1
|
Vohra M, Kumar S, Sohnen P, Kaur S, Swamynathan S, Hirose T, Kozmik Z, Swamynathan SK. Pard3 promotes corneal epithelial stratification and homeostasis by regulating apical-basal polarity, cytoskeletal organization and tight junction-mediated barrier function. Ocul Surf 2025; 37:201-215. [PMID: 40188986 DOI: 10.1016/j.jtos.2025.04.001] [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: 12/16/2024] [Revised: 03/24/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
PURPOSE To document the expression of apical-basal polarity (ABP) determinants in the mouse corneal epithelium (CE) and elucidate the functions of Pard3 in establishment and maintenance of ABP, stratification, homeostasis, and barrier function in the CE. METHODS Pard3Δ/ΔC mice (Pard3LoxP/LoxP; Aldh3A1-Cre/+) with cornea-specific Pard3 ablation were generated by breeding Aldh3A1-Cre/+ with Pard3LoxP/LoxP mice. The control (Aldh3A1-Cre/+ or Pard3LoxP/LoxP alone) and Pard3Δ/ΔC corneal histology, ocular surface properties, barrier function, and actin cytoskeleton were assessed by Haematoxylin and Eosin staining of paraformaldehyde-fixed, paraffin-embedded tissues, scanning electron microscopy, fluorescein staining, and phalloidin staining, respectively. The expression of specific markers of interest was evaluated by qRT-PCR, immunoblots and immunofluorescent staining. RESULTS Dynamic changes were observed in the expression and localization of ABP determinants as the CE stratified and matured between post-natal day 5 (PN5) and PN52. Adult Pard3Δ/ΔC CE contained fewer cell layers with rounded basal cells, and loosely adherent superficial cells lacking microplicae. Adult Pard3Δ/ΔC CE also displayed impaired barrier function with decreased expression of tight junction, adherens junction, and desmosome components, disrupted actin cytoskeletal organization, increased proliferation, and upregulation of transcription factors that drive epithelial-mesenchymal transition (EMT). CONCLUSIONS Disruption of ABP in Pard3Δ/ΔC CE, altered expression of cell junction complex components and disorganized actin cytoskeleton, increased cell proliferation, and upregulated EMT transcription factors suggest that the ABP-determinant Pard3 promotes CE features while suppressing mesenchymal cell fate. Collectively, these results elucidate that Pard3-mediated ABP is essential for CE stratification, homeostasis and barrier function.
Collapse
Affiliation(s)
- Mehak Vohra
- Department of Ophthalmology, University of South Florida, Tampa, FL, USA
| | - Simran Kumar
- Department of Ophthalmology, University of South Florida, Tampa, FL, USA
| | - Peri Sohnen
- Department of Ophthalmology, University of South Florida, Tampa, FL, USA
| | - Satinder Kaur
- Department of Ophthalmology, University of South Florida, Tampa, FL, USA
| | - Sudha Swamynathan
- Department of Ophthalmology, University of South Florida, Tampa, FL, USA
| | - Tomonori Hirose
- Department of Molecular Biology, Yokohama City University, Yokohama, Japan
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | | |
Collapse
|
2
|
Morabito M, Thibodot P, Gigandet A, Compagnon P, Toso C, Berishvili E, Lacotte S, Peloso A. Liver Extracellular Matrix in Colorectal Liver Metastasis. Cancers (Basel) 2025; 17:953. [PMID: 40149289 PMCID: PMC11939972 DOI: 10.3390/cancers17060953] [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: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
The liver is the most common site of metastasis of colorectal cancer (CRC), and colorectal liver metastasis is one of the major causes of CRC-related deaths worldwide. The tumor microenvironment, particularly the extracellular matrix (ECM), plays a critical role in CRC metastasis and chemoresistance. Based on findings from clinical and basic research, this review attempts to offer a complete understanding of the role of the ECM in colorectal liver metastasis and to suggest potential ways for therapeutic intervention. First, the ECMs' role in regulating cancer cell fate is explored. We then discuss the hepatic ECM fingerprint and its influence on the metastatic behavior of CRC cells, highlighting key molecular interactions that promote metastasis. In addition, we examine how changes in the ECM within the metastatic niche contribute to chemoresistance, focusing on ECM remodeling by ECM stiffening and the activation of specific signaling pathways. Understanding these mechanisms is crucial for the development of novel strategies to overcome metastasis and improve outcomes for CRC patients.
Collapse
Affiliation(s)
- Marika Morabito
- General, Emergency and Transplant Surgery Department, ASST Settelaghi, University Hospital and Faculty of Medicine of Insubria, 21100 Varese, Italy
| | - Pauline Thibodot
- Hepato-Biliary Center, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, 94800 Villejuif, France
| | - Anthony Gigandet
- School of Medecine, Faculty of Medecine, University of Geneva, 1211 Geneva, Switzerland
| | - Philippe Compagnon
- Division of Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland;
| | - Christian Toso
- Division of Abdominal Surgery and Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland;
| | - Stéphanie Lacotte
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland;
| | - Andrea Peloso
- Hepato-Biliary Center, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, 94800 Villejuif, France
- Division of Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland;
- Division of Abdominal Surgery and Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland
| |
Collapse
|
3
|
Nguyen A, Heim JB, Cordara G, Chan MC, Johannesen H, Charlesworth C, Li M, Azumaya CM, Madden B, Krengel U, Meves A, Campbell MG. Structural and functional characterization of integrin α5-targeting antibodies for anti-angiogenic therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631572. [PMID: 39829743 PMCID: PMC11741253 DOI: 10.1101/2025.01.08.631572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Integrins are a large family of heterodimeric receptors important for cell adhesion and signaling. Integrin α5β1, also known as the fibronectin receptor, is a key mediator of angiogenesis and its dysregulation is associated with tumor proliferation, progression, and metastasis. Despite numerous efforts, α5β1-targeting therapeutics have been unsuccessful in large part due to efficacy and off-target effects. To mediate activation and signaling, integrins undergo drastic conformational changes. However, how therapeutics influence or are affected by integrin conformation remains incompletely characterized. Using cell biology, biophysics, and electron microscopy, we shed light on these relationships by characterizing two potentially therapeutic anti-α5β1 antibodies, BIIG2 and MINT1526A. We show that both antibodies bind α5β1 with nanomolar affinity and reduce angiogenesis in vitro. We demonstrate BIIG2 reduces tumor growth in two human xenograft mouse models and exhibits a strong specificity for connective tissue-resident fibroblasts and melanoma cells. Using electron microscopy, we map out the molecular interfaces mediating the integrin-antibody interactions and reveal that although both antibodies have overlapping epitopes and block fibronectin binding via steric hindrance, the effect on the conformational equilibrium is drastically different. While MINT1526A constricts α5β1's range of flexibility, BIIG2 binds without restricting the available conformational states. These mechanistic insights, coupled with the functional analysis, guide which aspects should be prioritized to avoid off-target effects or partial agonism in the design of future integrin-targeted therapeutics.
Collapse
Affiliation(s)
- Adam Nguyen
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington 98109, USA
- Biological Physics Structure and Design Program, University of Washington, Seattle, Washington 98195, USA
| | - Joel B. Heim
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota 55905, USA
- Department of Chemistry, University of Oslo, NO-0315 Oslo, Norway
- Current Address: Nykode Therapeutics, Oslo Science Park, 0349 Oslo, Norway
| | - Gabriele Cordara
- Department of Chemistry, University of Oslo, NO-0315 Oslo, Norway
| | - Matthew C. Chan
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington 98109, USA
| | - Hedda Johannesen
- Department of Chemistry, University of Oslo, NO-0315 Oslo, Norway
| | - Cristine Charlesworth
- Medical Genome Facility, Proteomics Core, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Ming Li
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Caleigh M. Azumaya
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington 98109, USA
- Current Address: Genentech, South San Francisco, California 94080, USA
| | - Benjamin Madden
- Medical Genome Facility, Proteomics Core, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Ute Krengel
- Department of Chemistry, University of Oslo, NO-0315 Oslo, Norway
| | - Alexander Meves
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Melody G. Campbell
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington 98109, USA
- Biological Physics Structure and Design Program, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
4
|
Arnaout MA. The Integrin Receptors: From Discovery to Structure to Medicines. Immunol Rev 2025; 329:e13433. [PMID: 39724488 PMCID: PMC11752789 DOI: 10.1111/imr.13433] [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: 11/13/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
Innate immune cells perform vital tasks in detecting, seeking, and eliminating invading pathogens, thus ensuring host survival. However, loss of function of these cells or their overactive response to tissue injury often causes serious ailments. It is, therefore, crucial to understand at a basic level how these cells function in health and disease. A major step toward this goal came from studies I conducted in the late 1970s investigating the cause of life-threatening bacterial infections in a pediatric patient. This work led us to trace this disease to the inability of the patient's neutrophils to seek and clear infections due to an inherited deficiency in leukocyte adhesion caused by the loss of a plasma membrane glycoprotein complex now known as CD11/CD18 or β2 integrins. I followed this work by determining the 3-dimensional structures of integrins. These studies provided the foundation for understanding the unique properties of integrins in mediating bidirectional cell adhesion signaling and enabled a structure-guided design of compounds to dial down overactive integrins in common disorders, including thromboinflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- M Amin Arnaout
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Sekiya T, Holley MC. The Glial Scar: To Penetrate or Not for Motor Pathway Restoration? Cell Transplant 2025; 34:9636897251315271. [PMID: 40152462 PMCID: PMC11951902 DOI: 10.1177/09636897251315271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/25/2024] [Accepted: 01/08/2025] [Indexed: 03/29/2025] Open
Abstract
Although notable progress has been made, restoring motor function from the brain to the muscles continues to be a substantial clinical challenge in motor neuron diseases/disorders such as spinal cord injury (SCI). While cell transplantation has been widely explored as a potential therapeutic method for reconstructing functional motor pathways, there remains considerable opportunity for enhancing its therapeutic effectiveness. We reviewed studies on motor pathway regeneration to identify molecular and ultrastructural cues that could enhance the efficacy of cell transplantation. While the glial scar is often cited as an intractable barrier to axon regeneration, this mainly applies to axons trying to penetrate its "core" to reach the opposite side. However, the glial scar exhibits a "duality," with an anti-regenerative core and a pro-regenerative "surface." This surface permissiveness is attributed to pro-regenerative molecules, such as laminin in the basement membrane (BM). Transplanting donor cells onto the BM, which forms plastically after injury, may significantly enhance the efficacy of cell transplantation. Specifically, forming detour pathways between transplanted cells and endogenous propriospinal neurons on the pro-regenerative BM may efficiently bypass the intractable scar core and promote motor pathway regeneration. We believe harnessing the tissue's innate repair capacity is crucial, and targeting post-injury plasticity in astrocytes and Schwann cells, especially those associated with the BM that has predominantly been overlooked in the field of SCI research, can advance motor system restoration to a new stage. A shift in cell delivery routes-from the traditional intra-parenchymal (InP) route to the transplantation of donor cells onto the pro-regenerative BM via the extra-parenchymal (ExP) route-may signify a transformative step forward in neuro-regeneration research. Practically, however, the complementary use of both InP and ExP methods may offer the most substantial benefit for restoring motor pathways. We aim for this review to deepen the understanding of cell transplantation and provide a framework for evaluating the efficacy of this therapeutic modality in comparison to others.
Collapse
Affiliation(s)
- Tetsuji Sekiya
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Neurological Surgery, Hikone Chuo Hospital, Hikone, Japan
| | - Matthew C. Holley
- Department of Biomedical Science, University of Sheffield, Sheffield, England
| |
Collapse
|
6
|
Chinvattanachot G, Rivas D, Duque G. Mechanisms of muscle cells alterations and regeneration decline during aging. Ageing Res Rev 2024; 102:102589. [PMID: 39566742 DOI: 10.1016/j.arr.2024.102589] [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: 07/13/2024] [Revised: 10/27/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Skeletal muscles are essential for locomotion and body metabolism regulation. As muscles age, they lose strength, elasticity, and metabolic capability, leading to ineffective motion and metabolic derangement. Both cellular and extracellular alterations significantly influence muscle aging. Satellite cells (SCs), the primary muscle stem cells responsible for muscle regeneration, become exhausted, resulting in diminished population and functionality during aging. This decline in SC function impairs intercellular interactions as well as extracellular matrix production, further hindering muscle regeneration. Other muscle-resident cells, such as fibro-adipogenic progenitors (FAPs), pericytes, and immune cells, also deteriorate with age, reducing local growth factor activities and responsiveness to stress or injury. Systemic signaling, including hormonal changes, contributes to muscle cellular catabolism and disrupts muscle homeostasis. Collectively, these cellular and environmental components interact, disrupting muscle homeostasis and regeneration in advancing age. Understanding these complex interactions offers insights into potential regenerative strategies to mitigate age-related muscle degeneration.
