1
|
Burboa PC, Corrêa-Velloso JC, Arriagada C, Thomas AP, Durán WN, Lillo MA. Impact of Matrix Gel Variations on Primary Culture of Microvascular Endothelial Cell Function. Microcirculation 2024; 31:e12859. [PMID: 38818977 PMCID: PMC11227414 DOI: 10.1111/micc.12859] [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: 08/31/2023] [Revised: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE The endothelium regulates crucial aspects of vascular function, including hemostasis, vasomotor tone, proliferation, immune cell adhesion, and microvascular permeability. Endothelial cells (ECs), especially in arterioles, are pivotal for flow distribution and peripheral resistance regulation. Investigating vascular endothelium physiology, particularly in microvascular ECs, demands precise isolation and culturing techniques. METHODS Freshly isolated ECs are vital for examining protein expression, ion channel behavior, and calcium dynamics. Establishing primary endothelial cell cultures is crucial for unraveling vascular functions and understanding intact microvessel endothelium roles. Despite the significance, detailed protocols and comparisons with intact vessels are scarce in microvascular research. We developed a reproducible method to isolate microvascular ECs, assessing substrate influence by cultivating cells on fibronectin and gelatin matrix gels. This comparative approach enhances our understanding of microvascular endothelial cell biology. RESULTS Microvascular mesenteric ECs expressed key markers (VE-cadherin and eNOS) in both matrix gels, confirming cell culture purity. Under uncoated conditions, ECs were undetected, whereas proteins linked to smooth muscle cells and fibroblasts were evident. Examining endothelial cell (EC) physiological dynamics on distinct matrix substrates revealed comparable cell length, shape, and Ca2+ elevations in both male and female ECs on gelatin and fibronectin matrix gels. Gelatin-cultured ECs exhibited analogous membrane potential responses to acetylcholine (ACh) or adenosine triphosphate (ATP), contrasting with their fibronectin-cultured counterparts. In the absence of stimulation, fibronectin-cultured ECs displayed a more depolarized resting membrane potential than gelatin-cultured ECs. CONCLUSIONS Gelatin-cultured ECs demonstrated electrical behaviors akin to intact endothelium from mouse mesenteric arteries, thus advancing our understanding of endothelial cell behavior within diverse microenvironments.
Collapse
Affiliation(s)
- Pía C. Burboa
- Department of Pharmacology; Physiology & Neuroscience; New Jersey Medical School; Rutgers, The State University of New Jersey, Newark, NJ 07103, U.S.A
| | - Juliana C. Corrêa-Velloso
- Department of Pharmacology; Physiology & Neuroscience; New Jersey Medical School; Rutgers, The State University of New Jersey, Newark, NJ 07103, U.S.A
| | - Cecilia Arriagada
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Campus Los Leones, Lota 2465, Providencia, Santiago, Chile
| | - Andrew P. Thomas
- Department of Pharmacology; Physiology & Neuroscience; New Jersey Medical School; Rutgers, The State University of New Jersey, Newark, NJ 07103, U.S.A
| | - Walter N. Durán
- Department of Pharmacology; Physiology & Neuroscience; New Jersey Medical School; Rutgers, The State University of New Jersey, Newark, NJ 07103, U.S.A
| | - Mauricio A. Lillo
- Department of Pharmacology; Physiology & Neuroscience; New Jersey Medical School; Rutgers, The State University of New Jersey, Newark, NJ 07103, U.S.A
| |
Collapse
|
2
|
Cai X, Zhu J, Luo X, Jin G, Huang Y, Li L. A Thermally Stable Recombinant Human Fibronectin Peptide-Fused Protein (rhFN3C) for Faster Aphthous Ulcer (AU) Healing. Bioengineering (Basel) 2023; 11:38. [PMID: 38247915 PMCID: PMC10813363 DOI: 10.3390/bioengineering11010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Approximately 59.4-100% of head and neck cancer patients receiving radiotherapy or radio chemotherapy suffer from aphthous ulcers (AUs), which seriously affect the subsequent treatment. At the same time, AUs are a common oral mucosal disease with a high incidence rate among the population, often accompanied by severe pain, and affect both physical and mental health. Strategies to increase the ulcer healing rate and relieve pain symptoms quickly is a long-term clinical objective. Oral mucosal discontinuity is the main histological hallmark of AUs. So, covering the inner mucosal defect with an in vitro engineered oral mucosal equivalent shows good prospects for AU alleviation. Fibronectin (FN) is a glycopeptide in the extracellular matrix and exhibits opsonic properties, aiding the phagocytosis and clearance of foreign pathogens through all stages of ulcer healing. But native FN comes from animal blood, which has potential health risks. rhFN3C was designed with multi-domains of native FN, whose core functions are the recruitment of cells and growth factors to accelerate AU healing. rhFN3C is a peptide-fused recombinant protein. The peptides are derived from the positions of 1444-1545 (FNIII10) and 1632-1901 (FNIII12-14) in human native FN. We optimized the fermentation conditions of rhFN3C in E. coli BL21 to enable high expression levels. rhFN3C is thermally stable and nontoxic for L929, strongly promotes the migration and adhesion of HaCaT, decreases the incidence of wound infection, and shortens the mean healing time by about 2 days compared to others (p < 0.01). rhFN3C may have great potential for use in the treatment of AUs. The specific methods and mechanisms of rhFN3C are yet to be investigated.
Collapse
Affiliation(s)
- Xiang Cai
- State Key Laboratory of Bioactive Molecules and Drug Gability Assessment, Jinan University, Guangzhou 510632, China; (X.C.); (J.Z.); (X.L.); (G.J.); (Y.H.)
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- Department of Materials Science and Engineering, Institute of Biomedical Engineering, Engineering Research Center of Artificial Organs and Materials, Jinan University, Guangzhou 510632, China
- Biopharmaceutical R&D Center of Jinan University, Guangzhou 510632, China
| | - Jiawen Zhu
- State Key Laboratory of Bioactive Molecules and Drug Gability Assessment, Jinan University, Guangzhou 510632, China; (X.C.); (J.Z.); (X.L.); (G.J.); (Y.H.)
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- Biopharmaceutical R&D Center of Jinan University, Guangzhou 510632, China
| | - Xin Luo
- State Key Laboratory of Bioactive Molecules and Drug Gability Assessment, Jinan University, Guangzhou 510632, China; (X.C.); (J.Z.); (X.L.); (G.J.); (Y.H.)
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- Biopharmaceutical R&D Center of Jinan University, Guangzhou 510632, China
| | - Guoguo Jin
- State Key Laboratory of Bioactive Molecules and Drug Gability Assessment, Jinan University, Guangzhou 510632, China; (X.C.); (J.Z.); (X.L.); (G.J.); (Y.H.)
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- Biopharmaceutical R&D Center of Jinan University, Guangzhou 510632, China
| | - Yadong Huang
- State Key Laboratory of Bioactive Molecules and Drug Gability Assessment, Jinan University, Guangzhou 510632, China; (X.C.); (J.Z.); (X.L.); (G.J.); (Y.H.)
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- Biopharmaceutical R&D Center of Jinan University, Guangzhou 510632, China
| | - Lihua Li
- State Key Laboratory of Bioactive Molecules and Drug Gability Assessment, Jinan University, Guangzhou 510632, China; (X.C.); (J.Z.); (X.L.); (G.J.); (Y.H.)
- Department of Materials Science and Engineering, Institute of Biomedical Engineering, Engineering Research Center of Artificial Organs and Materials, Jinan University, Guangzhou 510632, China
| |
Collapse
|
3
|
Matsuoka T, Yashiro M. Molecular Insight into Gastric Cancer Invasion-Current Status and Future Directions. Cancers (Basel) 2023; 16:54. [PMID: 38201481 PMCID: PMC10778111 DOI: 10.3390/cancers16010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. There has been no efficient therapy for stage IV GC patients due to this disease's heterogeneity and dissemination ability. Despite the rapid advancement of molecular targeted therapies, such as HER2 and immune checkpoint inhibitors, survival of GC patients is still unsatisfactory because the understanding of the mechanism of GC progression is still incomplete. Invasion is the most important feature of GC metastasis, which causes poor mortality in patients. Recently, genomic research has critically deepened our knowledge of which gene products are dysregulated in invasive GC. Furthermore, the study of the interaction of GC cells with the tumor microenvironment has emerged as a principal subject in driving invasion and metastasis. These results are expected to provide a profound knowledge of how biological molecules are implicated in GC development. This review summarizes the advances in our current understanding of the molecular mechanism of GC invasion. We also highlight the future directions of the invasion therapeutics of GC. Compared to conventional therapy using protease or molecular inhibitors alone, multi-therapy targeting invasion plasticity may seem to be an assuring direction for the progression of novel strategies.
Collapse
Affiliation(s)
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan;
| |
Collapse
|
4
|
Lee HJ, Tomasini-Johansson BR, Gupta N, Kwon GS. Fibronectin-targeted FUD and PEGylated FUD peptides for fibrotic diseases. J Control Release 2023; 360:69-81. [PMID: 37315694 PMCID: PMC10527082 DOI: 10.1016/j.jconrel.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
Tissue fibrosis is characterized by excessive deposition of extracellular matrix (ECM) molecules. Fibronectin (FN) is a glycoprotein found in the blood and tissues, a key player in the assembly of ECM through interaction with cellular and extracellular components. Functional Upstream Domain (FUD), a peptide derived from an adhesin protein of bacteria, has a high binding affinity for the N-terminal 70-kDa domain of FN that plays a crucial role in FN polymerization. In this regard, FUD peptide has been characterized as a potent inhibitor of FN matrix assembly, reducing excessive ECM accumulation. Furthermore, PEGylated FUD was developed to prevent rapid elimination of FUD and enhance its systemic exposure in vivo. Herein, we summarize the development of FUD peptide as a potential anti-fibrotic agent and its application in experimental fibrotic diseases. In addition, we discuss how modification of the FUD peptide via PEGylation impacts pharmacokinetic profiles of the FUD peptide and can potentially contribute to anti-fibrosis therapy.
