Engineering, University of North Texas, 3940 N Elm St., Denton, TX 76207, USA
- BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, TX 76203-5017, USA
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, 3940 N Elm St., Denton, TX 76207, USA
| | - Ala Hessami
- Department of Biomedical Engineering, University of North Texas, 3940 N Elm St., Denton, TX 76207, USA
| | - Katelyn Mathis
- Department of Biomedical Engineering, University of North Texas, 3940 N Elm St., Denton, TX 76207, USA
- BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, TX 76203-5017, USA
| | - Jade Webb
- Department of Biomedical Engineering, University of North Texas, 3940 N Elm St., Denton, TX 76207, USA
| | - Joanna Fang
- Department of Biomedical Engineering, University of North Texas, 3940 N Elm St., Denton, TX 76207, USA
| | - Parsa Radfar
- Department of Biomedical Engineering, University of North Texas, 3940 N Elm St., Denton, TX 76207, USA
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, 3940 N Elm St., Denton, TX 76207, USA
| | - Brian Meckes
- Department of Biomedical Engineering, University of North Texas, 3940 N Elm St., Denton, TX 76207, USA
- BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, TX 76203-5017, USA
| |
Collapse
|
42
|
Redin E, Quintanal-Villalonga Á, Rudin CM. Small cell lung cancer profiling: an updated synthesis of subtypes, vulnerabilities, and plasticity. Trends Cancer 2024; 10:935-946. [PMID: 39164163 DOI: 10.1016/j.trecan.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/16/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024]
Abstract
Small cell lung cancer (SCLC) is a devastating disease with high proliferative and metastatic capacity. SCLC has been classified into molecular subtypes based on differential expression of lineage-defining transcription factors. Recent studies have proposed new subtypes that are based on both tumor-intrinsic and -extrinsic factors. SCLC demonstrates substantial intratumoral subtype heterogeneity characterized by highly plastic transcriptional states, indicating that the initially dominant subtype can shift during disease progression and in association with resistance to therapy. Strategies to promote or constrain plasticity and cell fate transitions have nominated novel targets that could prompt the development of more durably effective therapies for patients with SCLC. In this review, we describe the latest advances in SCLC subtype classification and their biological and clinical implications.
Collapse
Affiliation(s)
- Esther Redin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Álvaro Quintanal-Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Pharmacology Program, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
43
|
Ewerling A, May-Simera HL. Evolutionary trajectory for nuclear functions of ciliary transport complex proteins. Microbiol Mol Biol Rev 2024; 88:e0000624. [PMID: 38995044 PMCID: PMC11426024 DOI: 10.1128/mmbr.00006-24] [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] [Indexed: 07/13/2024] Open
Abstract
SUMMARYCilia and the nucleus were two defining features of the last eukaryotic common ancestor. In early eukaryotic evolution, these structures evolved through the diversification of a common membrane-coating ancestor, the protocoatomer. While in cilia, the descendants of this protein complex evolved into parts of the intraflagellar transport complexes and BBSome, the nucleus gained its selectivity by recruiting protocoatomer-like proteins to the nuclear envelope to form the selective nuclear pore complexes. Recent studies show a growing number of proteins shared between the proteomes of the respective organelles, and it is currently unknown how ciliary transport proteins could acquire nuclear functions and vice versa. The nuclear functions of ciliary proteins are still observable today and remain relevant for the understanding of the disease mechanisms behind ciliopathies. In this work, we review the evolutionary history of cilia and nucleus and their respective defining proteins and integrate current knowledge into theories for early eukaryotic evolution. We postulate a scenario where both compartments co-evolved and that fits current models of eukaryotic evolution, explaining how ciliary proteins and nucleoporins acquired their dual functions.