Collapse
Affiliation(s)
- Guntarat Chinvattanachot
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| | - Daniel Rivas
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Gustavo Duque
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
7
|
Hang Q, Zuo S, Yang Y, Wang Y, Li C, Li W, Guo J, Hou S, Huang H. USP33 is an integrin α6 deubiquitinase and promotes esophageal squamous cell carcinoma cell migration and metastasis. J Cancer Res Clin Oncol 2024; 150:511. [PMID: 39589547 PMCID: PMC11599434 DOI: 10.1007/s00432-024-06041-5] [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: 10/22/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE The deubiquitinating enzymes (DUBs) have been linked to cancer initiation and progression. Although ubiquitin-specific protease 33 (USP33) represents a significant factor in regulating various tumor cell behaviors, its specific biological functions and precise mechanisms in esophageal squamous cell carcinoma (ESCC) progression remain unclear. METHODS The expressions of USP33 mRNA in GEO databases, clinical ESCC samples, and USP33 protein were analyzed using bioinformatics, RT-PCR, and immunohistochemistry, respectively. Using Kaplan-Meier survival curves, the log-rank test was used to determine the cumulative survival rate. Western blotting was used to determine indicated protein expression. The cell biological functions were evaluated by cell growth assay, transwell, cell adhesion, and cell spreading assay, respectively. The interaction between USP33 and integrins was detected by immunoprecipitation, and the deubiquitination was performed by deubiquitination assay. The metastatic ability was checked by tail vein injection. RESULTS A significant positive correlation was found between USP33 expression and clinical TNM stage, T classification, and poor prognosis in patients with ESCC. USP33 promoted laminin-dependent adhesion, spreading, and migration of ESCC cells but not their proliferation. Mechanistically, USP33 mediates cell migration through binding, deubiquinating, and stabilizing integrin α6. USP33 knockdown could inhibit ESCC cell migration and metastasis majorly through integrin α6. CONCLUSION This study reveals a novel mechanism of USP33 in promoting laminin-dependent ESCC cell migration and metastasis through integrin α6, suggesting that USP33 may be a promising target for treating ESCC.
Collapse
Affiliation(s)
- Qinglei Hang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China.
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, Suining, Jiangsu Province, 221200, China.
| | - Shiying Zuo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Yawen Yang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Yuanzhi Wang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Caimin Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Wenqian Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Jingya Guo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Sicong Hou
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China.
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China.
| | - Haifeng Huang
- Department of Laboratory Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, 224006, China.
- Department of Laboratory Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224006, China.
| |
Collapse
|
8
|
Xia T, Pan Z, Wan H, Li Y, Mao G, Zhao J, Zhang F, Pan S. Mechanisms of mechanical stimulation in the development of respiratory system diseases. Am J Physiol Lung Cell Mol Physiol 2024; 327:L724-L739. [PMID: 39316681 DOI: 10.1152/ajplung.00122.2024] [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: 04/09/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
During respiration, mechanical stress can initiate biological responses that impact the respiratory system. Mechanical stress plays a crucial role in the development of the respiratory system. However, pathological mechanical stress can impact the onset and progression of respiratory diseases by influencing the extracellular matrix and cell transduction processes. In this article, we explore the mechanisms by which mechanical forces communicate with and influence cells. We outline the basic knowledge of respiratory mechanics, elucidating the important role of mechanical stimulation in influencing respiratory system development and differentiation from a microscopic perspective. We also explore the potential mechanisms of mechanical transduction in the pathogenesis and development of respiratory diseases such as asthma, lung injury, pulmonary fibrosis, and lung cancer. Finally, we look forward to new research directions in cellular mechanotransduction, aiming to provide fresh insights for future therapeutic research on respiratory diseases.
Collapse
Affiliation(s)
- Tian Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ziyin Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Haoxin Wan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yongsen Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Guocai Mao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fangbiao Zhang
- Department of Cardiothoracic Surgery, Lishui Municipal Central Hospital, Lishui, People's Republic of China
| | - Shu Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
9
|
Nonnast E, Mira E, Mañes S. Biomechanical properties of laminins and their impact on cancer progression. Biochim Biophys Acta Rev Cancer 2024; 1879:189181. [PMID: 39299492 DOI: 10.1016/j.bbcan.2024.189181] [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: 03/19/2024] [Revised: 07/30/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Laminins (LMs) constitute a family of heterotrimeric glycoproteins essential for the formation of basement membranes (BM). They act as molecular bridges between cells and the extracellular matrix (ECM), thereby transmitting signals influencing cell behavior and tissue organization. In the realm of cancer pathobiology, LMs regulate key processes such as migration, differentiation, or fibrosis. This review critically examines the multifaceted impact of LMs on tumor progression, with a particular focus on the isoform-specific structure-function relationships, and how this structural diversity contributes to the biomechanical properties of BMs. LM interactions with integrin and non-integrin cell surface receptors, as well as with other ECM proteins, modify the response of cancer cells to the ECM stiffness, ultimately influencing the capacity of malignant cells to breach the BM, a limiting step in metastatic dissemination. Comprehension of the mechanisms underlying LM-driven tumor biomechanics holds potential for better understand cancer pathobiology and design new targeted therapeutic strategies.
Collapse
Affiliation(s)
- Elena Nonnast
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049 Madrid, Spain
| | - Emilia Mira
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049 Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049 Madrid, Spain.
| |
Collapse
|
10
|
Nowwarote N, Chahlaoui Z, Petit S, Duong LT, Dingli F, Loew D, Chansaenroj A, Kornsuthisopon C, Osathanon T, Ferre FC, Fournier BPJ. Decellularized extracellular matrix derived from dental pulp stem cells promotes gingival fibroblast adhesion and migration. BMC Oral Health 2024; 24:1166. [PMID: 39354504 PMCID: PMC11443845 DOI: 10.1186/s12903-024-04882-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/06/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Decellularized extracellular matrix (dECM) has been proposed as a useful source of biomimetic materials for regenerative medicine due to its biological properties that regulate cell behaviors. The present study aimed to investigate the influence of decellularized ECM derived from dental pulp stem cells (DPSCs) on gingival fibroblast (GF) cell behaviors. Cells were isolated from dental pulp and gingival tissues. ECM was derived from culturing dental pulp stem cells in growth medium supplemented with ascorbic acid. A bioinformatic database of the extracellular matrix was constructed using Metascape. GFs were reseeded onto dECM, and their adhesion, spreading, and organization were subsequently observed. The migration ability of the cells was determined using a scratch assay. Protein expression was evaluated using immunofluorescence staining. RESULTS Type 1 collagen and fibronectin were detected on the ECM and dECM derived from DPSCs. Negative phalloidin and nuclei were noted in the dECM. The proteomic database revealed enrichment of several proteins involved in ECM organization, ECM-receptor interaction, and focal adhesion. Compared with those on the controls, the GFs on the dECM exhibited more organized stress fibers. Furthermore, cultured GFs on dECM exhibited significantly enhanced migration and proliferation abilities. Interestingly, GFs seeded on dECM showed upregulation of FN1, ITGB3, and CTNNB1 mRNA levels. CONCLUSIONS ECM derived from DSPCs generates a crucial microenvironment for regulating GF adhesion, migration and proliferation. Therefore, decellularized ECM from DPSCs could serve as a matrix for oral tissue repair.
Collapse
Affiliation(s)
- Nunthawan Nowwarote
- Centre de Recherche des Cordeliers, Molecular Oral Pathophysiology, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université, Paris, 75006, France.
- Department of Oral Biology, Dental Faculty Garancière, Université Paris Cité, Paris, 75006, France.
| | - Zakaria Chahlaoui
- Centre de Recherche des Cordeliers, Molecular Oral Pathophysiology, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université, Paris, 75006, France
| | - Stephane Petit
- Centre de Recherche des Cordeliers, Molecular Oral Pathophysiology, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université, Paris, 75006, France
| | - Lucas T Duong
- Centre de Recherche des Cordeliers, Molecular Oral Pathophysiology, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université, Paris, 75006, France
| | - Florent Dingli
- Centre de Recherche, CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Damarys Loew
- Centre de Recherche, CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Ajjima Chansaenroj
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chatvadee Kornsuthisopon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Thanaphum Osathanon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Francois Come Ferre
- Centre de Recherche des Cordeliers, Molecular Oral Pathophysiology, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université, Paris, 75006, France
- Department of Oral Biology, Dental Faculty Garancière, Université Paris Cité, Paris, 75006, France
| | - Benjamin P J Fournier
- Centre de Recherche des Cordeliers, Molecular Oral Pathophysiology, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université, Paris, 75006, France
- Department of Oral Biology, Dental Faculty Garancière, Université Paris Cité, Paris, 75006, France
| |
Collapse
|
11
|
Tollabi M, Poursalehi Z, Mehrafshar P, Bakhtiari R, Sarmadi VH, Tayebi L, Haramshahi SMA. Insight into the role of integrins and integrins-targeting biomaterials in bone regeneration. Connect Tissue Res 2024; 65:343-363. [PMID: 39297793 PMCID: PMC11541888 DOI: 10.1080/03008207.2024.2396002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/06/2024] [Accepted: 08/19/2024] [Indexed: 10/17/2024]
Abstract
Features of the extracellular matrix, along with biochemical factors, have a momentous impress in making genes on and/or off. The interaction of cells and the extracellular matrix is mediated by integrins. Therefore, these molecules have pivotal roles in regulating cell behaviors. Integrins include a group of molecules with a variety of characteristics that can affect different molecular cascades. Considering the importance of these molecules in tissue regeneration after injury, it is necessary to know well the integrins involved in the process of connecting cells to the extracellular matrix in each tissue.With the increase in life expectancy, bone tissue engineering has received more attention from researchers. Integrins are critical components in osteoblast differentiation, survival, and bone mechanotransduction. During osteogenic differentiation in stem cells, specific integrins facilitate multiple signaling pathways through their cytoplasmic domain, leading to the induction of osteogenic differentiation. Also, due to the importance of using biomaterials in bone tissue engineering, efforts have been made to design and use biomaterials with maximum interaction with integrins. Notably, the use of RGD peptide or fibronectin for surface modification is a well-established and commonly employed approach to manipulate integrin activity.This review article looks into integrins' role in bone development and regeneration. It then goes on to explore the complex mechanisms by which integrins contribute to these processes. In addition, this review discusses the use of natural and synthetic biomaterials that target integrins to promote bone regeneration.
Collapse
Affiliation(s)
- Mohammad Tollabi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Poursalehi
- Department of Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Parichehr Mehrafshar
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Vahid Hosseinpour Sarmadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Seyed Mohammad Amin Haramshahi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Hazrate Fatemeh Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Totoki Y, Mutsuzaki H, Yanagisawa Y, Sogo Y, Yasunaga M, Noguchi H, Matsumoto Y, Koda M, Ito A, Yamazaki M. Do Stainless-Steel Pins Coated with Fibroblast Growth Factor-Calcium Phosphatase Composite Layers Have Anti-Infective Effects? MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1419. [PMID: 39336460 PMCID: PMC11434512 DOI: 10.3390/medicina60091419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024]
Abstract
Background: The most problematic complication of external fixation is infection at the pin insertion site. Technology that improves the adhesion of the external fixation pin to the skin, subcutaneous tissue, and bone may prevent infection at the pin site. The purpose of this study is to formulate a calcium phosphate-fibroblast growth factor (Cp-FGF) coating on a stainless-steel external fixation pin and to verify its effectiveness in reducing infection at the pin site and its possible influence on bone fixation in animal experiments. Methods: We compared stainless-steel screws without coating (SS group; n = 32), those with a calcium phosphate coating (Cp group; n = 30), those with a Cp-FGF coating (FGF group; n = 32), and those with a Cp-FGF coating having enhanced biological activity (FGF+ group; n = 32) in male Japanese white domesticated rabbits. Screws were inserted percutaneously into the bilateral proximal tibial diaphysis of the rabbits and implanted for 4 weeks. Screws and periscrew tissue were observed postoperatively for qualitatively assessing infection. Results: Infection assessment by gross findings after 4 weeks (at screw removal) showed no significant differences between the groups. Histopathological evaluation of soft tissue infection and bone tissue infection showed no significant differences between the groups for either soft tissue or bone tissue. Since neither the FGF+ group nor the FGF group showed anti-infective effects, the biological activity of FGF is not the only determining factor. We compared SEM, XRD, coating detaching test, sustained release test, and bioassay to examine physicochemical properties among the coatings but found no sufficient differences. Conclusions: It is suggested that improving the tissue adhesion to and/or biocompatibility of pins is also important to improve the in vivo performance of Cp-FGF-coated external fixation pins.