Collapse
Affiliation(s)
- Hye Jin Lee
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, WI 53705, USA
| | - Bianca R Tomasini-Johansson
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, 1111 Highland Avenue, WIMRII, Madison, WI 53705, USA
| | - Nikesh Gupta
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, WI 53705, USA
| | - Glen S Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, WI 53705, USA; Carbone Cancer Center, University of Wisconsin - Madison, 600 Highland Avenue, Madison, WI 53705, USA.
| |
Collapse
|
5
|
Gutiérrez-Galindo E, Yilmaz ZH, Hausser A. Membrane trafficking in breast cancer progression: protein kinase D comes into play. Front Cell Dev Biol 2023; 11:1173387. [PMID: 37293129 PMCID: PMC10246754 DOI: 10.3389/fcell.2023.1173387] [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: 02/24/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Protein kinase D (PKD) is a serine/threonine kinase family that controls important cellular functions, most notably playing a key role in the secretory pathway at the trans-Golgi network. Aberrant expression of PKD isoforms has been found mainly in breast cancer, where it promotes various cellular processes such as growth, invasion, survival and stem cell maintenance. In this review, we discuss the isoform-specific functions of PKD in breast cancer progression, with a particular focus on how the PKD controlled cellular processes might be linked to deregulated membrane trafficking and secretion. We further highlight the challenges of a therapeutic approach targeting PKD to prevent breast cancer progression.
Collapse
Affiliation(s)
| | - Zeynep Hazal Yilmaz
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
6
|
Cheng SH, Chiou HYC, Wang JW, Lin MH. Reciprocal Regulation of Cancer-Associated Fibroblasts and Tumor Microenvironment in Gastrointestinal Cancer: Implications for Cancer Dormancy. Cancers (Basel) 2023; 15:2513. [PMID: 37173977 PMCID: PMC10177044 DOI: 10.3390/cancers15092513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Gastrointestinal (GI) cancers remain a major cause of cancer-related deaths worldwide. Despite the progress made in current treatments, patients with GI cancers still have high recurrence rates after initial treatment. Cancer dormancy, which involves the entry and escape of cancer cells from dormancy, is linked to treatment resistance, metastasis, and disease relapse. Recently, the role of the tumor microenvironment (TME) in disease progression and treatment has received increasing attention. The crosstalk between cancer-associated fibroblasts (CAF)-secreted cytokines/chemokines and other TME components, for example, extracellular matrix remodeling and immunomodulatory functions, play crucial roles in tumorigenesis. While there is limited direct evidence of a relationship between CAFs and cancer cell dormancy, this review explores the potential of CAF-secreted cytokines/chemokines to either promote cancer cell dormancy or awaken dormant cancer cells under different conditions, and the therapeutic strategies that may be applicable. By understanding the interactions between cytokines/chemokines released by CAFs and the TME, and their impact on the entry/escape of cancer dormancy, researchers may develop new strategies to reduce the risk of therapeutic relapse in patients with GI cancers.
Collapse
Affiliation(s)
- Shih-Hsuan Cheng
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hsin-Ying Clair Chiou
- Teaching and Research Center, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
- Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jiunn-Wei Wang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Hong Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Post Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
7
|
Raskov H, Gaggar S, Tajik A, Orhan A, Gögenur I. The Matrix Reloaded-The Role of the Extracellular Matrix in Cancer. Cancers (Basel) 2023; 15:2057. [PMID: 37046716 PMCID: PMC10093330 DOI: 10.3390/cancers15072057] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
As the core component of all organs, the extracellular matrix (ECM) is an interlocking macromolecular meshwork of proteins, glycoproteins, and proteoglycans that provides mechanical support to cells and tissues. In cancer, the ECM can be remodelled in response to environmental cues, and it controls a plethora of cellular functions, including metabolism, cell polarity, migration, and proliferation, to sustain and support oncogenesis. The biophysical and biochemical properties of the ECM, such as its structural arrangement and being a reservoir for bioactive molecules, control several intra- and intercellular signalling pathways and induce cytoskeletal changes that alter cell shapes, behaviour, and viability. Desmoplasia is a major component of solid tumours. The abnormal deposition and composition of the tumour matrix lead to biochemical and biomechanical alterations that determine disease development and resistance to treatment. This review summarises the complex roles of ECM in cancer and highlights the possible therapeutic targets and how to potentially remodel the dysregulated ECM in the future. Furthering our understanding of the ECM in cancer is important as the modification of the ECM will probably become an important tool in the characterisation of individual tumours and personalised treatment options.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Shruti Gaggar
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Asma Tajik
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Oncology, Zealand University Hospital, 4000 Roskilde, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
8
|
Chavez T, Gerecht S. Engineering of the microenvironment to accelerate vascular regeneration. Trends Mol Med 2023; 29:35-47. [PMID: 36371337 PMCID: PMC9742290 DOI: 10.1016/j.molmed.2022.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/11/2022]
Abstract
Blood vessels are crucial for tissue development, functionality, and homeostasis and are typically a determinant in the progression of healing and regeneration. The tissue microenvironment provides physicochemical cues that affect cellular function, and the study of the microenvironment can be accelerated by the engineering of approaches capable of mimicking various aspects of the microenvironment. In this review, we introduce the major components of the vascular niche and focus on the roles of oxygen and the extracellular matrix (ECM). We demonstrate how vascular engineering approaches enhance our understanding of the microenvironment's impact on the vasculature towards vascular regeneration and describe the current limitations and future directions towards clinical utilization.
Collapse
Affiliation(s)
- Taylor Chavez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, NC, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
9
|
The application of 3D bioprinting in urological diseases. Mater Today Bio 2022; 16:100388. [PMID: 35967737 PMCID: PMC9364106 DOI: 10.1016/j.mtbio.2022.100388] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/24/2022] [Accepted: 07/27/2022] [Indexed: 12/02/2022] Open
Abstract
Urologic diseases are commonly diagnosed health problems affecting people around the world. More than 26 million people suffer from urologic diseases and the annual expenditure was more than 11 billion US dollars. The urologic cancers, like bladder cancer, prostate cancer and kidney cancer are always the leading causes of death worldwide, which account for approximately 22% and 10% of the new cancer cases and death, respectively. Organ transplantation is one of the major clinical treatments for urological diseases like end-stage renal disease and urethral stricture, albeit strongly limited by the availability of matching donor organs. Tissue engineering has been recognized as a highly promising strategy to solve the problems of organ donor shortage by the fabrication of artificial organs/tissue. This includes the prospective technology of three-dimensional (3D) bioprinting, which has been adapted to various cell types and biomaterials to replicate the heterogeneity of urological organs for the investigation of organ transplantation and disease progression. This review discusses various types of 3D bioprinting methodologies and commonly used biomaterials for urological diseases. The literature shows that advances in this field toward the development of functional urological organs or disease models have progressively increased. Although numerous challenges still need to be tackled, like the technical difficulties of replicating the heterogeneity of urologic organs and the limited biomaterial choices to recapitulate the complicated extracellular matrix components, it has been proved by numerous studies that 3D bioprinting has the potential to fabricate functional urological organs for clinical transplantation and in vitro disease models. Outline the advantages and characteristics of 3D printing compared with traditional methods for urological diseases. Guide the selection of 3D bioprinting technology and material in urological tissue engineering. Discuss the challenges and future perspectives of 3D bioprinting in urological diseases and clinical translation.
Collapse
|
10
|
Potekaev NN, Borzykh OB, Shnayder NA, Petrova MM, Karpova EI, Nasyrova RF. Collagen synthesis in the skin: genetic and epigenetic aspects. BULLETIN OF SIBERIAN MEDICINE 2022. [DOI: 10.20538/1682-0363-2022-3-217-226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
One of the most important functions of the skin, mechanical, is provided by collagen fibers and their interaction with other elements of the extracellular matrix. Synthesis of collagen fibers is a complex multistep process. At each stage, disturbances may occur, leading, as a result, to a decrease in the mechanical properties of the connective tissue. In clinical practice, disorders of collagen synthesis are manifested through increased skin laxity and looseness and premature aging. In addition to the clinical presentation, it is important for the cosmetologist and dermatologist to understand the etiology and pathogenesis of collagenopathies. The present review summarizes and systematizes available information about the role of genetic and epigenetic factors in the synthesis of collagen fibers in the skin. Understanding the etiology of collagen synthesis disorders can allow doctors to prescribe pathogenetically grounded treatment with the most effective results and minimize adverse reactions.
Collapse
Affiliation(s)
- N. N. Potekaev
- Pirogov Russian National Research Medical University; Moscow Research and Practical Center for Dermatology and Cosmetology, Department of Healthcare
| | - O. B. Borzykh
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University
| | - N. A. Shnayder
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University; Bekhterev Psychoneurological Research Institute
| | - M. M. Petrova
- V.F. Voino-Yasenetsky Krasnoyarsk State Medical University
| | - E. I. Karpova
- Pirogov Russian National Research Medical University
| | - R. F. Nasyrova
- Bekhterev Psychoneurological Research Institute; Kazan Federal University
| |
Collapse
|
11
|
Jamieson JJ, Lin Y, Malloy N, Soto D, Searson PC, Gerecht S. Hypoxia-induced blood-brain barrier dysfunction is prevented by pericyte-conditioned media via attenuated actomyosin contractility and claudin-5 stabilization. FASEB J 2022; 36:e22331. [PMID: 35476363 PMCID: PMC9060394 DOI: 10.1096/fj.202200010rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 11/11/2022]
Abstract
The blood-brain barrier (BBB) regulates molecular and cellular entry from the cerebrovasculature into the surrounding brain parenchyma. Many diseases of the brain are associated with dysfunction of the BBB, where hypoxia is a common stressor. However, the contribution of hypoxia to BBB dysfunction is challenging to study due to the complexity of the brain microenvironment. In this study, we used a BBB model with brain microvascular endothelial cells and pericytes differentiated from iPSCs to investigate the effect of hypoxia on barrier function. We found that hypoxia-induced barrier dysfunction is dependent upon increased actomyosin contractility and is associated with increased fibronectin fibrillogenesis. We propose a role for actomyosin contractility in mediating hypoxia-induced barrier dysfunction through modulation of junctional claudin-5. Our findings suggest pericytes may protect brain microvascular endothelial cells from hypoxic stresses and that pericyte-derived factors could be candidates for treatment of pathological barrier-forming tissues.