Collapse
Affiliation(s)
- Alexander Ewerling
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| | - Helen Louise May-Simera
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
44
|
Komori T. Regulation of Skeletal Development and Maintenance by Runx2 and Sp7. Int J Mol Sci 2024; 25:10102. [PMID: 39337587 PMCID: PMC11432631 DOI: 10.3390/ijms251810102] [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: 07/29/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Runx2 (runt related transcription factor 2) and Sp7 (Sp7 transcription factor 7) are crucial transcription factors for bone development. The cotranscription factor Cbfb (core binding factor beta), which enhances the DNA-binding capacity of Runx2 and stabilizes the Runx2 protein, is necessary for bone development. Runx2 is essential for chondrocyte maturation, and Sp7 is partly involved. Runx2 induces the commitment of multipotent mesenchymal cells to osteoblast lineage cells and enhances the proliferation of osteoprogenitors. Reciprocal regulation between Runx2 and the Hedgehog, fibroblast growth factor (Fgf), Wnt, and parathyroid hormone-like hormone (Pthlh) signaling pathways and Dlx5 (distal-less homeobox 5) plays an important role in these processes. The induction of Fgfr2 (Fgf receptor 2) and Fgfr3 expression by Runx2 is important for the proliferation of osteoblast lineage cells. Runx2 induces Sp7 expression, and Runx2+ osteoprogenitors become Runx2+Sp7+ preosteoblasts. Sp7 induces the differentiation of preosteoblasts into osteoblasts without enhancing their proliferation. In osteoblasts, Runx2 is required for bone formation by inducing the expression of major bone matrix protein genes, including Col1a1 (collagen type I alpha 1), Col1a2, Spp1 (secreted phosphoprotein 1), Ibsp (integrin binding sialoprotein), and Bglap (bone gamma carboxyglutamate protein)/Bglap2. Bglap/Bglap2 (osteocalcin) regulates the alignment of apatite crystals parallel to collagen fibrils but does not function as a hormone that regulates glucose metabolism, testosterone synthesis, and muscle mass. Sp7 is also involved in Co1a1 expression and regulates osteoblast/osteocyte process formation, which is necessary for the survival of osteocytes and the prevention of cortical porosity. SP7 mutations cause osteogenesis imperfecta in rare cases. Runx2 is an important pathogenic factor, while Runx1, Runx3, and Cbfb are protective factors in osteoarthritis development.
Collapse
Affiliation(s)
- Toshihisa Komori
- Department of Molecular Tumor Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
45
|
Roe K. The epithelial cell types and their multi-phased defenses against fungi and other pathogens. Clin Chim Acta 2024; 563:119889. [PMID: 39117034 DOI: 10.1016/j.cca.2024.119889] [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: 07/02/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024]
Abstract
Mucus and its movements are essential to epithelial tissue immune defenses against pathogens, including fungal pathogens, which can infect respiratory, gastrointestinal or the genito-urinary tracts. Several epithelial cell types contribute to their immune defense. This review focuses on the respiratory tract because of its paramount importance, but the observations will apply to epithelial cell defenses of other mucosal tissue, including the gastrointestinal and genito-urinary tracts. Mucus and its movements can enhance or degrade the immune defenses of the respiratory tract, particularly the lungs. The enhancements include inhaled pathogen entrapments, including fungal pathogens, pollutants and particulates, for their removal. The detriments include smaller lung airway obstructions by mucus, impairing the physical removal of pathogens and impairing vital transfers of oxygen and carbon dioxide between the alveolar circulatory system and the pulmonary air. Inflammation, edema and/or alveolar cellular damage can also reduce vital transfers of oxygen and carbon dioxide between the lung alveolar circulatory system and the pulmonary air. Furthermore, respiratory tract defenses are affected by several fatty acid mediators which activate cellular receptors to manipulate neutrophils, macrophages, dendritic cells, various innate lymphoid cells including the natural killer cells, T cells, γδ T cells, mucosal-associated invariant T cells, NKT cells and mast cells. These mediators include the inflammatory and frequently immunosuppressive prostaglandins and leukotrienes, and the special pro-resolving mediators, which normally resolve inflammation and immunosuppression. The total effects on the various epithelial cell and immune cell types, after exposures to pathogens, pollutants or particulates, will determine respiratory tract health or disease.
Collapse
Affiliation(s)
- Kevin Roe
- Retired United States Patent and Trademark Office, San Jose, CA, United States.