Collapse
Affiliation(s)
- Yasukazu Totoki
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Hirotaka Mutsuzaki
- Center for Medical Science, Ibaraki Prefectural University of Health Sciences, Ami 300-0394, Ibaraki, Japan
- Department of Orthopedic Surgery, Ibaraki Prefectural University of Health Sciences Hospital, Ami 300-0331, Ibaraki, Japan
| | - Yohei Yanagisawa
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Yu Sogo
- National Institute of Advanced Industrial Science and Technology (AIST), Health and Medical Research Institute, Tsukuba 305-8566, Ibaraki, Japan; (Y.S.); (M.Y.); (A.I.)
| | - Mayu Yasunaga
- National Institute of Advanced Industrial Science and Technology (AIST), Health and Medical Research Institute, Tsukuba 305-8566, Ibaraki, Japan; (Y.S.); (M.Y.); (A.I.)
| | - Hiroshi Noguchi
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Yukei Matsumoto
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Masao Koda
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
| | - Atsuo Ito
- National Institute of Advanced Industrial Science and Technology (AIST), Health and Medical Research Institute, Tsukuba 305-8566, Ibaraki, Japan; (Y.S.); (M.Y.); (A.I.)
| | - Masashi Yamazaki
- Department of Orthopaedic Surgery, University of Tsukuba, Tsukuba 305-8571, Ibaraki, Japan; (Y.T.); (Y.Y.); (H.N.); (Y.M.); (M.K.); (M.Y.)
- Department of Orthopaedic Surgery, Ichihara Hospital, Tsukuba 300-3295, Ibaraki, Japan
| |
Collapse
|
13
|
Miller LG, Chiok K, Mariasoosai C, Mohanty I, Pandit S, Deol P, Mehari L, Teng MN, Haas AL, Natesan S, Miura TA, Bose S. Extracellular ISG15 triggers ISGylation via a type-I interferon independent non-canonical mechanism to regulate host response during virus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.602290. [PMID: 39026703 PMCID: PMC11257485 DOI: 10.1101/2024.07.05.602290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Type-I interferons (IFN) induce cellular proteins with antiviral activity. One such protein is Interferon Stimulated Gene 15 (ISG15). ISG15 is conjugated to proteins during ISGylation to confer antiviral activity and regulate cellular activities associated with inflammatory and neurodegenerative diseases and cancer. Apart from ISGylation, unconjugated free ISG15 is also released from cells during various conditions, including virus infection. The role of extracellular ISG15 during virus infection was unknown. We show that extracellular ISG15 triggers ISGylation and acts as a soluble antiviral factor to restrict virus infection via an IFN-independent mechanism. Specifically, extracellular ISG15 acts post-translationally to markedly enhance the stability of basal intracellular ISG15 protein levels to support ISGylation. Furthermore, extracellular ISG15 interacts with cell surface integrin (α5β1 integrins) molecules via its RGD-like motif to activate the integrin-FAK (Focal Adhesion Kinase) pathway resulting in IFN-independent ISGylation. Thus, our studies have identified extracellular ISG15 protein as a new soluble antiviral factor that confers IFN-independent non-canonical ISGylation via the integrin-FAK pathway by post-translational stabilization of intracellular ISG15 protein.
Collapse
|
14
|
Gurung SK, Shevde LA, Rao SS. Laminin I mediates resistance to lapatinib in HER2-positive brain metastatic breast cancer cells in vitro. Biochem Biophys Res Commun 2024; 720:150142. [PMID: 38788545 DOI: 10.1016/j.bbrc.2024.150142] [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: 03/15/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024]
Abstract
The role of extracellular matrix (ECM) prevalent in the brain metastatic breast cancer (BMBC) niche in mediating cancer cell growth, survival, and response to therapeutic agents is not well understood. Emerging evidence suggests a vital role of ECM of the primary breast tumor microenvironment (TME) in tumor progression and survival. Possibly, the BMBC cells are also similarly influenced by the ECM of the metastatic niche; therefore, understanding the effect of the metastatic ECM on BMBC cells is imperative. Herein, we assessed the impact of various ECM components (i.e., Tenascin C, Laminin I, Collagen I, Collagen IV, and Fibronectin) on brain metastatic human epidermal growth factor receptor 2 (HER2)-positive and triple negative breast cancer (TNBC) cell lines in vitro. The highly aggressive TNBC cell line was minimally affected by ECM components exhibiting no remarkable changes in viability and morphology. On the contrary, amongst various ECM components tested, the HER2-positive cell line was significantly affected by Laminin I with higher viability and demonstrated a distinct spread morphology. In addition, HER2-positive BMBC cells exhibited resistance to Lapatinib in presence of Laminin I. Mechanistically, Laminin I-induced resistance to Lapatinib was mediated in part by phosphorylation of Erk 1/2 and elevated levels of Vimentin. Laminin I also significantly enhanced the migratory potential and replicative viability of HER2-positive BMBC cells. In sum, our findings show that presence of Laminin I in the TME of BMBC cells imparts resistance to targeted therapeutic agent Lapatinib, while increasing the possibility of its dispersal and clonogenic survival.
Collapse
Affiliation(s)
- Sumiran Kumar Gurung
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487, USA.
| |
Collapse
|
15
|
Ren G, Zhang Y, Liu J, Cheng W, Wu D, Han M, Zeng Y, Zhao X, Hu L, Zeng M, Gurram RK, Hu X, Zhou B, Hou Z, Zhu J, Jin W, Zhong C. Decreased GATA3 levels cause changed mouse cutaneous innate lymphoid cell fate, facilitating hair follicle recycling. Dev Cell 2024; 59:1809-1823.e6. [PMID: 38723629 PMCID: PMC11265981 DOI: 10.1016/j.devcel.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/08/2024] [Accepted: 04/16/2024] [Indexed: 07/25/2024]
Abstract
In mice, skin-resident type 2 innate lymphoid cells (ILC2s) exhibit some ILC3-like characteristics. However, the underlying mechanism remains elusive. Here, we observed lower expression of the ILC2 master regulator GATA3 specifically in cutaneous ILC2s (cILC2s) compared with canonical ILC2s, in line with its functionally divergent role in transcriptional control in cILC2s. Decreased levels of GATA3 enabled the expansion of RORγt fate-mapped (RORγtfm+) cILC2s after postnatal days, displaying certain similarities to ILC3s. Single-cell trajectory analysis showed a sequential promotion of the RORγtfm+ cILC2 divergency by RORγt and GATA3. Notably, during hair follicle recycling, these RORγtfm+ cILC2s accumulated around the hair follicle dermal papilla (DP) region to facilitate the process. Mechanistically, we found that GATA3-mediated integrin α3β1 upregulation on RORγtfm+ cILC2s was required for their positioning around the DP. Overall, our study demonstrates a distinct regulatory role of GATA3 in cILC2s, particularly in promoting the divergence of RORγtfm+ cILC2s to facilitate hair follicle recycling.
Collapse
Affiliation(s)
- Guanqun Ren
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Yime Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Jiamin Liu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Wenwen Cheng
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Di Wu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Mengwei Han
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Yanyu Zeng
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Xingyu Zhao
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Luni Hu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Min Zeng
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Rama Krishna Gurram
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20892, USA
| | - Xiaole Hu
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Bo Zhou
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zhiyuan Hou
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20892, USA
| | - Wenfei Jin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
| | - Chao Zhong
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China.
| |
Collapse
|
16
|
Yurchenco PD, Kulczyk AW. Polymerizing laminins in development, health, and disease. J Biol Chem 2024; 300:107429. [PMID: 38825010 PMCID: PMC11260871 DOI: 10.1016/j.jbc.2024.107429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Polymerizing laminins are multi-domain basement membrane (BM) glycoproteins that self-assemble into cell-anchored planar lattices to establish the initial BM scaffold. Nidogens, collagen-IV and proteoglycans then bind to the scaffold at different domain loci to create a mature BM. The LN domains of adjacent laminins bind to each other to form a polymer node, while the LG domains attach to cytoskeletal-anchoring integrins and dystroglycan, as well as to sulfatides and heparan sulfates. The polymer node, the repeating unit of the polymer scaffold, is organized into a near-symmetrical triskelion. The structure, recently solved by cryo-electron microscopy in combination with AlphaFold2 modeling and biochemical studies, reveals how the LN surface residues interact with each other and how mutations cause failures of self-assembly in an emerging group of diseases, the LN-lamininopathies, that include LAMA2-related dystrophy and Pierson syndrome.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Arkadiusz W Kulczyk
- Department of Biochemistry and Microbiology, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
17
|
Wyle Y, Lu N, Hepfer J, Sayal R, Martinez T, Wang A. The Role of Biophysical Factors in Organ Development: Insights from Current Organoid Models. Bioengineering (Basel) 2024; 11:619. [PMID: 38927855 PMCID: PMC11200479 DOI: 10.3390/bioengineering11060619] [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: 04/17/2024] [Revised: 05/26/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Biophysical factors play a fundamental role in human embryonic development. Traditional in vitro models of organogenesis focused on the biochemical environment and did not consider the effects of mechanical forces on developing tissue. While most human tissue has a Young's modulus in the low kilopascal range, the standard cell culture substrate, plasma-treated polystyrene, has a Young's modulus of 3 gigapascals, making it 10,000-100,000 times stiffer than native tissues. Modern in vitro approaches attempt to recapitulate the biophysical niche of native organs and have yielded more clinically relevant models of human tissues. Since Clevers' conception of intestinal organoids in 2009, the field has expanded rapidly, generating stem-cell derived structures, which are transcriptionally similar to fetal tissues, for nearly every organ system in the human body. For this reason, we conjecture that organoids will make their first clinical impact in fetal regenerative medicine as the structures generated ex vivo will better match native fetal tissues. Moreover, autologously sourced transplanted tissues would be able to grow with the developing embryo in a dynamic, fetal environment. As organoid technologies evolve, the resultant tissues will approach the structure and function of adult human organs and may help bridge the gap between preclinical drug candidates and clinically approved therapeutics. In this review, we discuss roles of tissue stiffness, viscoelasticity, and shear forces in organ formation and disease development, suggesting that these physical parameters should be further integrated into organoid models to improve their physiological relevance and therapeutic applicability. It also points to the mechanotransductive Hippo-YAP/TAZ signaling pathway as a key player in the interplay between extracellular matrix stiffness, cellular mechanics, and biochemical pathways. We conclude by highlighting how frontiers in physics can be applied to biology, for example, how quantum entanglement may be applied to better predict spontaneous DNA mutations. In the future, contemporary physical theories may be leveraged to better understand seemingly stochastic events during organogenesis.
Collapse
Affiliation(s)
- Yofiel Wyle
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
| | - Nathan Lu
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Jason Hepfer
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Rahul Sayal
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Taylor Martinez
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California-Davis, Davis, CA 95616, USA
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA 95817, USA
| |
Collapse
|
18
|
Teixeira Polez R, Huynh N, Pridgeon CS, Valle-Delgado JJ, Harjumäki R, Österberg M. Insights into spheroids formation in cellulose nanofibrils and Matrigel hydrogels using AFM-based techniques. Mater Today Bio 2024; 26:101065. [PMID: 38706731 PMCID: PMC11066555 DOI: 10.1016/j.mtbio.2024.101065] [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: 11/11/2023] [Revised: 03/30/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024] Open
Abstract
The recent FDA decision to eliminate animal testing requirements emphasises the role of cell models, such as spheroids, as regulatory test alternatives for investigations of cellular behaviour, drug responses, and disease modelling. The influence of environment on spheroid formation are incompletely understood, leading to uncertainty in matrix selection for scaffold-based 3D culture. This study uses atomic force microscopy-based techniques to quantify cell adhesion to Matrigel and cellulose nanofibrils (CNF), and cell-cell adhesion forces, and their role in spheroid formation of hepatocellular carcinoma (HepG2) and induced pluripotent stem cells (iPS(IMR90)-4). Results showed different cell behaviour in CNF and Matrigel cultures. Both cell lines formed compact spheroids in CNF but loose cell aggregates in Matrigel. Interestingly, the type of cell adhesion protein, and not the bond strength, appeared to be a key factor in the formation of compact spheroids. The gene expression of E- and N-cadherins, proteins on cell membrane responsible for cell-cell interactions, was increased in CNF culture, leading to formation of compact spheroids while Matrigel culture induced integrin-laminin binding and downregulated E-cadherin expression, resulting in looser cell aggregates. These findings enhance our understanding of cell-biomaterial interactions in 3D cultures and offer insights for improved 3D cell models, culture biomaterials, and applications in drug research.