Collapse
Affiliation(s)
- John J Jamieson
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - YingYu Lin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicholas Malloy
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel Soto
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Duke University, Duke, North Carolina, USA
| |
Collapse
|
12
|
Xue J, Liao Q, Luo M, Hua C, Zhao J, Yu G, Chen X, Li X, Zhang X, Ran R, Lu F, Wang Y, Qiao L. Cigarette smoke-induced oxidative stress activates NRF2 to mediate fibronectin disorganization in vascular formation. Open Biol 2022; 12:210310. [PMID: 35472288 PMCID: PMC9042581 DOI: 10.1098/rsob.210310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/20/2022] [Indexed: 01/09/2023] Open
Abstract
Cigarette smoke significantly induces oxidative stress, resulting in cardiovascular disease. NRF2, a well-known antioxidative stress response factor, is generally considered to play protective roles in cardiovascular dysfunction triggered by oxidative stress. Interestingly, recent studies reported adverse effects of NRF2 on the cardiovascular system. These unfavourable pathogenic effects of NRF2 need to be further investigated. Our work shows that cigarette smoke extract (CSE)-induced oxidative stress disturbs fibronectin (FN) assembly during angiogenesis. Furthermore, this effect largely depends on hyperactive NRF2-STAT3 signalling, which consequently promotes abnormal FN deposition. Consistently, disruption of this pathway by inhibiting NRF2 or STAT3 prevents CSE-induced FN disorganization and vasculature disruption in human umbilical vein endothelial cells or zebrafish. Taken together, these findings demonstrate the cardiovascular dysfunction caused by CSE from a novel perspective that NRF2-dependent signalling engages in FN disorganization.
Collapse
Affiliation(s)
- Jinjiang Xue
- College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
- Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Qiong Liao
- Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Man Luo
- Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Chenfeng Hua
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, 2 Fengyang Street, Zhengzhou 450001, People's Republic of China
| | - Junwei Zhao
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, 2 Fengyang Street, Zhengzhou 450001, People's Republic of China
| | - Gangfeng Yu
- Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Xiangyu Chen
- Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Xueru Li
- Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Xinchun Zhang
- Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Ruiguo Ran
- Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Fanghui Lu
- Institute of Life Sciences, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Yingxiong Wang
- College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| | - Liangjun Qiao
- College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, People's Republic of China
| |
Collapse
|
13
|
He X, Lee B, Jiang Y. Extracellular matrix in cancer progression and therapy. MEDICAL REVIEW (2021) 2022; 2:125-139. [PMID: 37724245 PMCID: PMC10471113 DOI: 10.1515/mr-2021-0028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/31/2022] [Indexed: 09/20/2023]
Abstract
The tumor ecosystem with heterogeneous cellular compositions and the tumor microenvironment has increasingly become the focus of cancer research in recent years. The extracellular matrix (ECM), the major component of the tumor microenvironment, and its interactions with the tumor cells and stromal cells have also enjoyed tremendously increased attention. Like the other components of the tumor microenvironment, the ECM in solid tumors differs significantly from that in normal organs and tissues. We review recent studies of the complex roles the tumor ECM plays in cancer progression, from tumor initiation, growth to angiogenesis and invasion. We highlight that the biomolecular, biophysical, and mechanochemical interactions between the ECM and cells not only regulate the steps of cancer progression, but also affect the efficacy of systemic cancer treatment. We further discuss the strategies to target and modify the tumor ECM to improve cancer therapy.
Collapse
Affiliation(s)
- Xiuxiu He
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Byoungkoo Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
14
|
Lickert S, Kenny M, Selcuk K, Mehl JL, Bender M, Früh SM, Burkhardt MA, Studt JD, Nieswandt B, Schoen I, Vogel V. Platelets drive fibronectin fibrillogenesis using integrin αIIbβ3. SCIENCE ADVANCES 2022; 8:eabj8331. [PMID: 35275711 PMCID: PMC8916723 DOI: 10.1126/sciadv.abj8331] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Platelets interact with multiple adhesion proteins during thrombogenesis, yet little is known about their ability to assemble fibronectin matrix. In vitro three-dimensional superresolution microscopy complemented by biophysical and biochemical methods revealed fundamental insights into how platelet contractility drives fibronectin fibrillogenesis. Platelets adhering to thrombus proteins (fibronectin and fibrin) versus basement membrane components (laminin and collagen IV) pull fibronectin fibrils along their apical membrane versus underneath their basal membrane, respectively. In contrast to other cell types, platelets assemble fibronectin nanofibrils using αIIbβ3 rather than α5β1 integrins. Apical fibrillogenesis correlated with a stronger activation of integrin-linked kinase, higher platelet traction forces, and a larger tension in fibrillar-like adhesions compared to basal fibrillogenesis. Our findings have potential implications for how mechanical thrombus integrity might be maintained during remodeling and vascular repair.
Collapse
Affiliation(s)
- Sebastian Lickert
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Martin Kenny
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Kateryna Selcuk
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Johanna L. Mehl
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Markus Bender
- Institute of Experimental Biomedicine – Chair I, University Hospital, and Rudolf Virchow Center, Julius Maximilian University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Susanna M. Früh
- Hahn-Schickard, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
- Laboratory for MEMS Applications, IMTEK Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
| | - Melanie A. Burkhardt
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Jan-Dirk Studt
- Division of Hematology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine – Chair I, University Hospital, and Rudolf Virchow Center, Julius Maximilian University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Ingmar Schoen
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
- Corresponding author. (V.V.); (I.S.)
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
- Corresponding author. (V.V.); (I.S.)
| |
Collapse
|
15
|
Duan K, Dash BC, Sasson DC, Islam S, Parker J, Hsia HC. Human iPSC-Derived Vascular Smooth Muscle Cells in a Fibronectin Functionalized Collagen Hydrogel Augment Endothelial Cell Morphogenesis. Bioengineering (Basel) 2021; 8:223. [PMID: 34940376 PMCID: PMC8698933 DOI: 10.3390/bioengineering8120223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/19/2023] Open
Abstract
Tissue-engineered constructs have immense potential as autologous grafts for wound healing. Despite the rapid advancement in fabrication technology, the major limitation is controlling angiogenesis within these constructs to form a vascular network. Here, we aimed to develop a 3D hydrogel that can regulate angiogenesis. We tested the effect of fibronectin and vascular smooth muscle cells derived from human induced pluripotent stem cells (hiPSC-VSMC) on the morphogenesis of endothelial cells. The results demonstrate that fibronectin increases the number of EC networks. However, hiPSC-VSMC in the hydrogel further substantiated the number and size of EC networks by vascular endothelial growth factor and basic fibroblast growth factor secretion. A mechanistic study shows that blocking αvβ3 integrin signaling between hiPSC-VSMC and fibronectin impacts the EC network formation via reduced cell viability and proangiogenic growth factor secretion. Collectively, this study set forth initial design criteria in developing an improved pre-vascularized construct.
Collapse
Affiliation(s)
- Kaiti Duan
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (K.D.); (D.C.S.); (S.I.); (J.P.)
| | - Biraja C. Dash
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (K.D.); (D.C.S.); (S.I.); (J.P.)
| | - Daniel C. Sasson
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (K.D.); (D.C.S.); (S.I.); (J.P.)
| | - Sara Islam
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (K.D.); (D.C.S.); (S.I.); (J.P.)
| | - Jackson Parker
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (K.D.); (D.C.S.); (S.I.); (J.P.)
| | - Henry C. Hsia
- Section of Plastic Surgery, Department of Surgery Yale School of Medicine, Yale University, New Haven, CT 06510, USA; (K.D.); (D.C.S.); (S.I.); (J.P.)
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
16
|
Mohd Yunus MH, Rashidbenam Z, Fauzi MB, Bt Hj Idrus R, Bin Saim A. Evaluating Feasibility of Human Tissue Engineered Respiratory Epithelium Construct as a Potential Model for Tracheal Mucosal Reconstruction. Molecules 2021; 26:molecules26216724. [PMID: 34771136 PMCID: PMC8587409 DOI: 10.3390/molecules26216724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/15/2022] Open
Abstract
The normal function of the airway epithelium is vital for the host’s well-being. Conditions that might compromise the structure and functionality of the airway epithelium include congenital tracheal anomalies, infection, trauma and post-intubation injuries. Recently, the onset of COVID-19 and its complications in managing respiratory failure further intensified the need for tracheal tissue replacement. Thus far, plenty of naturally derived, synthetic or allogeneic materials have been studied for their applicability in tracheal tissue replacement. However, a reliable tracheal replacement material is missing. Therefore, this study used a tissue engineering approach for constructing tracheal tissue. Human respiratory epithelial cells (RECs) were isolated from nasal turbinate, and the cells were incorporated into a calcium chloride-polymerized human blood plasma to form a human tissue respiratory epithelial construct (HTREC). The quality of HTREC in vitro, focusing on the cellular proliferation, differentiation and distribution of the RECs, was examined using histological, gene expression and immunocytochemical analysis. Histological analysis showed a homogenous distribution of RECs within the HTREC, with increased proliferation of the residing RECs within 4 days of investigation. Gene expression analysis revealed a significant increase (p < 0.05) in gene expression level of proliferative and respiratory epithelial-specific markers Ki67 and MUC5B, respectively, within 4 days of investigation. Immunohistochemical analysis also confirmed the expression of Ki67 and MUC5AC markers in residing RECs within the HTREC. The findings show that calcium chloride-polymerized human blood plasma is a suitable material, which supports viability, proliferation and mucin secreting phenotype of RECs, and this suggests that HTREC can be a potential candidate for respiratory epithelial tissue reconstruction.