| |
Collapse
|
46
|
Guo ZF, Tongmuang N, Li C, Zhang C, Hu L, Capreri D, Zuo MX, Summer R, Sun J. Inhibiting endothelial cell Mst1 attenuates acute lung injury in mice. JCI Insight 2024; 9:e178208. [PMID: 39253972 PMCID: PMC11385092 DOI: 10.1172/jci.insight.178208] [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: 12/06/2023] [Accepted: 07/25/2024] [Indexed: 09/11/2024] Open
Abstract
Lung endothelium plays a pivotal role in the orchestration of inflammatory responses to acute pulmonary insults. Mammalian sterile 20-like kinase 1 (Mst1) is a serine/threonine kinase that has been shown to play an important role in the regulation of apoptosis, stress responses, and organ growth. This study investigated the role of Mst1 in lung endothelial activation and acute lung injury (ALI). We found that Mst1 was significantly activated in inflamed lung endothelial cells (ECs) and mouse lung tissues. Overexpression of Mst1 promoted nuclear factor κ-B (NF-κB) activation through promoting JNK and p38 activation in lung ECs. Inhibition of Mst1 by either its dominant negative form (DN-Mst1) or its pharmacological inhibitor markedly attenuated cytokine-induced expression of cytokines, chemokines, and adhesion molecules in lung ECs. Importantly, in a mouse model of lipopolysaccharide-induced (LPS-induced) ALI, both deletion of Mst1 in lung endothelium and treatment of WT mice with a pharmacological Mst1 inhibitor significantly protected mice from LPS-induced ALI. Together, our findings identified Mst1 kinase as a key regulator in controlling lung EC activation and suggest that therapeutic strategies aimed at inhibiting Mst1 activation might be effective in the prevention and treatment of inflammatory lung diseases.
Collapse
Affiliation(s)
- Zhi-Fu Guo
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai
| | - Nopprarat Tongmuang
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Chao Li
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Chen Zhang
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Louis Hu
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Daniel Capreri
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mei-Xing Zuo
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ross Summer
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jianxin Sun
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
47
|
Villares E, Gerecht S. Engineered Biomaterials and Model Systems to Study YAP/TAZ in Cancer. ACS Biomater Sci Eng 2024; 10:5550-5561. [PMID: 39190867 DOI: 10.1021/acsbiomaterials.4c01170] [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] [Indexed: 08/29/2024]
Abstract
The transcriptional coactivators yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are master regulators involved in a multitude of cancer types and a wide range of tumorigenic events, including cancer stem cell renewal, invasion, metastasis, tumor precursor emergence, and drug resistance. YAP/TAZ are known to be regulated by several external cues and stimuli, such as extracellular matrix stiffness, cell spreading, cell geometry, and shear stress. Therefore, there is a need in the field of cancer research to develop and design relevant in vitro models that can accurately reflect the complex biochemical and biophysical cues of the tumor microenvironment central to the YAP/TAZ signaling nexus. While much progress has been made, this remains a major roadblock to advancing research in this field. In this review, we highlight the current engineered biomaterials and in vitro model systems that can be used to advance our understanding of how YAP/TAZ shapes several aspects of cancer. We begin by discussing current 2D and 3D hydrogel systems that model the YAP/TAZ response to ECM stiffness. We then examine the current trends in organoid culture systems and the use of microfluidics to model the effects of cellular density and shear stress on YAP/TAZ. Finally, we analyze the ongoing pitfalls of the present models used and important future directions in engineering systems that will advance our current knowledge of YAP/TAZ in cancer.
Collapse
Affiliation(s)
- Emma Villares
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27705, United States
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27705, United States
| |
Collapse
|
48
|
Dumoulin A, Wilson NH, Tucker KL, Stoeckli ET. A cell-autonomous role for primary cilium-mediated signaling in long-range commissural axon guidance. Development 2024; 151:dev202788. [PMID: 39157903 PMCID: PMC11423920 DOI: 10.1242/dev.202788] [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: 02/23/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Ciliopathies are characterized by the absence or dysfunction of primary cilia. Despite the fact that cognitive impairments are a common feature of ciliopathies, how cilia dysfunction affects neuronal development has not been characterized in detail. Here, we show that primary cilium-mediated signaling is required cell-autonomously by neurons during neural circuit formation. In particular, a functional primary cilium is crucial during axonal pathfinding for the switch in responsiveness of axons at a choice point or intermediate target. Using different animal models and in vivo, ex vivo and in vitro experiments, we provide evidence for a crucial role of primary cilium-mediated signaling in long-range axon guidance. The primary cilium on the cell body of commissural neurons transduces long-range guidance signals sensed by growth cones navigating an intermediate target. In extension of our finding that Shh is required for the rostral turn of post-crossing commissural axons, we suggest a model implicating the primary cilium in Shh signaling upstream of a transcriptional change of axon guidance receptors, which in turn mediate the repulsive response to floorplate-derived Shh shown by post-crossing commissural axons.