Collapse
Affiliation(s)
- Roberta Teixeira Polez
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Ngoc Huynh
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Chris S. Pridgeon
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Juan José Valle-Delgado
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Riina Harjumäki
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Monika Österberg
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| |
Collapse
|
19
|
Li X, Lin H, Yu Y, Lu Y, He B, Liu M, Zhuang L, Xu Y, Li W. In Situ Rapid-Formation Sprayable Hydrogels for Challenging Tissue Injury Management. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400310. [PMID: 38298099 DOI: 10.1002/adma.202400310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 01/20/2024] [Indexed: 02/02/2024]
Abstract
Rapid-acting, convenient, and broadly applicable medical materials are in high demand for the treatment of extensive and intricate tissue injuries in extremely medical scarcity environment, such as battlefields, wilderness, and traffic accidents. Conventional biomaterials fail to meet all the high criteria simultaneously for emergency management. Here, a multifunctional hydrogel system capable of rapid gelation and in situ spraying, addressing clinical challenges related to hemostasis, barrier establishment, support, and subsequent therapeutic treatment of irregular, complex, and urgent injured tissues, is designed. This hydrogel can be fast formed in less than 0.5 s under ultraviolet initiation. The precursor maintains an impressively low viscosity of 0.018 Pa s, while the hydrogel demonstrates a storage modulus of 0.65 MPa, achieving the delicate balance between sprayable fluidity and the mechanical strength requirements in practice, allowing flexible customization of the hydrogel system for differentiated handling and treatment of various tissues. Notably, the interactions between the component of this hydrogel and the cell surface protein confer upon its inherently bioactive functionalities such as osteogenesis, anti-inflammation, and angiogenesis. This research endeavors to provide new insights and designs into emergency management and complex tissue injuries treatment.
Collapse
Affiliation(s)
- Xiaolei Li
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Fels Cancer Institute for Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Han Lin
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
| | - Yilin Yu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
| | - Yukun Lu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
| | - Bin He
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
| | - Meng Liu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
| | - Lin Zhuang
- School of Physics, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Provincial Key Laboratory of Photovoltaics Technologies, Sun Yat-sen University, Guangzhou, 510006, P. R. China
| | - Yue Xu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
| | - Weichang Li
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
| |
Collapse
|
20
|
Mesquita FCP, King M, da Costa Lopez PL, Thevasagayampillai S, Gunaratne PH, Hochman-Mendez C. Laminin Alpha 2 Enhances the Protective Effect of Exosomes on Human iPSC-Derived Cardiomyocytes in an In Vitro Ischemia-Reoxygenation Model. Int J Mol Sci 2024; 25:3773. [PMID: 38612582 PMCID: PMC11011704 DOI: 10.3390/ijms25073773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Ischemic heart disease, a leading cause of death worldwide, manifests clinically as myocardial infarction. Contemporary therapies using mesenchymal stromal cells (MSCs) and their derivative (exosomes, EXOs) were developed to decrease the progression of cell damage during ischemic injury. Laminin alpha 2 (LAMA2) is an important extracellular matrix protein of the heart. Here, we generated MSC-derived exosomes cultivated under LAMA2 coating to enhance human-induced pluripotent stem cell (hiPSC)-cardiomyocyte recognition of LAMA2-EXOs, thus, increasing cell protection during ischemia reoxygenation. We mapped the mRNA content of LAMA2 and gelatin-EXOs and identified 798 genes that were differentially expressed, including genes associated with cardiac muscle development and extracellular matrix organization. Cells were treated with LAMA2-EXOs 2 h before a 4 h ischemia period (1% O2, 5% CO2, glucose-free media). LAMA2-EXOs had a two-fold protective effect compared to non-treatment on plasma membrane integrity and the apoptosis activation pathway; after a 1.5 h recovery period (20% O2, 5% CO2, cardiomyocyte-enriched media), cardiomyocytes treated with LAMA2-EXOs showed faster recovery than did the control group. Although EXOs had a protective effect on endothelial cells, there was no LAMA2-enhanced protection on these cells. This is the first report of LAMA2-EXOs used to treat cardiomyocytes that underwent ischemia-reoxygenation injury. Overall, we showed that membrane-specific EXOs may help improve cardiomyocyte survival in treating ischemic cardiovascular disease.
Collapse
Affiliation(s)
- Fernanda C. P. Mesquita
- Department of Regenerative Medicine Research, The Texas Heart Institute, Houston, TX 77030, USA; (F.C.P.M.); (M.K.); (P.L.d.C.L.)
| | - Madelyn King
- Department of Regenerative Medicine Research, The Texas Heart Institute, Houston, TX 77030, USA; (F.C.P.M.); (M.K.); (P.L.d.C.L.)
| | - Patricia Luciana da Costa Lopez
- Department of Regenerative Medicine Research, The Texas Heart Institute, Houston, TX 77030, USA; (F.C.P.M.); (M.K.); (P.L.d.C.L.)
| | | | - Preethi H. Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Camila Hochman-Mendez
- Department of Regenerative Medicine Research, The Texas Heart Institute, Houston, TX 77030, USA; (F.C.P.M.); (M.K.); (P.L.d.C.L.)
| |
Collapse
|
21
|
Yamamoto M, Funada R, Taki R, Shiroishi M. Production and characterization of an Fv-clasp of rheumatoid factor, a low-affinity human autoantibody. J Biochem 2024; 175:387-394. [PMID: 38102734 DOI: 10.1093/jb/mvad112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Rheumatoid factor (RF) is an autoantibody against IgG that affects autoimmune diseases and inhibits the effectiveness of pharmaceuticals and diagnostic agents. Although RFs derived from various germline genes have been identified, little is known about their molecular recognition mechanisms. In this study, the Fv-clasp format was used to prepare YES8c, an RF. We developed an Escherichia coli secretion expression system capable of producing milligram-scale of YES8c Fv-clasp per 1 L of culture. Although YES8c is an autoantibody with very low affinity, the produced Fv-clasp maintained specific binding to IgG. Interestingly, the molecules prepared by E. coli secretion had a higher affinity than those prepared by refolding. In the structure of the YES8c-Fc complex, the N-terminus of the light chain is close to Fc; therefore, it is suggested that the addition of the N-terminal methionine may cause collisions with Fc, resulting in reduced affinity. Our findings suggest that the Fv-clasp, which provides sufficient stability and a high bacterial yield, is a useful format for studying RFs with very low affinity. Furthermore, the Fv-clasp produced from a secretion expression system, which can properly process the N-terminus, would be suitable for analysis of RFs in which the N-terminus may be involved in interactions.
Collapse
Affiliation(s)
- Maruto Yamamoto
- Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| | - Ryoma Funada
- Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| | - Ryota Taki
- Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| | - Mitsunori Shiroishi
- Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| |
Collapse
|
22
|
Filgueiras LA, de Andrade FDCP, Iwao Horita S, Shirsat SD, Achal V, Rai M, Henriques-Pons A, Mendes AN. Analysis of SIKVAV's receptor affinity, pharmacokinetics, and pharmacological characteristics: a matrikine with potent biological function. J Biomol Struct Dyn 2024:1-23. [PMID: 38345036 DOI: 10.1080/07391102.2024.2313709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/27/2024] [Indexed: 03/08/2025]
Abstract
Matrikines are biologically active peptides generated from fragments fragmentation of extracellular matrix components (ECM) that are functionally distinct from the original full-length molecule. The active matricryptic sites can be unmasked by ECM components enzymatic degradation or multimerization, heterotypic binding, adsorption to other molecules, cell-mediated mechanical forces, exposure to reactive oxygen species, ECM denaturation, and others. Laminin α1-derived peptide (SIKVAV) is a bioactive peptide derived from laminin-111 that participates in tumor development, cell proliferation, angiogenesis in various cell types. SIKVAV has also a potential pharmaceutical activity that may be used for tissue regeneration and bioengineering in Alzheimer's disease and muscular dystrophies. In this work, we made computational analyzes of SIKVAV regarding the ADMET panel, that stands for Administration, Distribution, Metabolism, Excretion, and Toxicity. Docking analyzes using the α3β1 and α6β1 integrin receptors were performed to fill in the gaps in the SIKVAV's signaling pathway and coupling tests showed that SIKVAV can interact with both receptors. Moreover, there is no indication of cytotoxicity, mutagenic or carcinogenic activity, skin or oral sensitivity. Our analysis suggests that SIKVAV has a high probability of interacting with peroxisome proliferator-activated receptor-gamma (NR-PPAR-γ), which has anti-inflammatory activity. The results of bioinformatics can help understand the participation of SIKVAV in homeostasis and influence the understanding of how this peptide can act as a biological asset in the control of dystrophies, neurodegenerative diseases, and tissue engineering.
Collapse
Affiliation(s)
- Livia Alves Filgueiras
- Laboratory of Innovation in Science and Technology - LACITEC, Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | | | - Samuel Iwao Horita
- Laboratory of Innovation in Therapies, Education, and Bioproducts - LITEB, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Shubhangi D Shirsat
- Laboratory of Innovation in Therapies, Education, and Bioproducts - LITEB, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Varenyam Achal
- Environmental Engineering Program, Guangdong Technion - Israel Institute of Technology, Shantou, China
- Technion - Israel Institute of Technology, Haifa, Israel
| | - Mahendra Rai
- Department of Biotechnology, SGB Amravati University, Amravati, India
| | - Andrea Henriques-Pons
- Laboratory of Innovation in Therapies, Education, and Bioproducts - LITEB, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Anderson Nogueira Mendes
- Laboratory of Innovation in Science and Technology - LACITEC, Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
23
|
Akter L, Flechsig H, Marchesi A, Franz CM. Observing Dynamic Conformational Changes within the Coiled-Coil Domain of Different Laminin Isoforms Using High-Speed Atomic Force Microscopy. Int J Mol Sci 2024; 25:1951. [PMID: 38396630 PMCID: PMC10888245 DOI: 10.3390/ijms25041951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Laminins are trimeric glycoproteins with important roles in cell-matrix adhesion and tissue organization. The laminin α, ß, and γ-chains have short N-terminal arms, while their C-termini are connected via a triple coiled-coil domain, giving the laminin molecule a well-characterized cross-shaped morphology as a result. The C-terminus of laminin alpha chains contains additional globular laminin G-like (LG) domains with important roles in mediating cell adhesion. Dynamic conformational changes of different laminin domains have been implicated in regulating laminin function, but so far have not been analyzed at the single-molecule level. High-speed atomic force microscopy (HS-AFM) is a unique tool for visualizing such dynamic conformational changes under physiological conditions at sub-second temporal resolution. After optimizing surface immobilization and imaging conditions, we characterized the ultrastructure of laminin-111 and laminin-332 using HS-AFM timelapse imaging. While laminin-111 features a stable S-shaped coiled-coil domain displaying little conformational rearrangement, laminin-332 coiled-coil domains undergo rapid switching between straight and bent conformations around a defined central molecular hinge. Complementing the experimental AFM data with AlphaFold-based coiled-coil structure prediction enabled us to pinpoint the position of the hinge region, as well as to identify potential molecular rearrangement processes permitting hinge flexibility. Coarse-grained molecular dynamics simulations provide further support for a spatially defined kinking mechanism in the laminin-332 coiled-coil domain. Finally, we observed the dynamic rearrangement of the C-terminal LG domains of laminin-111 and laminin-332, switching them between compact and open conformations. Thus, HS-AFM can directly visualize molecular rearrangement processes within different laminin isoforms and provide dynamic structural insight not available from other microscopy techniques.
Collapse
Affiliation(s)
- Lucky Akter
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa 920-1167, Japan; (L.A.); (H.F.); (A.M.)
| | - Holger Flechsig
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa 920-1167, Japan; (L.A.); (H.F.); (A.M.)
| | - Arin Marchesi
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa 920-1167, Japan; (L.A.); (H.F.); (A.M.)