Collapse
Affiliation(s)
- Mohd Heikal Mohd Yunus
- Department of Physiology, Faculty of Medicine, UKM Medical Centre, Jalan Yaacob Latiff, Cheras, Kuala Lumpur 56000, Malaysia;
- Correspondence: ; Tel.: +60-123-137-644
| | - Zahra Rashidbenam
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (Z.R.); (M.B.F.)
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (Z.R.); (M.B.F.)
| | - Ruszymah Bt Hj Idrus
- Department of Physiology, Faculty of Medicine, UKM Medical Centre, Jalan Yaacob Latiff, Cheras, Kuala Lumpur 56000, Malaysia;
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (Z.R.); (M.B.F.)
| | - Aminuddin Bin Saim
- Ear, Nose & Throat Consultation Clinic, Ampang Puteri Specialist Hospital, Ampang 68000, Selangor, Malaysia;
| |
Collapse
|
17
|
Walji N, Kheiri S, Young EWK. Angiogenic Sprouting Dynamics Mediated by Endothelial-Fibroblast Interactions in Microfluidic Systems. Adv Biol (Weinh) 2021; 5:e2101080. [PMID: 34655165 DOI: 10.1002/adbi.202101080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/18/2021] [Indexed: 11/09/2022]
Abstract
Angiogenesis, the development of new blood vessels from existing vasculature, is a key process in normal development and pathophysiology. In vitro models are necessary for investigating mechanisms of angiogenesis and developing antiangiogenic therapies. Microfluidic cell culture models of angiogenesis are favored for their ability to recapitulate 3D tissue structures and control spatiotemporal aspects of the microenvironments. To capture the angiogenesis process, microfluidic models often include endothelial cells and a fibroblast component. However, the influence of fibroblast organization on resulting angiogenic behavior remains unclear. Here a comparative study of angiogenic sprouting on a microfluidic chip induced by fibroblasts in 2D monolayer, 3D dispersed, and 3D spheroid culture formats, is conducted. Vessel morphology and sprout distribution for each configuration are measured, and these observations are correlated with measurements of secreted factors and numerical simulations of diffusion gradients. The results demonstrate that angiogenic sprouting varies in response to fibroblast organization with correlating variations in secretory profile and secreted factor gradients across the microfluidic device. This study is anticipated to shed light on how sprouting dynamics are mediated by fibroblast configuration such that the microfluidic cell culture design process includes the selection of a fibroblast component where the effects are known and leveraged.
Collapse
Affiliation(s)
- Noosheen Walji
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, M5S 3G8, Canada.,Institute of Biomedical Engineering, University of Toronto, 160 College St., Toronto, M5S 3E1, Canada
| | - Sina Kheiri
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, M5S 3G8, Canada
| | - Edmond W K Young
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, M5S 3G8, Canada.,Institute of Biomedical Engineering, University of Toronto, 160 College St., Toronto, M5S 3E1, Canada
| |
Collapse
|
18
|
Abstract
One of the most important functions of the skin, i.e., protection from mechanical damage, is ensured by collagen fibers and their interaction with other elements in the extracellular matrix. Collagen fiber turnover is a complex multi-stage process. At each stage, a disruption may occur, leading to a decrease in the mechanical properties of the connective tissue. Clinically, collagen formation disorders manifest themselves as increased flabbiness and looseness of the skin and as early signs of facial aging. In addition to the clinical picture, it is important for cosmetologists and dermatologists to understand the etiology and pathogenesis of collagenopathies. In our review, we summarized and systematized the available information concerning the role of genetic and epigenetic factors in skin collagen fiber turnover. Furthermore, we focused on the functions of different types of collagens present in the skin. Understanding the etiology of impaired collagen formation can allow doctors to prescribe pathogenetically based treatments, achieve the most effective results, and minimize adverse reactions.
Collapse
|
19
|
Abstract
Significance: The vascular extracellular matrix (ECM) not only provides mechanical stability but also manipulates vascular cell behaviors, which are crucial for vascular function and homeostasis. ECM remodeling, which alters vascular wall mechanical properties and exposes vascular cells to bioactive molecules, is involved in the development and progression of hypertension. Recent Advances: This brief review summarized the dynamic changes in ECM components and their modification and degradation during hypertension and after antihypertensive treatment. We also discussed how alterations in the ECM amount, assembly, mechanical properties, and degradation fragment generation provide input into the pathological process of hypertension. Critical Issues: Although the relevance between ECM remodeling and hypertension has been recognized, the underlying mechanism by which ECM remodeling initiates the development of hypertension remains unclear. Therefore, the modulation of ECM remodeling on arterial stiffness and hypertension in genetically modified rodent models is summarized in this review. The circulating biomarkers based on ECM metabolism and therapeutic strategies targeting ECM disorders in hypertension are also introduced. Future Directions: Further research will provide more comprehensive understanding of ECM remodeling in hypertension by the application of matridomic and degradomic approaches. The better understanding of mechanisms underlying vascular ECM remodeling may provide novel potential therapeutic strategies for preventing and treating hypertension. Antioxid. Redox Signal. 34, 765-783.
Collapse
Affiliation(s)
- Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
20
|
do Monte FA, Ahuja N, Awad KR, Pan Z, Young S, Kim HKW, Aswath P, Brotto M, Varanasi VG. Silicon Oxynitrophosphide Nanoscale Coating Enhances Antioxidant Marker-Induced Angiogenesis During in vivo Cranial Bone-Defect Healing. JBMR Plus 2021; 5:e10425. [PMID: 33869985 PMCID: PMC8046063 DOI: 10.1002/jbm4.10425] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 09/18/2020] [Accepted: 10/01/2020] [Indexed: 11/08/2022] Open
Abstract
Critical-sized bone defects are challenging to heal because of the sudden and large volume of lost bone. Fixative plates are often used to stabilize defects, yet oxidative stress and delayed angiogenesis are contributing factors to poor biocompatibility and delayed bone healing. This study tests the angiogenic and antioxidant properties of amorphous silicon oxynitrophosphide (SiONPx) nanoscale-coating material on endothelial cells to regenerate vascular tissue in vitro and in bone defects. in vitro studies evaluate the effect of silicon oxynitride (SiONx) and two different SiONPx compositions on human endothelial cells exposed to ROS (eg, hydrogen peroxide) that simulates oxidative stress conditions. in vivo studies using adult male Sprague Dawley rats (approximately 450 g) were performed to compare a bare plate, a SiONPx-coated implant plate, and a sham control group using a rat standard-sized calvarial defect. Results from this study showed that plates coated with SiONPx significantly reduced cell death, and enhanced vascular tubule formation and matrix deposition by upregulating angiogenic and antioxidant expression (eg, vascular endothelial growth factor A, angiopoetin-1, superoxide dismutase 1, nuclear factor erythroid 2-related factor 2, and catalase 1). Moreover, endothelial cell markers (CD31) showed a significant tubular structure in the SiONPx coating group compared with an empty and uncoated plate group. This reveals that atomic doping of phosphate into the nanoscale coating of SiONx produced markedly elevated levels of antioxidant and angiogenic markers that enhance vascular tissue regeneration. This study found that SiONPx or SiONx nanoscale-coated materials enhance antioxidant expression, angiogenic marker expression, and reduce ROS levels needed for accelerating vascular tissue regeneration. These results further suggest that SiONPx nanoscale coating could be a promising candidate for titanium plate for rapid and enhanced cranial bone-defect healing. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Felipe A do Monte
- Department of BioengineeringUniversity of Texas at ArlingtonArlingtonTXUSA
- Center for Excellence in Hip DisordersTexas Scottish Rite HospitalDallasTXUSA
| | - Neelam Ahuja
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Kamal R Awad
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
- Department of Materials Science and EngineeringUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Zui Pan
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Simon Young
- Department of Oral and Maxillofacial SurgeryThe University of Texas Health Science Center at Houston, School of DentistryHoustonTXUSA
| | - Harry KW Kim
- Center for Excellence in Hip DisordersTexas Scottish Rite HospitalDallasTXUSA
- Department of Orthopedic SurgeryUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Pranesh Aswath
- Department of Materials Science and EngineeringUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Marco Brotto
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
| | - Venu G Varanasi
- Bone‐Muscle Research CenterUniversity of Texas at ArlingtonArlingtonTXUSA
- Department of Materials Science and EngineeringUniversity of Texas at ArlingtonArlingtonTXUSA
| |
Collapse
|
21
|
Identification of extracellular matrix proteins secreted by human dermal fibroblasts cultured in 3D electrospun scaffolds. Sci Rep 2021; 11:6655. [PMID: 33758206 PMCID: PMC7988018 DOI: 10.1038/s41598-021-85742-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
The appreciation that cell interactions in tissues is dependent on their three dimensional (3D) distribution has stimulated the development of 3D cell culture models. We constructed an artificial 3D tumour by culturing human breast cancer JIMT-1 cells and human dermal fibroblasts (HDFs) in a 3D network of electrospun polycaprolactone fibres. Here, we investigate ECM components produced by the cells in the artificial 3D tumour, which is an important step in validating the model. Immunostaining and confocal fluorescence microscopy show that the ECM proteins fibronectin, collagen I, and laminin are deposited throughout the entire 3D structure. Secreted soluble factors including matrix metalloproteinases (MMPs) and interleukine-6 (IL-6) were analysed in collected medium and were found to be mainly derived from the HDFs. Treatment with transforming growth factor-β1 (TGF-β1), a major cytokine found in a tumour, significantly alters the MMP activity and IL-6 concentration. In addition, TGF-β1 treatment, changes the morphology of the HDFs to become more elongated and with increased linearized actin filaments compared to non-treated HDFs. Collectively, these novel findings suggest that the artificial 3D tumour displays a clear cell distribution and ECM deposition that resembles a tumour environment in vivo, suggesting an innovative biological model to study a human tumour.