Collapse
Affiliation(s)
- Alexandre Dumoulin
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Nicole H Wilson
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Kerry L Tucker
- University of New England, College of Osteopathic Medicine, Department of Biomedical Sciences, Center for Excellence in the Neurosciences, Biddeford, ME 04005, USA
| | - Esther T Stoeckli
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- University Research Priority Program 'Adaptive Brain Circuits in Development and Learning' (URPP AdaBD), University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
49
|
Yuze Ma, Liu N, Shao X, Shi T, Lin J, Liu B, Shen T, Guo B, Jiang Q. Mechanical loading on osteocytes regulates thermogenesis homeostasis of brown adipose tissue by influencing osteocyte-derived exosomes. J Orthop Translat 2024; 48:39-52. [PMID: 39087139 PMCID: PMC11287067 DOI: 10.1016/j.jot.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/25/2024] [Accepted: 06/19/2024] [Indexed: 08/02/2024] Open
Abstract
Background Osteocytes are the main stress-sensing cells in bone. The substances secreted by osteocytes under mechanical loading play a crucial role in maintaining body homeostasis. Osteocytes have recently been found to release exosomes into the circulation, but whether they are affected by mechanical loading or participate in the regulation of systemic homeostasis remains unclear. Methods We used a tail-suspension model to achieve mechanical unloading on osteocytes. Osteocyte-specific CD63 reporter mice were used for osteocyte exosome tracing. Exosome detection and inhibitor treatment were performed to confirm the effect of mechanical loading on exosome secretion by osteocytes. Co-culture, GW4869 and exosome treatment were used to investigate the biological functions of osteocyte-derived exosomes on brown adipose tissue (BAT) and primary brown adipocytes. Osteocyte-specific Dicer KO mice were used to screen for loading-sensitive miRNAs. Dual luciferase assay was performed to validate the selected target gene. Results Firstly, we found the thermogenic activity was increased in BAT of mice subjected to tail suspension, which is due to the effect of unloaded bone on circulating exosomes. Further, we showed that the secretion of exosomes from osteocytes is regulated by mechanical loading, and osteocyte-derived exosomes can reach BAT and affect thermogenic activity. More importantly, we confirmed the effect of osteocyte exosomes on BAT both in vivo and in vitro. Finally, we discovered that let-7e-5p contained in exosomes is under regulation of mechanical loading and regulates thermogenic activity of BAT by targeting Ppargc1a. Conclusion Exosomes derived from osteocytes are loading-sensitive, and play a vital role in regulation on BAT, suggesting that regulation of exosomes secretion can restore homeostasis. The translational potential of this article This study provides a biological rationale for using osteocyte exosomes as potential agents to modulate BAT and even whole-body homeostasis. It also provides a new pathological basis and a new treatment approach for mechanical unloading conditions such as spaceflight.
Collapse
Affiliation(s)
- Yuze Ma
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Na Liu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xiaoyan Shao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Tianshu Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jiaquan Lin
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Bin Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tao Shen
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Baosheng Guo
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
50
|
Wright NJ, Zhang F, Suo Y, Kong L, Yin Y, Fedor JG, Sharma K, Borgnia MJ, Im W, Lee SY. Antiviral drug recognition and elevator-type transport motions of CNT3. Nat Chem Biol 2024; 20:1144-1153. [PMID: 38418906 PMCID: PMC11625470 DOI: 10.1038/s41589-024-01559-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024]
Abstract
Nucleoside analogs have broad clinical utility as antiviral drugs. Key to their systemic distribution and cellular entry are human nucleoside transporters. Here, we establish that the human concentrative nucleoside transporter 3 (CNT3) interacts with antiviral drugs used in the treatment of coronavirus infections. We report high-resolution single-particle cryo-electron microscopy structures of bovine CNT3 complexed with antiviral nucleosides N4-hydroxycytidine, PSI-6206, GS-441524 and ribavirin, all in inward-facing states. Notably, we found that the orally bioavailable antiviral molnupiravir arrests CNT3 in four distinct conformations, allowing us to capture cryo-electron microscopy structures of drug-loaded outward-facing and drug-loaded intermediate states. Our studies uncover the conformational trajectory of CNT3 during membrane transport of a nucleoside analog antiviral drug, yield new insights into the role of interactions between the transport and the scaffold domains in elevator-like domain movements during drug translocation, and provide insights into the design of nucleoside analog antiviral prodrugs with improved oral bioavailability.
Collapse
Affiliation(s)
- Nicholas J Wright
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Feng Zhang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Yang Suo
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Lingyang Kong
- Departments of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Ying Yin
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Justin G Fedor
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Kedar Sharma
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Mario J Borgnia
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Wonpil Im
- Departments of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|