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Via Tronto, 10/A Torrette di Ancona, 60126 Ancona, Italy
| | - Clemens M. Franz
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa 920-1167, Japan; (L.A.); (H.F.); (A.M.)
| |
Collapse
|
24
|
Arase N, Sasaoka Y, Narita J, Kiyohara E, Hashimoto K, Shinzaki S, Nojima S, Takagi J, Fujimoto M. Anti-α6β4 integrin autoantibodies inhibit the binding of laminins to α6β4 integrin in patients with pemphigoid and affect the gastrointestinal tract. J Eur Acad Dermatol Venereol 2024; 38:404-412. [PMID: 37857595 DOI: 10.1111/jdv.19582] [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: 07/27/2023] [Accepted: 09/27/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Anti-α6β4 integrin autoantibodies can be observed in some patients with mucous membrane pemphigoid. We have previously identified anti-α6β4 integrin extracellular domain autoantibodies together with anti-BP180 NC16A antibodies in a patient with DPP-4 inhibitor-induced bullous pemphigoid. However, the significance and impact of anti-α6β4 integrin antibodies are unknown. OBJECTIVES To characterize anti-α6β4 integrin extracellular domain autoantibodies in pemphigoid patients, to determine whether these antibodies inhibit laminin-α6β4 integrin binding and to observe their systemic effects. METHODS Anti-α6β4 integrin autoantibodies were analysed by staining cells expressing the extracellular region of α6β4 integrin with sera from 20 patients with pemphigoid. The anti-α6β4 integrin autoantibodies were characterized using different transfectants. The binding of laminins to α6β4 integrin was studied using cells expressing the activated conformation of α6β4 integrin and the inhibitory effect of the autoantibodies on the binding of laminins to α6β4 integrin was tested. Trends in antibody titres and clinical symptoms were quantified and analysed. RESULTS IgG autoantibodies against the extracellular domain of anti-α6β4 integrin were found in some patients with pemphigoid. Laminin binding to α6β4 integrin was observed in the active conformation of α6β4 integrin, and serum from a patient with a high titre of anti-α6β4 integrin antibodies inhibited the binding of both laminin-511 and laminin-332 to α6β4 integrin. α6β4 integrin is expressed on the basement membrane of both skin and small intestine, and exfoliation was observed in the patient's epidermis and small intestinal epithelium. A reduction in the titre of the anti-α6β4 integrin antibody was associated with improvement in both skin and gastrointestinal symptoms. CONCLUSIONS This study demonstrated the presence of anti-α6β4 integrin extracellular domain-specific autoantibodies in some patients with pemphigoid. In addition, these autoantibodies showed inhibitory activity on α6β4 integrin-laminin binding. Anti-α6β4 integrin antibodies can affect the gastrointestinal tract as well as the skin and oral mucosa.
Collapse
Affiliation(s)
- Noriko Arase
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Sasaoka
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Narita
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eiji Kiyohara
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuhisa Hashimoto
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinichiro Shinzaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Gastroenterology, Hyogo Medical University School of Medicine, Hyogo, Japan
| | - Satoshi Nojima
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory for Cutaneous Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
25
|
Cho Y, Choi Y, Seong H. Nanoscale surface coatings and topographies for neural interfaces. Acta Biomater 2024; 175:55-75. [PMID: 38141934 DOI: 10.1016/j.actbio.2023.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 12/25/2023]
Abstract
With the lack of minimally invasive tools for probing neuronal systems across spatiotemporal scales, understanding the working mechanism of the nervous system and limited assessments available are imperative to prevent or treat neurological disorders. In particular, nanoengineered neural interfaces can provide a solution to this technological barrier. This review covers recent surface engineering approaches, including nanoscale surface coatings, and a range of topographies from the microscale to the nanoscale, primarily focusing on neural-interfaced biosystems. Specifically, the immobilization of bioactive molecules to fertilize the neural cell lineage, topographical engineering to induce mechanotransduction in neural cells, and enhanced cell-chip coupling using three-dimensional structured surfaces are highlighted. Advances in neural interface design will help us understand the nervous system, thereby achieving the effective treatments for neurological disorders. STATEMENT OF SIGNIFICANCE: • This review focuses on designing bioactive neural interface with a nanoscale chemical modification and topographical engineering at multiscale perspective. • Versatile nanoscale surface coatings and topographies for neural interface are summarized. • Recent advances in bioactive materials applicable for neural cell culture, electrophysiological sensing, and neural implants are reviewed.
Collapse
Affiliation(s)
- Younghak Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Yunyoung Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyejeong Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea.
| |
Collapse
|
26
|
Austin TT, Thomas CL, Warren B. Auditory robustness and resilience in the aging auditory system of the desert locust. Neurobiol Aging 2024; 133:39-50. [PMID: 37913625 DOI: 10.1016/j.neurobiolaging.2023.09.009] [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: 07/12/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 11/03/2023]
Abstract
After overexposure to loud music, we experience a decrease in our ability to hear (robustness), which usually recovers (resilience). Here, we exploited the amenable auditory system of the desert locust, Schistocerca gregaria, to measure how robustness and resilience depend on age. We found that gene expression changes are dominated by age as opposed to noise exposure. We measured sound-evoked nerve activity for young and aged locusts directly, after 24 hours and 48 hours after noise exposure. We found that both young and aged locusts recovered their auditory nerve function over 48 hours. We also measured the sound-evoked transduction current in individual auditory neurons, and although the transduction current magnitude recovered in the young locusts after noise exposure, it failed to recover in the aged locusts. A plastic mechanism compensates for the decreased transduction current in aged locusts. We suggest key genes upregulated in young noise-exposed locusts that mediate robustness to noise exposure and find potential candidates responsible for compensatory mechanisms in the auditory neurons of aged noise-exposed locusts.
Collapse
Affiliation(s)
- Thomas T Austin
- Neurogenetics Group, College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Christian L Thomas
- Neurogenetics Group, College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Ben Warren
- Neurogenetics Group, College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK.
| |
Collapse
|
27
|
Thapa BV, Banerjee M, Glimm T, Saini DK, Bhat R. The senescent mesothelial matrix accentuates colonization by ovarian cancer cells. Cell Mol Life Sci 2023; 81:2. [PMID: 38043093 PMCID: PMC10694112 DOI: 10.1007/s00018-023-05017-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 12/05/2023]
Abstract
Ovarian cancer is amongst the most morbid of gynecological malignancies due to its diagnosis at an advanced stage, a transcoelomic mode of metastasis, and rapid transition to chemotherapeutic resistance. Like all other malignancies, the progression of ovarian cancer may be interpreted as an emergent outcome of the conflict between metastasizing cancer cells and the natural defense mounted by microenvironmental barriers to such migration. Here, we asked whether senescence in coelom-lining mesothelia, brought about by drug exposure, affects their interaction with disseminated ovarian cancer cells. We observed that cancer cells adhered faster on senescent human and murine mesothelial monolayers than on non-senescent controls. Time-lapse epifluorescence microscopy showed that mesothelial cells were cleared by a host of cancer cells that surrounded the former, even under sub-confluent conditions. A multiscale computational model predicted that such colocalized mesothelial clearance under sub-confluence requires greater adhesion between cancer cells and senescent mesothelia. Consistent with the prediction, we observed that senescent mesothelia expressed an extracellular matrix with higher levels of fibronectin, laminins and hyaluronan than non-senescent controls. On senescent matrix, cancer cells adhered more efficiently, spread better, and moved faster and persistently, aiding the spread of cancer. Inhibition assays using RGD cyclopeptides suggested the adhesion was predominantly contributed by fibronectin and laminin. These findings led us to propose that the senescence-associated matrisomal phenotype of peritoneal barriers enhances the colonization of invading ovarian cancer cells contributing to the metastatic burden associated with the disease.
Collapse
Affiliation(s)
- Bharat Vivan Thapa
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
- Undergraduate Program, Indian Institute of Science, Bangalore, 560012, India
| | - Mallar Banerjee
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Tilmann Glimm
- Department of Mathematics, Western Washington University, Bellingham, WA, 98229, USA
| | - Deepak K Saini
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India.
| | - Ramray Bhat
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
28
|
Kersten R, Trampert DC, Herta T, Hubers LM, Maillette de Buy Wenniger LJ, Verheij J, van de Graaf SFJ, Beuers U. IgG4-related cholangitis - a mimicker of fibrosing and malignant cholangiopathies. J Hepatol 2023; 79:1502-1523. [PMID: 37598939 DOI: 10.1016/j.jhep.2023.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/24/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
IgG4-related cholangitis (IRC) is the major hepatobiliary manifestation of IgG4-related disease (IgG4-RD), a systemic fibroinflammatory disorder. The pathogenesis of IgG4-RD and IRC is currently viewed as multifactorial, as there is evidence of a genetic predisposition while environmental factors, such as blue-collar work, are major risk factors. Various autoantigens have been described in IgG4-RD, including annexin A11 and laminin 511-E8, proteins which may exert a partially protective function in cholangiocytes by enhancing secretion and barrier function, respectively. For the other recently described autoantigens, galectin-3 and prohibitin 1, a distinct role in cholangiocytes appears less apparent. In relation to these autoantigens, oligoclonal expansions of IgG4+ plasmablasts are present in patients with IRC and disappear upon successful treatment. More recently, specific T-cell subtypes including regulatory T cells, follicular T helper 2 cells, peripheral T helper cells and cytotoxic CD8+ and CD4+ SLAMF7+ T cells have been implicated in the pathogenesis of IgG4-RD. The clinical presentation of IRC often mimics other biliary diseases such as primary sclerosing cholangitis or cholangiocarcinoma, which may lead to inappropriate medical and potentially invalidating surgical interventions. As specific biomarkers are lacking, diagnosis is made according to the HISORt criteria comprising histopathology, imaging, serology, other organ manifestations and response to therapy. Treatment of IRC aims to prevent or alleviate organ damage and to improve symptoms and consists of (i) remission induction, (ii) remission maintenance and (iii) long-term management. Glucocorticosteroids are highly effective for remission induction, after which immunomodulators can be introduced for maintenance of remission as glucocorticosteroid-sparing alternatives. Increased insight into the pathogenesis of IRC will lead to improved diagnosis and novel therapeutic strategies in the future.
Collapse
Affiliation(s)
- Remco Kersten
- Department of Gastroenterology & Hepatology, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - David C Trampert
- Department of Gastroenterology & Hepatology, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Toni Herta
- Department of Gastroenterology & Hepatology, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam University Medical Centers, Amsterdam, the Netherlands; Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Lowiek M Hubers
- Department of Gastroenterology & Hepatology, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | | - Joanne Verheij
- Department of Pathology, Amsterdam University Medical Centers, the Netherlands
| | - Stan F J van de Graaf
- Department of Gastroenterology & Hepatology, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Ulrich Beuers
- Department of Gastroenterology & Hepatology, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| |
Collapse
|
29
|
Kulczyk AW. Artificial intelligence and the analysis of cryo-EM data provide structural insight into the molecular mechanisms underlying LN-lamininopathies. Sci Rep 2023; 13:17825. [PMID: 37857770 PMCID: PMC10587063 DOI: 10.1038/s41598-023-45200-5] [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: 07/31/2023] [Accepted: 10/17/2023] [Indexed: 10/21/2023] Open
Abstract
Laminins (Lm) are major components of basement membranes (BM), which polymerize to form a planar lattice on cell surface. Genetic alternations of Lm affect their oligomerization patterns and lead to failures in BM assembly manifesting in a group of human disorders collectively defined as Lm N-terminal domain lamininopathies (LN-lamininopathies). We have employed a recently determined cryo-EM structure of the Lm polymer node, the basic repeating unit of the Lm lattice, along with structure prediction and modeling to systematically analyze structures of twenty-three pathogenic Lm polymer nodes implicated in human disease. Our analysis provides the detailed mechanistic explanation how Lm mutations lead to failures in Lm polymerization underlining LN-lamininopathies. We propose the new categorization scheme of LN-lamininopathies based on the insight gained from the structural analysis. Our results can help to facilitate rational drug design aiming in the treatment of Lm deficiencies.
Collapse
Affiliation(s)
- Arkadiusz W Kulczyk
- Institute for Quantitative Biomedicine, Rutgers University, 174 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
- Department of Biochemistry & Microbiology, Rutgers University, 75 Lipman Drive, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
30
|
Zhang H, Zhu DS, Zhu J. Family-wide analysis of integrin structures predicted by AlphaFold2. Comput Struct Biotechnol J 2023; 21:4497-4507. [PMID: 37753178 PMCID: PMC10518446 DOI: 10.1016/j.csbj.2023.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/17/2023] [Accepted: 09/17/2023] [Indexed: 09/28/2023] Open
Abstract
Recent advances in protein structure prediction using AlphaFold2, known for its high efficiency and accuracy, have opened new avenues for comprehensive analysis of all structures within a single protein family. In this study, we evaluated the capabilities of AphaFold2 in analyzing integrin structures. Integrins are heterodimeric cell surface receptors composed of a combination of 18 α and 8 β subunits, resulting in a family of 24 different members. Both α and β subunits consist of a large extracellular domain, a short transmembrane domain, and typically, a short cytoplasmic tail. Integrins play a pivotal role in a wide range of cellular functions by recognizing diverse ligands. Despite significant advances in integrin structural studies in recent decades, high-resolution structures have only been determined for a limited subsets of integrin members, thus limiting our understanding of the entire integrin family. Here, we first analyzed the single-chain structures of 18 α and 8 β integrins in the AlphaFold2 protein structure database. We then employed the newly developed AlphaFold2-multimer program to predict the α/β heterodimer structures of all 24 human integrins. The predicted structures show a high level of accuracy for the subdomains of both α and β subunits, offering high-resolution structure insights for all integrin heterodimers. Our comprehensive structural analysis of the entire integrin family unveils a potentially diverse range of conformations among the 24 members, providing a valuable structure database for studies related to integrin structure and function. We further discussed the potential applications and limitations of the AlphaFold2-derived integrin structures.