Collapse
|
22
|
Barrs RW, Jia J, Ward M, Richards DJ, Yao H, Yost MJ, Mei Y. Engineering a Chemically Defined Hydrogel Bioink for Direct Bioprinting of Microvasculature. Biomacromolecules 2021; 22:275-288. [PMID: 33332959 PMCID: PMC7870577 DOI: 10.1021/acs.biomac.0c00947] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Vascularizing printed tissues is a critical challenge in bioprinting. While protein-based hydrogel bioinks have been successfully used to bioprint microvasculature, their compositions are ill-defined and subject to batch variation. Few studies have focused on engineering proangiogenic bioinks with defined properties to direct endogenous microvascular network formation after printing. Here, a peptide-functionalized alginate hydrogel bioink with defined mechanical, rheological, and biochemical properties is developed for direct bioprinting of microvascularized tissues. An integrin-binding peptide (RGD) and a vascular endothelial growth factor-mimetic peptide with a protease-sensitive linker are conjugated onto a biodegradable alginate to synergistically promote vascular morphogenesis and capillary-scale endothelial tube formation. Partial ionic crosslinking before printing converts the otherwise unprintable hydrogel into a viscoelastic bioink with excellent printability and cytocompatibility. We use the bioink to fabricate a compartmentalized vascularized tissue construct, wherein we observe pericyte-endothelial cell colocalization and angiogenic sprouting across a tissue interface, accompanied by deposition of fibronectin and collagen in vascular and tissue components, respectively. This study provides a tunable and translational "off-the-shelf" hydrogel bioink with defined composition for vascularized bioprinting.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Department of Bioengineering, Clemson University, USA
- Department of Surgery, Medical University of South
Carolina, USA
| | - Jia Jia
- Department of Bioengineering, Clemson University, USA
| | - Michael Ward
- Department of Bioengineering, Clemson University, USA
| | | | - Hai Yao
- Department of Bioengineering, Clemson University, USA
| | - Michael J. Yost
- Department of Surgery, Medical University of South
Carolina, USA
- Department of Regenerative Medicine and Cell Biology,
Medical University of South Carolina, USA
| | - Ying Mei
- Department of Bioengineering, Clemson University, USA
- Department of Regenerative Medicine and Cell Biology,
Medical University of South Carolina, USA
| |
Collapse
|
23
|
Influence of microcurrent on the modulation of remodelling genes in a wound healing assay. Mol Biol Rep 2021; 48:1233-1241. [PMID: 33475929 DOI: 10.1007/s11033-021-06135-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/02/2021] [Indexed: 10/22/2022]
Abstract
The literature has shown the beneficial effects of microcurrent (MC) therapy on tissue repair. We investigated if the application of MC at 10 μA/90 s could modulate the expression of remodeling genes transforming growth factor beta (Tgfb), connective tissue growth factor (Ctgf), insulin-like growth factor 1 (Igf1), tenascin C (Tnc), Fibronectin (Fn1), Scleraxis (Scx), Fibromodulin (Fmod) and tenomodulin in NIH/3T3 fibroblasts in a wound healing assay. The cell migration was analyzed between days 0 and 4 in both fibroblasts (F) and fibroblasts + MC (F+MC) groups. On the 4th day, cell viability and gene expression were also analyzed after daily MC application. Higher expression of Ctgf and lower expression of Tnc and Fmod, respectively, were observed in the F+MC group in relation to F group (p < 0.05), and no difference was observed between the groups for the genes Tgfb, Fn1 and Scx. In cell migration, a higher number of cells in the scratch region was observed in group F+MC (p < 0.05) compared to group F on the 4th day, and the cell viability assay showed no difference between the groups. In conclusion, MC therapy at an intensity/time of 10 μA/90 s with 4 daily applications did not affect cell viability, stimulated fibroblasts migration with the involvement of Ctgf, and reduced the Tnc and Fmod expression.
Collapse
|
24
|
He S, Ouyang S, Li X, Ma B. Inhibition of laser induced rats choroidal neovascularization by intravitreous injection of sEphB4-HSA. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:18. [PMID: 33553311 PMCID: PMC7859820 DOI: 10.21037/atm-20-3810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background Choroidal neovascularization (CNV) is a leading cause of central vision loss complicated with age-related macular degeneration. Although intravitreal anti-VEGF therapy is widely used in wet age-related macular degeneration, optimal treatment regimens for the disease are still under investigation. EphrinB2 and EphB4 regulate angiogenesis, and interruption of EphB4/ephrinB2 has been demonstrated to inhibit angiogenesis. In the current study, we studied the effects of soluble EphB4 (sEphB4) on laser induced CNV in a rat model by intravitreous injection and the underlying mechanism. Methods Male rats (Brown-Norway) were used in the study. CNV was induced by laser and the sEphB4 was injected intravitreous after laser at days 3 and 7. The CNV lesions were evaluated by three methods: fluorescein angiography (FA) in vivo, CNV volume by confocal analysis of choroidal flat-mounts and H&E staining. The expression of fibronectin (FN), VEGFR-2, phospho-VEGFR-2 (pVEGFR-2), the double labeling of EphB4 with FN was analyzed by immunofluorescence. The interaction of FN with EphB4 and the effects of intraocular injection of sEphB4 on the inhibition of pVEGFR-2 were determined by western blot. Results The FA leakage and CNV volume were significantly inhibited by the injection of the sEphB4. Further, histology analysis showed that CNV lesion was significantly smaller in the rats with sEphB4 injection than rats with placebo application. The expressions of pVEGFR-2 and FN in the CNV lesions were reduced compared with controls. Conclusions Our study suggests that the inhibition of CNV by sEphB4 may be through suppression of VEGFR-2 phosphorylation and the expression of FN. sEphB4 may be a new potential therapeutic strategy of CNV.
Collapse
Affiliation(s)
- Shikun He
- Department of Pathology and USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Sha Ouyang
- Department of Pathology and USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.,Department of Ophthalmology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaohua Li
- Department of Ophthalmology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Ophthalmology, Henan Eye Hospital, Zhengzhou, China
| | - Binyun Ma
- Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
25
|
Howe MD, McCullough LD, Urayama A. The Role of Basement Membranes in Cerebral Amyloid Angiopathy. Front Physiol 2020; 11:601320. [PMID: 33329053 PMCID: PMC7732667 DOI: 10.3389/fphys.2020.601320] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022] Open
Abstract
Dementia is a neuropsychiatric syndrome characterized by cognitive decline in multiple domains, often leading to functional impairment in activities of daily living, disability, and death. The most common causes of age-related progressive dementia include Alzheimer's disease (AD) and vascular cognitive impairment (VCI), however, mixed disease pathologies commonly occur, as epitomized by a type of small vessel pathology called cerebral amyloid angiopathy (CAA). In CAA patients, the small vessels of the brain become hardened and vulnerable to rupture, leading to impaired neurovascular coupling, multiple microhemorrhage, microinfarction, neurological emergencies, and cognitive decline across multiple functional domains. While the pathogenesis of CAA is not well understood, it has long been thought to be initiated in thickened basement membrane (BM) segments, which contain abnormal protein deposits and amyloid-β (Aβ). Recent advances in our understanding of CAA pathogenesis link BM remodeling to functional impairment of perivascular transport pathways that are key to removing Aβ from the brain. Dysregulation of this process may drive CAA pathogenesis and provides an important link between vascular risk factors and disease phenotype. The present review summarizes how the structure and composition of the BM allows for perivascular transport pathways to operate in the healthy brain, and then outlines multiple mechanisms by which specific dementia risk factors may promote dysfunction of perivascular transport pathways and increase Aβ deposition during CAA pathogenesis. A better understanding of how BM remodeling alters perivascular transport could lead to novel diagnostic and therapeutic strategies for CAA patients.
Collapse
Affiliation(s)
| | | | - Akihiko Urayama
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
26
|
Shiwarski DJ, Tashman JW, Tsamis A, Bliley JM, Blundon MA, Aranda-Michel E, Jallerat Q, Szymanski JM, McCartney BM, Feinberg AW. Fibronectin-based nanomechanical biosensors to map 3D surface strains in live cells and tissue. Nat Commun 2020; 11:5883. [PMID: 33208732 PMCID: PMC7675982 DOI: 10.1038/s41467-020-19659-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 10/19/2020] [Indexed: 01/07/2023] Open
Abstract
Mechanical forces are integral to cellular migration, differentiation and tissue morphogenesis; however, it has proved challenging to directly measure strain at high spatial resolution with minimal perturbation in living sytems. Here, we fabricate, calibrate, and test a fibronectin (FN)-based nanomechanical biosensor (NMBS) that can be applied to the surface of cells and tissues to measure the magnitude, direction, and strain dynamics from subcellular to tissue length-scales. The NMBS is a fluorescently-labeled, ultra-thin FN lattice-mesh with spatial resolution tailored by adjusting the width and spacing of the lattice from 2-100 µm. Time-lapse 3D confocal imaging of the NMBS demonstrates 2D and 3D surface strain tracking during mechanical deformation of known materials and is validated with finite element modeling. Analysis of the NMBS applied to single cells, cell monolayers, and Drosophila ovarioles highlights the NMBS's ability to dynamically track microscopic tensile and compressive strains across diverse biological systems where forces guide structure and function.