Collapse
Affiliation(s)
- Heng Zhang
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Daniel S. Zhu
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Jieqing Zhu
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
31
|
Zhang H, Zhu DS, Zhu J. Family-wide analysis of integrin structures predicted by AlphaFold2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539023. [PMID: 37205578 PMCID: PMC10187181 DOI: 10.1101/2023.05.02.539023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Recent advances in protein structure prediction using AlphaFold2, known for its high efficiency and accuracy, have opened new avenues for comprehensive analysis of all structures within a single protein family. In this study, we evaluated the capabilities of AphaFold2 in analyzing integrin structures. Integrins are heterodimeric cell surface receptors composed of a combination of 18 α and 8 β subunits, resulting in a family of 24 different members. Both α and β subunits consist of a large extracellular domain, a short transmembrane domain, and typically, a short cytoplasmic tail. Integrins play a pivotal role in a wide range of cellular functions by recognizing diverse ligands. Despite significant advances in integrin structural studies in recent decades, high-resolution structures have only been determined for a limited subsets of integrin members, thus limiting our understanding of the entire integrin family. Here, we first analyzed the single-chain structures of 18 α and 8 β integrins in the AlphaFold2 protein structure database. We then employed the newly developed AlphaFold2-multimer program to predict the α/β heterodimer structures of all 24 human integrins. The predicted structures show a high level of accuracy for the subdomains of both α and β subunits, offering high-resolution structure insights for all integrin heterodimers. Our comprehensive structural analysis of the entire integrin family unveils a potentially diverse range of conformations among the 24 members, providing a valuable structure database for studies related to integrin structure and function. We further discussed the potential applications and limitations of the AlphaFold2-derived integrin structures.
Collapse
Affiliation(s)
- Heng Zhang
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Daniel S. Zhu
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Jieqing Zhu
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
32
|
Yeh TC, Lin NY, Chiu CY, Hsu TW, Wu HY, Lin HY, Chen CH, Huang MC. TMTC1 promotes invasiveness of ovarian cancer cells through integrins β1 and β4. Cancer Gene Ther 2023; 30:1134-1143. [PMID: 37221403 PMCID: PMC10425284 DOI: 10.1038/s41417-023-00625-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/20/2023] [Accepted: 05/04/2023] [Indexed: 05/25/2023]
Abstract
Ovarian cancer is the most lethal gynecological malignancy and is characterized by peritoneal disseminated metastasis. Although O-mannosyltransferase TMTC1 is highly expressed by ovarian cancer, its pathophysiological role in ovarian cancer remains unclear. Here, immunohistochemistry showed that TMTC1 was overexpressed in ovarian cancer tissues compared with adjacent normal ovarian tissues, and high TMTC1 expression was associated with poor prognosis in patients with ovarian cancer. Silencing TMTC1 reduced ovarian cancer cell viability, migration, and invasion in vitro, as well as suppressed peritoneal tumor growth and metastasis in vivo. Moreover, TMTC1 knockdown reduced cell-laminin adhesion, which was associated with the decreased phosphorylation of FAK at pY397. Conversely, TMTC1 overexpression promoted these malignant properties in ovarian cancer cells. Glycoproteomic analysis and Concanavalin A (ConA) pull-down assays showed that integrins β1 and β4 were novel O-mannosylated protein substrates of TMTC1. Furthermore, TMTC1-mediated cell migration and invasion were significantly reversed by siRNA-mediated knockdown of integrin β1 or β4. Collectively, these results suggest that TMTC1-mediated invasive behaviors are primarily through integrins β1 and β4 and that TMTC1 is a potential therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Ting-Chih Yeh
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Neng-Yu Lin
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chin-Yu Chiu
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Wen Hsu
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yi Wu
- Instrumentation Center, National Taiwan University, Taipei, Taiwan
| | - Hsuan-Yu Lin
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Hau Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan.
| | - Min-Chuan Huang
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
33
|
Maldonado H, Leyton L. CSK-mediated signalling by integrins in cancer. Front Cell Dev Biol 2023; 11:1214787. [PMID: 37519303 PMCID: PMC10382208 DOI: 10.3389/fcell.2023.1214787] [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: 04/30/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Cancer progression and metastasis are processes heavily controlled by the integrin receptor family. Integrins are cell adhesion molecules that constitute the central components of mechanosensing complexes called focal adhesions, which connect the extracellular environment with the cell interior. Focal adhesions act as key players in cancer progression by regulating biological processes, such as cell migration, invasion, proliferation, and survival. Src family kinases (SFKs) can interplay with integrins and their downstream effectors. SFKs also integrate extracellular cues sensed by integrins and growth factor receptors (GFR), transducing them to coordinate metastasis and cell survival in cancer. The non-receptor tyrosine kinase CSK is a well-known SFK member that suppresses SFK activity by phosphorylating its specific negative regulatory loop (C-terminal Y527 residue). Consequently, CSK may play a pivotal role in tumour progression and suppression by inhibiting SFK oncogenic effects in several cancer types. Remarkably, CSK can localise near focal adhesions when SFKs are activated and even interact with focal adhesion components, such as phosphorylated FAK and Paxillin, among others, suggesting that CSK may regulate focal adhesion dynamics and structure. Even though SFK oncogenic signalling has been extensively described before, the specific role of CSK and its crosstalk with integrins in cancer progression, for example, in mechanosensing, remain veiled. Here, we review how CSK, by regulating SFKs, can regulate integrin signalling, and focus on recent discoveries of mechanotransduction. We additionally examine the cross talk of integrins and GFR as well as the membrane availability of these receptors in cancer. We also explore new pharmaceutical approaches to these signalling pathways and analyse them as future therapeutic targets.
Collapse
Affiliation(s)
- Horacio Maldonado
- Receptor Dynamics in Cancer Laboratory, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
34
|
Yu C, Jiang W, Li B, Hu Y, Liu D. The Role of Integrins for Mediating Nanodrugs to Improve Performance in Tumor Diagnosis and Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13111721. [PMID: 37299624 DOI: 10.3390/nano13111721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
Integrins are heterodimeric transmembrane proteins that mediate adhesive connections between cells and their surroundings, including surrounding cells and the extracellular matrix (ECM). They modulate tissue mechanics and regulate intracellular signaling, including cell generation, survival, proliferation, and differentiation, and the up-regulation of integrins in tumor cells has been confirmed to be associated with tumor development, invasion, angiogenesis, metastasis, and therapeutic resistance. Thus, integrins are expected to be an effective target to improve the efficacy of tumor therapy. A variety of integrin-targeting nanodrugs have been developed to improve the distribution and penetration of drugs in tumors, thereby, improving the efficiency of clinical tumor diagnosis and treatment. Herein, we focus on these innovative drug delivery systems and reveal the improved efficacy of integrin-targeting methods in tumor therapy, hoping to provide prospective guidance for the diagnosis and treatment of integrin-targeting tumors.
Collapse
Affiliation(s)
- Chi Yu
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Wei Jiang
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210093, China
| | - Bin Li
- Department of Biochemistry and Molecular Biology, Medical College, Guangxi University of Science and Technology, Liuzhou 545005, China
| | - Yong Hu
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210093, China
| | - Dan Liu
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| |
Collapse
|
35
|
Chen YC, Li Y, Yan CCS, Hsu CP, Cheng PL, Tu HL. DNA tension assays reveal that force-dependent integrin activation regulates neurite outgrowth in primary cortical neurons. BIOMATERIALS ADVANCES 2023; 150:213431. [PMID: 37116456 DOI: 10.1016/j.bioadv.2023.213431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/30/2023]
Abstract
Biomechanical inputs are ubiquitously present in biological systems and are known to regulate various cell functions. In particular, neural cell development is sensitive to mechanical regulation, as these cells reside in one of the softest microenvironments in the body. To fully characterize and comprehend how mechanical force modulates early neuronal processes, we prepared substrates functionalized with DNA probes displaying integrin ligands, including cRGD and laminin, to quantify integrin-mediated molecular tension during neurite initiation in primary cortical neurons. Our live-cell imaging analysis reveals that integrin-mediated tension force is highly dynamic and distributed across the cell body, with the overall tension signal gradually increasing during neurite outgrowth. Notably, we detected a consistent level of mechanical force (amplitude = 4.7-12 piconewtons, pN) for cell integrin-ligand interactions. Further quantifications reveal that neurons exhibit faster cell spreading and neurite outgrowth upon interacting with ligands functionalized with 4.7 pN relative to 12 pN probes. These findings indicate that the magnitude of integrin-mediated mechanical feedback regulates neuronal activity during early neuritogenesis. Additionally, we observed that mechanical tension is correlated with calcium signaling, since inhibiting calcium influx substantially reduced mechanical tension. Thus, our findings support that the magnitude of integrin-mediated mechanical feedback regulates neuronal activity during early neuritogenesis and that mechanical force is an essential element complementing well-known biochemical regulatory mechanisms orchestrating the integrin activation machinery and controlled neurite outgrowth in cortical neurons.
Collapse
Affiliation(s)
- Ying-Chi Chen
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Ying Li
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan
| | | | - Chao-Ping Hsu
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan; Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei 10617, Taiwan
| | - Pei-Lin Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan.
| | - Hsiung-Lin Tu
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan; Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
36
|
Tang T, Liu Y, Wang P, Xiang Y, Liu L, Xiao S, Wang G. Carbon quantum dots as a nitric oxide donor can promote wound healing of deep partial-thickness burns in rats. Eur J Pharm Sci 2023; 183:106394. [PMID: 36740102 DOI: 10.1016/j.ejps.2023.106394] [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: 07/24/2022] [Revised: 01/01/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
INTRODUCTION In this study, a new carbon quantum dots-NO (CQDs-NO) that is based on spermidine trihydrochloride and can be used as a nitric oxide donor was prepared using a two-step hyperthermia-intermittent ultrasonic method, after which its healing effect on deep partial-thickness burn wounds was tested in rats. MATERIALS AND METHODS CQDs-NO were prepared by a two-step hyperthermia-intermittent ultrasonic method. NO-released rate and biocompatibility of CQDs-NO were tested. The biological functions of CQDs-NO were measured by scratch assay, Western blotting, histology, and transcriptome sequencing. RESULTS CQDs-NO with a concentration of 1 μg/mL and 5 μg/mL showed no cytotoxicity. CQDs-NO could release NO when co-cultured with cells or glutathione peroxidase. We also found that CQDs-NO promotes the biological processes such as angiogenesis, cell-substrate adhesion, extracellular matrix organization, cell migration, and wound healing in human umbilical vein endothelial cells (HUVEC). Additionally, CQDs-NO promoted wound healing of deep partial-thickness burn by enhancing vascularization, matrix deposition, as well as regulating the inflammatory reactions of wounds. CONCLUSIONS CQDs-NO could be used as an alternative method for deep partial-thickness burn healing.
Collapse
Affiliation(s)
- Tao Tang
- Burn Institute of PLA, Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, China
| | - Yingying Liu
- Burn Institute of PLA, Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, China
| | - Peng Wang
- Department of Burns and Plastic Surgery Linfen Central Hospital, Linfen, Shanxi, 041000, China
| | - Yang Xiang
- Burn Institute of PLA, Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, China
| | - Lei Liu
- Department of Plastic Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Shichu Xiao
- Burn Institute of PLA, Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, China.
| | - Guangyi Wang
- Burn Institute of PLA, Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, Chinese Academy of Medical Sciences, Shanghai, 200433, China.
| |
Collapse
|
37
|
Du F, Shusta EV, Palecek SP. Extracellular matrix proteins in construction and function of in vitro blood-brain barrier models. FRONTIERS IN CHEMICAL ENGINEERING 2023. [DOI: 10.3389/fceng.2023.1130127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
The blood-brain barrier (BBB) is a highly impermeable barrier separating circulating blood and brain tissue. A functional BBB is critical for brain health, and BBB dysfunction has been linked to the pathophysiology of diseases such as stroke and Alzheimer’s disease. A variety of models have been developed to study the formation and maintenance of the BBB, ranging from in vivo animal models to in vitro models consisting of primary cells or cells differentiated from human pluripotent stem cells (hPSCs). These models must consider the composition and source of the cellular components of the neurovascular unit (NVU), including brain microvascular endothelial cells (BMECs), brain pericytes, astrocytes, and neurons, and how these cell types interact. In addition, the non-cellular components of the BBB microenvironment, such as the brain vascular basement membrane (BM) that is in direct contact with the NVU, also play key roles in BBB function. Here, we review how extracellular matrix (ECM) proteins in the brain vascular BM affect the BBB, with a particular focus on studies using hPSC-derived in vitro BBB models, and discuss how future studies are needed to advance our understanding of how the ECM affects BBB models to improve model performance and expand our knowledge on the formation and maintenance of the BBB.
Collapse
|
38
|
Tan M, Ge Y, Wang X, Wang Y, Liu Y, He F, Teng H. Extracellular Vesicles (EVs) in Tumor Diagnosis and Therapy. Technol Cancer Res Treat 2023; 22:15330338231171463. [PMID: 37122245 PMCID: PMC10134167 DOI: 10.1177/15330338231171463] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
In recent years, extracellular vesicles (EVs) have gained significant attention due to their tremendous potential for clinical applications. EVs play a crucial role in various aspects, including tumorigenesis, drug resistance, immune escape, and reconstruction of the tumor microenvironment. Despite the growing interest in EVs, many questions still need to be addressed before they can be practically applied in clinical settings. This paper aims to review EVs' isolation methods, structure research, the roles of EVs in tumorigenesis and their mechanisms in multiple types of tumors, their potential application in drug delivery, and the expectations for their future in clinical research.
Collapse
Affiliation(s)
- Mingdian Tan
- School of Medicine, Asian Liver Center, Stanford, CA, USA
| | - Yizhi Ge
- The Affiliated Cancer Hospital of Nanjing Medical University (Jiangsu Cancer Hospital) and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xiaogang Wang
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yan Wang
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford University School of Medicine, Stanford, CA, USA
| | - Yi Liu
- School of Medicine, Asian Liver Center, Stanford, CA, USA
| | - Feng He
- Stanford University School of Medicine, Stanford, CA, USA
| | - Hongqi Teng
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
39
|
What does it take to be a collagen receptor? Matrix Biol 2023; 115:128-132. [PMID: 36574820 DOI: 10.1016/j.matbio.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
|
40
|
Aleman J, Young CD, Karam SD, Wang XJ. Revisiting laminin and extracellular matrix remodeling in metastatic squamous cell carcinoma: What have we learned after more than four decades of research? Mol Carcinog 2023; 62:5-23. [PMID: 35596706 PMCID: PMC9676410 DOI: 10.1002/mc.23417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023]
Abstract
Patients with squamous cell carcinoma (SCC) have significantly lower survival upon the development of distant metastases. The extracellular matrix (ECM) is a consistent yet dynamic influence on the metastatic capacity of SCCs. The ECM encompasses a milieu of structural proteins, signaling molecules, and enzymes. Just over 40 years ago, the fibrous ECM glycoprotein laminin was identified. Roughly four decades of research have revealed a pivotal role of laminins in metastasis. However, trends in ECM alterations in some cancers have been applied broadly to all metastatic diseases, despite evidence that these characteristics vary by tumor type. We will summarize how laminins influence the SCC metastatic process exclusively. Enhanced laminin protein deposition occurs at the invasive edge of SCC tumors, which correlates with elevated levels of laminin-binding β1 integrins on SCC cells, increased MMP-3 presence, worse prognosis, and lymphatic dissemination. Although these findings are significant, gaps in knowledge of the formation of a premetastatic niche, the processes of intra- and extravasation, and the contributions of the ECM to SCC metastatic cell dormancy persist. Bridging these gaps requires novel in vitro systems and animal models that reproduce tumor-stromal interactions and spontaneous metastasis seen in the clinic. These advances will allow accurate assessment of laminins to predict responders to transforming growth factor-β inhibitors and immunotherapy, as well as potential combinatorial therapies with the standard of care. Such clinical interventions may drastically improve quality of life and patient survival by explicitly targeting SCC metastasis.
Collapse
Affiliation(s)
- John Aleman
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christian D. Young
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sana D. Karam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
- Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, Colorado, USA
| |
Collapse
|
41
|
Ngalim SH, Yusoff N, Johnson RR, Abdul Razak SR, Chen X, Hobbs JK, Lee YY. A review on mechanobiology of cell adhesion networks in different stages of sporadic colorectal cancer to explain its tumorigenesis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:63-72. [PMID: 36116549 DOI: 10.1016/j.pbiomolbio.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 06/15/2023]
Abstract
Sporadic colorectal cancer (CRC) is strongly linked to extraneous factors, like poor diet and lifestyle, but not to inherent factors like familial genetics. The changes at the epigenomics and signalling pathways are known across the sporadic CRC stages. The catch is that temporal information of the onset, the feedback loop, and the crosstalk of signalling and noise are still unclear. This makes it challenging to diagnose and treat colon cancer effectively with no relapse. Various microbial cells and native cells of the colon, contribute to sporadic CRC development. These cells secrete autocrine and paracrine for their bioenergetics and communications with other cell types. Imbalances of the biochemicals affect the epithelial lining of colon. One side of this epithelial lining is interfacing the dense colon tissue, while the other side is exposed to microbiota and excrement from the lumen. Hence, the epithelial lining is prone to tumorigenesis due to the influence of both biochemical and mechanical cues from its complex surrounding. The role of physical transformations in tumorigenesis have been limitedly discussed. In this context, cellular and tissue structures, and force transductions are heavily regulated by cell adhesion networks. These networks include cell anchoring mechanism to the surrounding, cell structural integrity mechanism, and cell effector molecules. This review will focus on the progression of the sporadic CRC stages that are governed by the underlaying cell adhesion networks within the epithelial cells. Additionally, current and potential technologies and therapeutics that target cell adhesion networks for treatments of sporadic CRC will be incorporated.
Collapse
Affiliation(s)
- Siti Hawa Ngalim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM) Bertam, 13200 Kepala Batas, Penang, Malaysia.
| | - Norwahida Yusoff
- School of Mechanical Engineering, Universiti Sains Malaysia (USM) Engineering Campus, 14300 Nibong Tebal, Penang, Malaysia
| | - Rayzel Renitha Johnson
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM) Bertam, 13200 Kepala Batas, Penang, Malaysia
| | - Siti Razila Abdul Razak
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM) Bertam, 13200 Kepala Batas, Penang, Malaysia
| | - Xinyue Chen
- Department of Physics and Astronomy, University of Sheffield, Hounsfield Road, Sheffield, S3 7RH, United Kingdom
| | - Jamie K Hobbs
- Department of Physics and Astronomy, University of Sheffield, Hounsfield Road, Sheffield, S3 7RH, United Kingdom
| | - Yeong Yeh Lee
- School of Medical Sciences, Universiti Sains Malaysia (USM) Kubang Kerian, 16150 Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
42
|
Nirwane A, Yao Y. Cell-specific expression and function of laminin at the neurovascular unit. J Cereb Blood Flow Metab 2022; 42:1979-1999. [PMID: 35796497 PMCID: PMC9580165 DOI: 10.1177/0271678x221113027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/08/2022] [Accepted: 06/19/2022] [Indexed: 11/17/2022]
Abstract
Laminin, a major component of the basal lamina (BL), is a heterotrimeric protein with many isoforms. In the CNS, laminin is expressed by almost all cell types, yet different cells synthesize distinct laminin isoforms. By binding to its receptors, laminin exerts a wide variety of important functions. However, due to the reciprocal and cell-specific expression of laminin in different cells at the neurovascular unit, its functions in blood-brain barrier (BBB) maintenance and BBB repair after injury are not fully understood. In this review, we focus on the expression and functions of laminin and its receptors in the neurovascular unit under both physiological and pathological conditions. We first briefly introduce the structures of laminin and its receptors. Next, the expression and functions of laminin and its receptors in the CNS are summarized in a cell-specific manner. Finally, we identify the knowledge gap in the field and discuss key questions that need to be answered in the future. Our goal is to provide a comprehensive overview on cell-specific expression of laminin and its receptors in the CNS and their functions on BBB integrity.
Collapse
Affiliation(s)
- Abhijit Nirwane
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
43
|
Zhao X, Liu C, He X, Wang M, Zhang H, Cheng J, Wang H. Laminin-bound integrin α6β4 promotes non-small cell lung cancer progression via the activation of YAP/TAZ signaling pathway. Front Oncol 2022; 12:1015709. [PMID: 36276068 PMCID: PMC9583390 DOI: 10.3389/fonc.2022.1015709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/23/2022] [Indexed: 11/26/2022] Open
Abstract
Laminin is an extracellular matrix multidomain trimeric glycoprotein, that has a potential role in tumor progression. Here, we studied the effects of non-small cell lung cancer (NSCLC) cells interaction on laminin and explored the underlying mechanism of laminin associated NSCLC progression. Culture of A549 and NCI-1299 cells on 2D collagen gels (containing laminin) significantly promoted the proliferative and tumorigenic characteristics, as well as cell invasion of tumor cells in vitro. Consistently, comparing the clinical NSCLC tumor tissues, a poor overall survival was observed in patients with high laminin expression. Mechanistically, the expression of integrin α6β4 was required for the pro-tumor effects of laminin. Meanwhile, we showed that the downstream signaling of integrin α6β4, involved the focal adhesion kinase (FAK)/Yes-Associated Protein (YAP)/TAZ signaling pathway. The activation of FAK/YAP/TAZ signaling pathway induced by laminin was validated in tumor tissues from NSCLC patients. Suppression of integrin α6β4/FAK/YAP/TAZ signaling pathway efficiently suppressed the laminin-induced tumor growth, and strengthened the anticancer effects of chemotherapy, describing a novel target for NSCLC treatment.
Collapse
Affiliation(s)
- Xiaopeng Zhao
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chuang Liu
- Department of Thoracic Surgery, The Fourth Central Hospital of Baoding City, Baoding, China
| | - Xu He
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Miao Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haoran Zhang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingge Cheng
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongyan Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Hongyan Wang,
| |
Collapse
|
44
|
Takada YK, Fujita M, Takada Y. Pro-Inflammatory Chemokines CCL5, CXCL12, and CX3CL1 Bind to and Activate Platelet Integrin αIIbβ3 in an Allosteric Manner. Cells 2022; 11:cells11193059. [PMID: 36231020 PMCID: PMC9563052 DOI: 10.3390/cells11193059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/16/2022] Open
Abstract
Activation of platelet integrin αIIbβ3, a key event for hemostasis and thrombus formation, is known to be mediated exclusively by inside-out signaling. We showed that inflammatory chemokines CX3CL1 and CXCL12 in previous studies, and CCL5 in this study, bound to the allosteric binding site (site 2) of vascular integrin αvβ3, in addition to the classical ligand binding site (site 1), and allosterically activated integrins independent of inside-out signaling. Since αIIbβ3 is exposed to inflammatory chemokines at increased concentrations during inflammation (e.g., cytokine/chemokine storm) and platelet activation, we hypothesized that these chemokines bind to and activate αIIbβ3 in an allosteric activation mechanism. We found that these chemokines bound to αIIbβ3. Notably, they activated soluble αIIbβ3 in 1 mM Ca2+ by binding to site 2. They activated cell-surface αIIbβ3 on CHO cells, which lack machinery for inside-out signaling or chemokine receptors, quickly (<1 min) and at low concentrations (1–10 ng/mL) compared to activation of soluble αIIbβ3, probably because chemokines bind to cell surface proteoglycans. Furthermore, activation of αIIbβ3 by the chemokines was several times more potent than 1 mM Mn2+. We propose that CCL5 and CXCL12 (stored in platelet granules) may allosterically activate αIIbβ3 upon platelet activation and trigger platelet aggregation. Transmembrane CX3CL1 on activated endothelial cells may mediate platelet–endothelial interaction by binding to and activating αIIbβ3. Additionally, these chemokines in circulation over-produced during inflammation may trigger αIIbβ3 activation, which is a possible missing link between inflammation and thrombosis.