Collapse
Affiliation(s)
- Daniel J Shiwarski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Joshua W Tashman
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Alkiviadis Tsamis
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Jaci M Bliley
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Malachi A Blundon
- Department of Biology, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Edgar Aranda-Michel
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Quentin Jallerat
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - John M Szymanski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Brooke M McCartney
- Department of Biology, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Department of Materials Science & Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
27
|
Ngo MT, Harley BAC. Angiogenic biomaterials to promote therapeutic regeneration and investigate disease progression. Biomaterials 2020; 255:120207. [PMID: 32569868 PMCID: PMC7396313 DOI: 10.1016/j.biomaterials.2020.120207] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023]
Abstract
The vasculature is a key component of the tissue microenvironment. Traditionally known for its role in providing nutrients and oxygen to surrounding cells, the vasculature is now also acknowledged to provide signaling cues that influence biological outcomes in regeneration and disease. These cues come from the cells that comprise vasculature, as well as the dynamic biophysical and biochemical properties of the surrounding extracellular matrix that accompany vascular development and remodeling. In this review, we illustrate the larger role of the vasculature in the context of regenerative biology and cancer progression. We describe cellular, biophysical, biochemical, and metabolic components of vascularized microenvironments. Moreover, we provide an overview of multidimensional angiogenic biomaterials that have been developed to promote therapeutic vascularization and regeneration, as well as to mimic elements of vascularized microenvironments as a means to uncover mechanisms by which vasculature influences cancer progression and therapy.
Collapse
Affiliation(s)
- Mai T Ngo
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
28
|
Trujillo S, Gonzalez-Garcia C, Rico P, Reid A, Windmill J, Dalby MJ, Salmeron-Sanchez M. Engineered 3D hydrogels with full-length fibronectin that sequester and present growth factors. Biomaterials 2020; 252:120104. [DOI: 10.1016/j.biomaterials.2020.120104] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/22/2022]
|
29
|
Avagliano A, Fiume G, Ruocco MR, Martucci N, Vecchio E, Insabato L, Russo D, Accurso A, Masone S, Montagnani S, Arcucci A. Influence of Fibroblasts on Mammary Gland Development, Breast Cancer Microenvironment Remodeling, and Cancer Cell Dissemination. Cancers (Basel) 2020; 12:E1697. [PMID: 32604738 PMCID: PMC7352995 DOI: 10.3390/cancers12061697] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
The stromal microenvironment regulates mammary gland development and tumorigenesis. In normal mammary glands, the stromal microenvironment encompasses the ducts and contains fibroblasts, the main regulators of branching morphogenesis. Understanding the way fibroblast signaling pathways regulate mammary gland development may offer insights into the mechanisms of breast cancer (BC) biology. In fact, the unregulated mammary fibroblast signaling pathways, associated with alterations in extracellular matrix (ECM) remodeling and branching morphogenesis, drive breast cancer microenvironment (BCM) remodeling and cancer growth. The BCM comprises a very heterogeneous tissue containing non-cancer stromal cells, namely, breast cancer-associated fibroblasts (BCAFs), which represent most of the tumor mass. Moreover, the different components of the BCM highly interact with cancer cells, thereby generating a tightly intertwined network. In particular, BC cells activate recruited normal fibroblasts in BCAFs, which, in turn, promote BCM remodeling and metastasis. Thus, comparing the roles of normal fibroblasts and BCAFs in the physiological and metastatic processes, could provide a deeper understanding of the signaling pathways regulating BC dissemination. Here, we review the latest literature describing the structure of the mammary gland and the BCM and summarize the influence of epithelial-mesenchymal transition (EpMT) and autophagy in BC dissemination. Finally, we discuss the roles of fibroblasts and BCAFs in mammary gland development and BCM remodeling, respectively.
Collapse
Affiliation(s)
- Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy;
| | - Nunzia Martucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Eleonora Vecchio
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Luigi Insabato
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Daniela Russo
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Antonello Accurso
- Department of General, Oncological, Bariatric and Endocrine-Metabolic Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Montagnani
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| |
Collapse
|
30
|
Moon SJ, Kim JH, Kong SH, Shin CS. Protein Expression of Cyclin B1, Transferrin Receptor, and Fibronectin Is Correlated with the Prognosis of Adrenal Cortical Carcinoma. Endocrinol Metab (Seoul) 2020; 35:132-141. [PMID: 32207273 PMCID: PMC7090291 DOI: 10.3803/enm.2020.35.1.132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/25/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Adrenal cortical carcinoma (ACC) is a rare cancer with a variable prognosis. Several prognostic factors of ACC have been previously reported, but a proteomic analysis has not yet been performed. This study aimed to investigate prognostic biomarkers for ACC using a proteomic approach. METHODS We used reverse-phase protein array data from The Cancer Proteome Atlas, and identified differentially expressed proteins in metastatic ACCs. Multivariate Cox regression analysis adjusted by age and staging was used for survival analysis, and the C-index and category-free net reclassification improvement (cfNRI) were utilized to evaluate additive prognostic value. RESULTS In 46 patients with ACC, cyclin B1, transferrin receptor (TfR1), and fibronectin were significantly overexpressed in patients with distant metastasis. In multivariate models, high expression of cyclin B1 and TfR1 was significantly associated with mortality (hazard ratio [HR], 6.13; 95% confidence interval [CI], 1.02 to 36.7; and HR, 6.59; 95% CI, 1.14 to 38.2; respectively), whereas high fibronectin expression was not (HR, 3.92; 95% CI, 0.75 to 20.4). Combinations of high cyclin B1/high TfR1, high cyclin B1/high fibronectin, and high TfR1/high fibronectin were strongly associated with mortality ([HR, 13.72; 95% CI, 1.89 to 99.66], [HR, 9.22; 95% CI, 1.34 to 63.55], and [HR, 18.59; 95% CI, 2.54 to 135.88], respectively). In reclassification analyses, cyclin B1, TfR1, fibronectin, and combinations thereof improved the prognostic performance (C-index, 0.78 to 0.82-0.86; cfNRI, all P values <0.05). CONCLUSION In ACC patients, the overexpression of cyclin B1, TfR1, and fibronectin and combinations thereof were associated with poor prognosis.
Collapse
Affiliation(s)
- Sun Joon Moon
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Hee Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University College of Medicine, Seoul, Korea.
| | - Sung Hye Kong
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Chan Soo Shin
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
31
|
Smith Q, Macklin B, Chan XY, Jones H, Trempel M, Yoder MC, Gerecht S. Differential HDAC6 Activity Modulates Ciliogenesis and Subsequent Mechanosensing of Endothelial Cells Derived from Pluripotent Stem Cells. Cell Rep 2020; 24:895-908.e6. [PMID: 30044986 DOI: 10.1016/j.celrep.2018.06.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/30/2018] [Accepted: 06/20/2018] [Indexed: 01/10/2023] Open
Abstract
The role of primary cilia in mechanosensation is essential in endothelial cell (EC) shear responsiveness. Here, we find that venous, capillary, and progenitor ECs respond to shear stress in vitro in a cilia-dependent manner. We then demonstrate that primary cilia assembly in human induced pluripotent stem cell (hiPSC)-derived ECs varies between different cell lines with marginal influence of differentiation protocol. hiPSC-derived ECs lacking cilia do not align to shear stress, lack stress fiber assembly, have uncoordinated migration during wound closure in vitro, and have aberrant calcium influx upon shear exposure. Transcriptional analysis reveals variation in regulatory genes involved in ciliogenesis among different hiPSC-derived ECs. Moreover, inhibition of histone deacetylase 6 (HDAC6) activity in hiPSC-ECs lacking cilia rescues cilia formation and restores mechanical sensing. Taken together, these results show the importance of primary cilia in hiPSC-EC mechano-responsiveness and its modulation through HDAC6 activity varies among hiPSC-ECs.
Collapse
Affiliation(s)
- Quinton Smith
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bria Macklin
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xin Yi Chan
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hannah Jones
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Michelle Trempel
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Mervin C Yoder
- Department of Pediatrics, Biochemistry, and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Physical Sciences-Oncology Center and the Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
32
|
do Monte FA, Awad KR, Ahuja N, Kim HK, Aswath P, Brotto M, Varanasi VG. Amorphous Silicon Oxynitrophosphide-Coated Implants Boost Angiogenic Activity of Endothelial Cells. Tissue Eng Part A 2020; 26:15-27. [PMID: 31044666 PMCID: PMC6983748 DOI: 10.1089/ten.tea.2019.0051] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/29/2019] [Indexed: 12/29/2022] Open
Abstract
Lack of osteointegration is a major cause of aseptic loosening and failure of implants used in bone replacement. Implants coated with angiogenic biomaterials can improve osteointegration and potentially reduce these complications. Silicon- and phosphorus-based materials have been shown to upregulate expression of angiogenic factors and improve endothelial cell functions. In the present study, we hypothesize that implants coated with amorphous silica-based coatings in the form of silicon oxynitrophosphide (SiONP) by using plasma-enhanced chemical vapor deposition (PECVD) technique could enhance human umbilical vein endothelial cell angiogenic properties in vitro. The tested groups were: glass coverslip (GCS), tissue culture plate, SiON, SiONP1 (O: 7.3 at %), and SiONP2 (O: 14.2 at %) implants. The SiONP2 composition demonstrated 3.5-fold more fibronectin deposition than the GCS (p < 0.001). The SiONP2 group also presented a significant improvement in the capillary tubule length and thickness compared with the other groups (p < 0.01). At 24 h, we observed at least a twofold upregulation of vascular endothelial growth factor A, hypoxia-inducible factor-1α, angiopoietin-1, and nesprin-2, more evident in the SiONP1 and SiONP2 groups. In conclusion, the studied amorphous silica-coated implants, especially the SiONP2 composition, could enhance the endothelial cell angiogenic properties in vitro and may induce faster osteointegration and healing. Impact Statement In this study, we report for the first time the significant enhancement of human umbilical vein endothelial cell angiogenic properties (in vitro) by the amorphous silica-based coatings in the form of silicon oxynitrophosphide (SiONP). The SiONP2 demonstrated 3.5-fold more fibronectin deposition than the glass coverslip and presented a significant improvement in the capillary tubule length and thickness. At 24 h, SiONP reported twofold upregulation of vascular endothelial growth factor A, hypoxia-inducible factor-1α, angiopoietin-1, and nesprin-2. The studied amorphous silica-coated implants enhance the endothelial cell angiogenic properties in vitro and may induce faster osteointegration and healing.
Collapse
Affiliation(s)
- Felipe A. do Monte
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital, Dallas, Texas
| | - Kamal R. Awad
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Neelam Ahuja
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Harry K.W. Kim
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital, Dallas, Texas
- Department of Orthopedic Surgery, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas
| | - Pranesh Aswath
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Venu G. Varanasi
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| |
Collapse
|
33
|
Lopes-Coelho F, Silva F, Gouveia-Fernandes S, Martins C, Lopes N, Domingues G, Brito C, Almeida AM, Pereira SA, Serpa J. Monocytes as Endothelial Progenitor Cells (EPCs), Another Brick in the Wall to Disentangle Tumor Angiogenesis. Cells 2020; 9:cells9010107. [PMID: 31906296 PMCID: PMC7016533 DOI: 10.3390/cells9010107] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/22/2019] [Accepted: 12/30/2019] [Indexed: 12/12/2022] Open
Abstract
Bone marrow contains endothelial progenitor cells (EPCs) that, upon pro-angiogenic stimuli, migrate and differentiate into endothelial cells (ECs) and contribute to re-endothelialization and neo-vascularization. There are currently no reliable markers to characterize EPCs, leading to their inaccurate identification. In the past, we showed that, in a panel of tumors, some cells on the vessel wall co-expressed CD14 (monocytic marker) and CD31 (EC marker), indicating a putative differentiation route of monocytes into ECs. Herein, we disclosed monocytes as potential EPCs, using in vitro and in vivo models, and also addressed the cancer context. Monocytes acquired the capacity to express ECs markers and were able to be incorporated into blood vessels, contributing to cancer progression, by being incorporated in tumor neo-vasculature. Reactive oxygen species (ROS) push monocytes to EC differentiation, and this phenotype is reverted by cysteine (a scavenger and precursor of glutathione), which indicates that angiogenesis is controlled by the interplay between the oxidative stress and the scavenging capacity of the tumor microenvironment.
Collapse
Affiliation(s)
- Filipa Lopes-Coelho
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Fernanda Silva
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Sofia Gouveia-Fernandes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Carmo Martins
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Nuno Lopes
- Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica, 2780-157 Oeiras, Portugal; (N.L.); (C.B.)
| | - Germana Domingues
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Catarina Brito
- Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica, 2780-157 Oeiras, Portugal; (N.L.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - António M Almeida
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
- Hospital da Luz, Av. Lusíada 100, 1500-650 Lisboa, Portugal
| | - Sofia A Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
| | - Jacinta Serpa
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
- Correspondence: ; Tel.: +350-217-229-800; Fax: +351-217-248-756
| |
Collapse
|
34
|
Egorikhina MN, Aleynik DY, Rubtsova YP, Levin GY, Charykova IN, Semenycheva LL, Bugrova ML, Zakharychev EA. Hydrogel scaffolds based on blood plasma cryoprecipitate and collagen derived from various sources: Structural, mechanical and biological characteristics. Bioact Mater 2019; 4:334-345. [PMID: 31720490 PMCID: PMC6838346 DOI: 10.1016/j.bioactmat.2019.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/04/2019] [Accepted: 10/13/2019] [Indexed: 01/19/2023] Open
Abstract
At present there is a growing need for tissue engineering products, including the products of scaffold-technologies. Biopolymer hydrogel scaffolds have a number of advantages and are increasingly being used to provide means of cell transfer for therapeutic treatments and for inducing tissue regeneration. This work presents original hydrogel biopolymer scaffolds based on a blood plasma cryoprecipitate and collagen and formed under conditions of enzymatic hydrolysis. Two differently originated collagens were used for the scaffold formation. During this work the structural and mechanical characteristics of the scaffold were studied. It was found that, depending on the origin of collagen, scaffolds possess differences in their structural and mechanical characteristics. Both types of hydrogel scaffolds have good biocompatibility and provide conditions that maintain the three-dimensional growth of adipose tissue stem cells. Hence, scaffolds based on such a blood plasma cryoprecipitate and collagen have good prospects as cell carriers and can be widely used in regenerative medicine.
Collapse
Affiliation(s)
- Marfa N. Egorikhina
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Diana Ya Aleynik
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Yulia P. Rubtsova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Grigory Ya Levin
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Irina N. Charykova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | | | - Marina L. Bugrova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | | |
Collapse
|
35
|
Pranda MA, Gray KM, DeCastro AJL, Dawson GM, Jung JW, Stroka KM. Tumor Cell Mechanosensing During Incorporation into the Brain Microvascular Endothelium. Cell Mol Bioeng 2019; 12:455-480. [PMID: 31719927 DOI: 10.1007/s12195-019-00591-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022] Open
Abstract
Introduction Tumor metastasis to the brain occurs in approximately 20% of all cancer cases and often occurs due to tumor cells crossing the blood-brain barrier (BBB). The brain microenvironment is comprised of a soft hyaluronic acid (HA)-rich extracellular matrix with an elastic modulus of 0.1-1 kPa, whose crosslinking is often altered in disease states. Methods To explore the effects of HA crosslinking on breast tumor cell migration, we developed a biomimetic model of the human brain endothelium, consisting of brain microvascular endothelial cell (HBMEC) monolayers on HA and gelatin (HA/gelatin) films with different degrees of crosslinking, as established by varying the concentration of the crosslinker Extralink. Results and Discussion Metastatic breast tumor cell migration speed, diffusion coefficient, spreading area, and aspect ratio increased with decreasing HA crosslinking, a mechanosensing trend that correlated with tumor cell actin organization but not CD44 expression. Meanwhile, breast tumor cell incorporation into endothelial monolayers was independent of HA crosslinking density, suggesting that alterations in HA crosslinking density affect tumor cells only after they exit the vasculature. Tumor cells appeared to exploit both the paracellular and transcellular routes of trans-endothelial migration. Quantitative phenotyping of HBMEC junctions via a novel Python software revealed a VEGF-dependent decrease in punctate VE-cadherin junctions and an increase in continuous and perpendicular junctions when HBMECs were treated with tumor cell-secreted factors. Conclusions Overall, our quantitative results suggest that a combination of biochemical and physical factors promote tumor cell migration through the BBB.
Collapse
Affiliation(s)
- Marina A Pranda
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Kelsey M Gray
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Ariana Joy L DeCastro
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Gregory M Dawson
- Department of Biology, University of Maryland, College Park, College Park, MD 20742 USA
| | - Jae W Jung
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA.,Biophysics Program, University of Maryland, College Park, College Park, MD 20742 USA.,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland - Baltimore, Baltimore, MD 21201 USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland - Baltimore, Baltimore, MD 21201 USA.,Fischell Department of Bioengineering, University of Maryland, College Park, 3110 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742 USA
| |
Collapse
|
36
|
Crosby CO, Zoldan J. Mimicking the physical cues of the ECM in angiogenic biomaterials. Regen Biomater 2019; 6:61-73. [PMID: 30967961 PMCID: PMC6447000 DOI: 10.1093/rb/rbz003] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/02/2018] [Accepted: 12/29/2018] [Indexed: 12/12/2022] Open
Abstract
A functional microvascular system is imperative to build and maintain healthy tissue. Impaired microvasculature results in ischemia, thereby limiting the tissue's intrinsic regeneration capacity. Therefore, the ability to regenerate microvascular networks is key to the development of effective cardiovascular therapies. To stimulate the formation of new microvasculature, researchers have focused on fabricating materials that mimic the angiogenic properties of the native extracellular matrix (ECM). Here, we will review biomaterials that seek to imitate the physical cues that are natively provided by the ECM to encourage the formation of microvasculature in engineered constructs and ischemic tissue in the body.
Collapse
Affiliation(s)
- Cody O Crosby
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
37
|
Ma W, Wang S, Liu X, Tang F, Zhao P, Cheng K, Zheng Q, Zhuo Y, Zhao X, Li X, Feng W. Protective effect of troxerutin and cerebroprotein hydrolysate injection on cerebral ischemia through inhibition of oxidative stress and promotion of angiogenesis in rats. Mol Med Rep 2019; 19:3148-3158. [PMID: 30816516 PMCID: PMC6423560 DOI: 10.3892/mmr.2019.9960] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 01/25/2019] [Indexed: 11/06/2022] Open
Abstract
Brain ischemia, including cerebral ischemia and cerebrovascular ischemia, leads to poor oxygen supply or cerebral hypoxia, and causes brain tissue death or cerebral infarction/ischemic stroke. The troxerutin and cerebroprotein hydrolysate injection (TCHI), is widely applied in China to improve blood supply in ischemic brain tissues and to enhance neuroprotective effects in clinical practice. However, the benefits and detailed underlying mechanism elaborating the effectiveness of TCHI in cerebrovascular diseases require further investigation. Therefore, in the present study, experimental in vivo and in vitro models were employed to investigate the potential mechanisms of TCHI on cerebral ischemic injury. The results demonstrated that TCHI increased the lactate dehydrogenase levels in the brain homogenate and conversely decreased lactic acid levels. TCHI was further observed to significantly increase superoxide dismutase activity and decrease malondialdehyde levels in ischemic brain tissues. In addition, TCHI significantly induced vascular maturation processes, including proliferation, adhesion, migration and tube formation in cultured human umbilical vein endothelial cells. Additionally, TCHI significantly stimulated microvessel formation in the rat aortic ring and chick chorioallantoic membrane assays. Taken together, these results provided strong evidence that TCHI stimulated angiogenesis at multiple steps, and indicated that TCHI attenuated cerebral ischemic damage through the amelioration of oxidative stress and promotion of angiogenesis.