Collapse
Affiliation(s)
- Yoko K. Takada
- Department of Dermatology, School of Medicine, University of California–Davis, 4645 Second Ave., Research III Suite 3300, Sacramento, CA 95817, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Masaaki Fujita
- Department of Dermatology, School of Medicine, University of California–Davis, 4645 Second Ave., Research III Suite 3300, Sacramento, CA 95817, USA
| | - Yoshikazu Takada
- Department of Dermatology, School of Medicine, University of California–Davis, 4645 Second Ave., Research III Suite 3300, Sacramento, CA 95817, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Correspondence:
| |
Collapse
|
45
|
Lin FY, Li J, Xie Y, Zhu J, Huong Nguyen TT, Zhang Y, Zhu J, Springer TA. A general chemical principle for creating closure-stabilizing integrin inhibitors. Cell 2022; 185:3533-3550.e27. [PMID: 36113427 PMCID: PMC9494814 DOI: 10.1016/j.cell.2022.08.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/07/2022] [Accepted: 08/04/2022] [Indexed: 01/26/2023]
Abstract
Integrins are validated drug targets with six approved therapeutics. However, small-molecule inhibitors to three integrins failed in late-stage clinical trials for chronic indications. Such unfavorable outcomes may in part be caused by partial agonism, i.e., the stabilization of the high-affinity, extended-open integrin conformation. Here, we show that the failed, small-molecule inhibitors of integrins αIIbβ3 and α4β1 stabilize the high-affinity conformation. Furthermore, we discovered a simple chemical feature present in multiple αIIbβ3 antagonists that stabilizes integrins in their bent-closed conformation. Closing inhibitors contain a polar nitrogen atom that stabilizes, via hydrogen bonds, a water molecule that intervenes between a serine residue and the metal in the metal-ion-dependent adhesion site (MIDAS). Expulsion of this water is a requisite for transition to the open conformation. This change in metal coordination is general to integrins, suggesting broad applicability of the drug-design principle to the integrin family, as validated with a distantly related integrin, α4β1.
Collapse
Affiliation(s)
- Fu-Yang Lin
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Li
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yonghua Xie
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, PRC
| | - Jianghai Zhu
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Thi Thu Huong Nguyen
- Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yonghui Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, PRC.
| | - Jieqing Zhu
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Timothy A Springer
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
46
|
Hu X, Roy SR, Jin C, Li G, Zhang Q, Asano N, Asahina S, Kajiwara T, Takahara A, Feng B, Aoki K, Xu C, Zhang Y. Control cell migration by engineering integrin ligand assembly. Nat Commun 2022; 13:5002. [PMID: 36008449 PMCID: PMC9411606 DOI: 10.1038/s41467-022-32686-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
Advances in mechanistic understanding of integrin-mediated adhesion highlight the importance of precise control of ligand presentation in directing cell migration. Top-down nanopatterning limited the spatial presentation to sub-micron placing restrictions on both fundamental study and biomedical applications. To break the constraint, here we propose a bottom-up nanofabrication strategy to enhance the spatial resolution to the molecular level using simple formulation that is applicable as treatment agent. Via self-assembly and co-assembly, precise control of ligand presentation is succeeded by varying the proportions of assembling ligand and nonfunctional peptide. Assembled nanofilaments fulfill multi-functions exerting enhancement to suppression effect on cell migration with tunable amplitudes. Self-assembled nanofilaments possessing by far the highest ligand density prevent integrin/actin disassembly at cell rear, which expands the perspective of ligand-density-dependent-modulation, revealing valuable inputs to therapeutic innovations in tumor metastasis.
Collapse
Affiliation(s)
- Xunwu Hu
- Active Soft Matter Group, CAS Songshan Lake Materials Laboratory, Dongguan, China
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Sona Rani Roy
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Chengzhi Jin
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Guangzhou Municipal and Guangdong Provincial Key of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guanying Li
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qizheng Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | | | | | - Tomoko Kajiwara
- Research Center for Negative Emission Technology, Kyushu University, Fukuoka, Japan
| | - Atsushi Takahara
- Research Center for Negative Emission Technology, Kyushu University, Fukuoka, Japan
| | - Bolu Feng
- Fluid Mechanics Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Kazuhiro Aoki
- Division of Quantitative Biology, National Institute for Basic Biology, National Institute of Natural Sciences, Aichi, Japan
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi, Japan
- Department of Basic Biology, School of Science, SOKENDAI (The Graduate University for Advanced Studies), Aichi, Japan
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Ye Zhang
- Active Soft Matter Group, CAS Songshan Lake Materials Laboratory, Dongguan, China.
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| |
Collapse
|
47
|
Nakashima Y, Tsukahara M. Laminin-511 activates the human induced pluripotent stem cell survival via α6β1 integrin-Fyn-RhoA-ROCK signaling. Stem Cells Dev 2022; 31:706-719. [PMID: 35726387 DOI: 10.1089/scd.2022.0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
In human induced pluripotent stem cells (hiPSCs), laminin-511/α6β1 integrin interacts with E-cadherin, an intercellular adhesion molecule, to induce the activation of the PI3K-dependent signaling pathway. The interaction between laminin-511/α6β1 integrin and E-cadherin, an intercellular adhesion molecule, results in protection against apoptosis via the proto-oncogene tyrosine-protein kinase Fyn(Fyn)-RhoA-ROCK signaling pathway and the Ras homolog gene family member A (RhoA)/Rho kinase (ROCK) signaling pathway (the major pathway for cell death). In this paper, the impact of laminin-511 on hiPSC on α6β1 integrin-Fyn-RhoA-ROCK signaling is discussed and explored along with validation experiments. PIK3CA mRNA (mean [standard deviation {SD}]: iMatrix-511, 1.00 [0. 61]; collagen+MFGE8, 0.023 [0.02]; **P<0.01; n=6) and PIK3R1 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 79]; collagen+MFGE8, 0.040 [0.06]; *P<0.05; n=6) were upregulated by iMatrix-511 resulting from an increased expression of Integrin α6 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 42]; collagen+MFGE8, 0.23 [0.05]; **P<0.01; n=6). iMatrix-511 increased the expression of p120-Catenin mRNA (mean [SD]: iMatrix-511, 1.00 [0. 71]; collagen+MFGE8, 0.025 [0.03]; **P<0.01; n=6) and RAC1 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 28]; collagen+MFGE8, 0.39 [0.15]; **P<0.01; n=6) by increasing the expression of E-cadherin mRNA (mean [SD]: iMatrix-511, 1.00 [0. 38]; collagen+MFGE8, 0.16 [0.11]; **P<0.01; n=6). As a result, iMatrix-511 increased the expression of P190 RhoGAP (GTPase-activating proteins) mRNA, such as ARHGAP1 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 57]; collagen+MFGE8, 0.032 [0.03]; **P<0.01; n=6), ARHGAP4 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 56]; collagen+MFGE8, 0.039 [0.049]; **P<0.01; n=6), and ARHGAP5 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 39]; collagen+MFGE8, 0.063 [0.043]; **P<0.01; n=6). Western blotting showed that phospho-Rac1 remained in the cytoplasm and phospho-Fyn showed nuclear transition in iPSCs cultured on iMatrix-511. Proteome analysis showed that PI3K signaling was enhanced and cytoskeletal actin was activated in iPSCs cultured on iMatrix-511. In conclusion, laminin-511 strongly activated the cell survival by promoting α6β1 integrin-Fyn-RhoA-ROCK signaling in hiPSCs.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto Research Park KISTIC Building Room 501, 5th floor, KISTIC building,, Kyoto Research Park KISTIC Building Room 501, 5th floor, KISTIC building, 134 Chudoji Minami-cho,, Shimogyo-ku,, Kyoto, Kyoto, Japan, 600-8813;
| | - Masayoshi Tsukahara
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), kyoto, Kyoto, Japan;
| |
Collapse
|
48
|
Superoxide Dismutase-3 Downregulates Laminin α5 Expression in Tumor Endothelial Cells via the Inhibition of Nuclear Factor Kappa B Signaling. Cancers (Basel) 2022; 14:cancers14051226. [PMID: 35267534 PMCID: PMC8909228 DOI: 10.3390/cancers14051226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 02/01/2023] Open
Abstract
The balance between laminin isoforms containing the α5 or the α4 chain in the endothelial basement membrane determines the site of leukocyte diapedesis under inflammatory conditions. Extracellular superoxide dismutase (SOD3) induces laminin α4 expression in tumor blood vessels, which is associated with enhanced intratumor T cell infiltration in primary human cancers. We show now that SOD3 overexpression in neoplastic and endothelial cells (ECs) reduces laminin α5 in tumor blood vessels. SOD3 represses the laminin α5 gene (LAMA5), but LAMA5 expression is not changed in SOD1-overexpressing cells. Transcriptomic analyses revealed SOD3 overexpression to change the transcription of 1682 genes in ECs, with the canonical and non-canonical NF-κB pathways as the major SOD3 targets. Indeed, SOD3 reduced the transcription of well-known NF-κB target genes as well as NF-κB-driven promoter activity in ECs stimulated with tumor necrosis factor (TNF)-α, an NF-κB signaling inducer. SOD3 inhibited the phosphorylation and degradation of IκBα (nuclear factor of the kappa light polypeptide gene enhancer in B-cells inhibitor alpha), an NF-κB inhibitor. Finally, TNF-α was found to be a transcriptional activator of LAMA5 but not of LAMA4; LAMA5 induction was prevented by SOD3. In conclusion, SOD3 is a major regulator of laminin balance in the basement membrane of tumor ECs, with potential implications for immune cell infiltration into tumors.
Collapse
|
49
|
Alhalhooly L, Confeld MI, Woo SO, Mamnoon B, Jacobson R, Ghosh S, Kim J, Mallik S, Choi Y. Single-Molecule Force Probing of RGD-Binding Integrins on Pancreatic Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:7671-7679. [PMID: 35113515 PMCID: PMC8890904 DOI: 10.1021/acsami.1c23361] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Integrin-targeting arginine-glycine-aspartic acid (RGD)-based nanocarriers have been widely used for tumor imaging, monitoring of tumor development, and delivery of anticancer drugs. However, the thermodynamics of an RGD-integrin formation and dissociation associated with binding dynamics, affinity, and stability remains unclear. Here, we probed the binding strength of the binary complex to live pancreatic cancer cells using single-molecule binding force spectroscopy methods, in which RGD peptides were functionalized on a force probe tip through poly(ethylene glycol) (PEG)-based bifunctional linker molecules. While the density of integrin αV receptors on the cell surface varies more than twofold from cell line to cell line, the individual RGD-integrin complexes exhibited a cell type-independent, monovalent bond strength. The load-dependent bond strength of multivalent RGD-integrin interactions scaled sublinearly with increasing bond number, consistent with the noncooperative, parallel bond model. Furthermore, the multivalent bonds ruptured sequentially either by one or in multiples, and the force strength was comparable to the synchronous rupture force. Comparison of energy landscapes of the bond number revealed a substantial decrease of kinetic off-rates for multivalent bonds, along with the increased width of the potential well and the increased potential barrier height between bound and unbound states, enhancing the stability of the multivalent bonds between them.
Collapse
Affiliation(s)
- Lina Alhalhooly
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Matthew I. Confeld
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Sung Oh Woo
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Babak Mamnoon
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Reed Jacobson
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Shrinwanti Ghosh
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Jiha Kim
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Yongki Choi
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, North Dakota 58108, United State
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United State
| |
Collapse
|
50
|
Anderson JM, Li J, Springer TA. Regulation by metal ions and the ADMIDAS of integrin α5β1 conformational states and intrinsic affinities. Mol Biol Cell 2022; 33:ar56. [PMID: 35108026 PMCID: PMC9265148 DOI: 10.1091/mbc.e21-11-0536] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Activation of integrins by Mn2+ is a benchmark in the integrin field, but how Mn2+ works and whether it reproduces physiological activation is unknown. We show that Mn2+ and high Mg2+ concentrations compete with Ca2+ at the ADMIDAS and shift the conformational equilibrium toward the open state, but the shift is far from complete. Additionally, replacement of Mg2+ by Mn2+ at the MIDAS increases the intrinsic affinities of both the high-affinity open and low-affinity closed states of integrins, in agreement with stronger binding of Mn2+ than Mg2+ to oxygen atoms. Mutation of the ADMIDAS increases the affinity of closed states and decreases the affinity of the open state and thus reduces the difference in affinity between the open and closed states. An important biological function of the ADMIDAS may be to stabilize integrins in highly discrete states, so that when integrins support cell adhesion and migration, their high and low affinity correspond to discrete on and off states, respectively.
Collapse
Affiliation(s)
- Jordan M Anderson
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston MA 02115.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA 02115
| | - Jing Li
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston MA 02115.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA 02115
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston MA 02115.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA 02115
| |
Collapse
|