Collapse
Affiliation(s)
- Wenbing Ma
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shixiang Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Xuanlin Liu
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fengru Tang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Peipei Zhao
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Kai Cheng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qiaowei Zheng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yingchen Zhuo
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xue Zhao
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xueqian Li
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Weiyi Feng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
38
|
Szewczyk G, Maciejewski TM, Szukiewicz D. Current progress in the inflammatory background of angiogenesis in gynecological cancers. Inflamm Res 2019; 68:247-260. [PMID: 30680411 PMCID: PMC6420455 DOI: 10.1007/s00011-019-01215-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 01/10/2019] [Accepted: 01/14/2019] [Indexed: 12/17/2022] Open
Abstract
A tumor growth depends on the potency of the tumor to support itself with nutrients and oxygen. The development of a vascular network within the tumor is key to its survival. The permanent contest between the tumor and its host involves tumor cells on one side and an immunological system and tissue stroma on the other. The angiogenesis is not only a specialty of the tumor, but it also depends on this complex multidirectional interaction. The most common gynecological cancers, cervical, endometrial and ovarian carcinoma are good examples for studying this problem. In this review, we aim to show that an inflammatory response against a tumor can be reverted into an undesirable process leading to the development of a vascular network within the tumor and, subsequently, further growth of the tumor and progression of a disease. Therefore, a key for tumor management should be searched within the immunological system, rather than focused on cell cycle and anti-angiogenic treatment only.
Collapse
Affiliation(s)
- Grzegorz Szewczyk
- Chair and Department of General and Experimental Pathology, Medical University of Warsaw, ul. Pawinskiego 3C, 02-106, Warsaw, Poland.
| | - Tomasz M Maciejewski
- Department of Gynecology and Obstetrics, Institute of Mother and Child, ul. Kasprzaka 17A, 01-211, Warsaw, Poland
| | - Dariusz Szukiewicz
- Chair and Department of General and Experimental Pathology, Medical University of Warsaw, ul. Pawinskiego 3C, 02-106, Warsaw, Poland
| |
Collapse
|
39
|
Diaz C, Neubauer S, Rechenmacher F, Kessler H, Missirlis D. Recruitment of integrin ανβ3 to integrin α5β1-induced clusters enables focal adhesion maturation and cell spreading. J Cell Sci 2019; 133:jcs.232702. [DOI: 10.1242/jcs.232702] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022] Open
Abstract
The major fibronectin (FN) binding integrins α5β1 and αvβ3 exhibit cooperativity during cell adhesion, migration and mechanosensing, through mechanisms that are not yet fully resolved. Exploiting mechanically-tunable, nano-patterned substrates, and peptidomimetic ligands designed to selectively bind corresponding integrins, we report that focal adhesions (FAs) of endothelial cells assembled on integrin α5β1-selective substrates, rapidly recruit αvβ3 integrins, but not vice versa. Blocking of integrin αvβ3 hindered FA maturation and cell spreading on α5β1-selective substrates, indicating a mechanism dependent on extracellular ligand binding and highlighting the requirement of αvβ3 engagement for efficient adhesion. Recruitment of αvβ3 integrins additionally occurred on hydrogel substrates of varying mechanical properties, above a threshold stiffness supporting FA formation. Mechanistic studies revealed the need for soluble factors present in serum to allow recruitment, and excluded exogenous, or endogenous, FN as the responsible ligand for integrin αvβ3 accumulation to adhesion clusters. Our findings highlight a novel mechanism of integrin co-operation and the critical role for αvβ3 integrins in promoting cell adhesion on α5β1-selective substrates.
Collapse
Affiliation(s)
- Carolina Diaz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research; postal address: Jahnstr. 29, D-69120, Heidelberg, Germany
- Department of Biophysical Chemistry, Physical Chemistry Institute, Heidelberg University; postal address: INF 253, D-69120 Heidelberg, Germany
| | - Stefanie Neubauer
- Institute for Advanced Study and Center for Integrated Protein Science (CIPSM), Technische Universität München; postal address: Lichtenbergstr. 4, D-85747, Garching, Germany
| | - Florian Rechenmacher
- Institute for Advanced Study and Center for Integrated Protein Science (CIPSM), Technische Universität München; postal address: Lichtenbergstr. 4, D-85747, Garching, Germany
| | - Horst Kessler
- Institute for Advanced Study and Center for Integrated Protein Science (CIPSM), Technische Universität München; postal address: Lichtenbergstr. 4, D-85747, Garching, Germany
| | - Dimitris Missirlis
- Department of Cellular Biophysics, Max Planck Institute for Medical Research; postal address: Jahnstr. 29, D-69120, Heidelberg, Germany
- Department of Biophysical Chemistry, Physical Chemistry Institute, Heidelberg University; postal address: INF 253, D-69120 Heidelberg, Germany
| |
Collapse
|
40
|
Hassanisaber H, Jafari L, Campeau MA, Drevelle O, Lauzon MA, Langelier E, Faucheux N, Rouleau L. The effect of substrate bulk stiffness on focal and fibrillar adhesion formation in human abdominal aortic endothelial cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 98:572-583. [PMID: 30813060 DOI: 10.1016/j.msec.2018.12.130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 12/07/2018] [Accepted: 12/28/2018] [Indexed: 01/01/2023]
Abstract
Endothelial cell (EC) dysfunction contributes to atherosclerosis, which is associated with arterial stiffening and fibronectin (FN) deposition, by ECs and smooth muscle cells (SMCs). The effect of stiffness on the EC/FN interaction and fibrillar adhesion formation has been poorly studied. An in vitro model was prepared that included FN-coated polydimethylsiloxane (PDMS) films with similar hydrophobicity and roughness but distinct Young's modulus values, mimicking healthy (1.0 MPa) and atherosclerotic (2.8 MPa) arteries. Human aortic abdominal endothelial cells (HAAECs) seeded on 1.0 MPa PDMS films spread over time and reached their maximum surface area faster than on 2.8 MPa PDMS films. In addition, HAAECs appeared to organize focal adhesion more rapidly on 1.0 MPa PDMS films, despite the similar cell binding domain accessibility to adsorbed FN. Interestingly, we also observed up to a ~5-fold increase in the percentage of HAAECs that had a well-developed fibrillar adhesion on 1.0 MPa compared to 2.8 MPa PDMS films as verified by integrin α5 subunits, tensin, and FN staining. This variation did not affect EC migration. These results suggest that there are favourable conditions for FN matrix assembly by ECs in early atherosclerosis rather than at advanced stages. Our in vitro model will therefore be helpful to understand the influence of bulk stiffness on cells involved in atherosclerosis.
Collapse
Affiliation(s)
- H Hassanisaber
- Cell-biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec, J1K 2R1, Canada.
| | - L Jafari
- Department of Mechanical Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec, J1K 2R1, Canada.
| | - M A Campeau
- Department of Chemical Engineering, McGill University, 3610 Rue University, Montréal, Québec, H3A 0C5, Canada.
| | - O Drevelle
- Cell-biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec, J1K 2R1, Canada; Thermal Plasma and Nanomaterial Synthesis Laboratory, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec J1K 2R1, Canada.
| | - M-A Lauzon
- 3D Cell Culture Systems Laboratory, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec J1K 2R1, Canada.
| | - E Langelier
- Department of Mechanical Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec, J1K 2R1, Canada; Clinical Research Centre of Centre Hospitalier Universitaire de Sherbrooke, 12e Avenue N, Sherbrooke, Québec J1H 5N4, Canada.
| | - N Faucheux
- Cell-biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec, J1K 2R1, Canada; Clinical Research Centre of Centre Hospitalier Universitaire de Sherbrooke, 12e Avenue N, Sherbrooke, Québec J1H 5N4, Canada.
| | - L Rouleau
- Department of Mechanical Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec, J1K 2R1, Canada.
| |
Collapse
|
41
|
Du P, Casavitri C, Suhaeri M, Wang PY, Lee JH, Koh WG, Park K. A Fibrous Hybrid Patch Couples Cell-Derived Matrix and Poly(l-lactide-co-caprolactone) for Endothelial Cells Delivery and Skin Wound Repair. ACS Biomater Sci Eng 2018; 5:900-910. [DOI: 10.1021/acsbiomaterials.8b01118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Ping Du
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Center for Human Tissues & Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Cininta Casavitri
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Muhammad Suhaeri
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Peng-Yuan Wang
- Center for Human Tissues & Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jong Ho Lee
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| |
Collapse
|
42
|
Izquierdo-Álvarez A, Vargas DA, Jorge-Peñas Á, Subramani R, Vaeyens MM, Van Oosterwyck H. Spatiotemporal Analyses of Cellular Tractions Describe Subcellular Effect of Substrate Stiffness and Coating. Ann Biomed Eng 2018; 47:624-637. [DOI: 10.1007/s10439-018-02164-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 10/29/2018] [Indexed: 12/21/2022]
|
43
|
Dias JR, Baptista-Silva S, Sousa A, Oliveira AL, Bártolo PJ, Granja PL. Biomechanical performance of hybrid electrospun structures for skin regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 93:816-827. [PMID: 30274117 DOI: 10.1016/j.msec.2018.08.050] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 08/03/2018] [Accepted: 08/22/2018] [Indexed: 12/19/2022]
Affiliation(s)
- J R Dias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Centre for Rapid and Sustainable Product Development (CDRsp), Polytechnic Institute of Leiria, Leiria, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.
| | - S Baptista-Silva
- CBQF - Center for Biotechnology and Fine Chemistry, School of Biotechnology, Portuguese Catholic University, Porto, Portugal
| | - A Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - A L Oliveira
- CBQF - Center for Biotechnology and Fine Chemistry, School of Biotechnology, Portuguese Catholic University, Porto, Portugal
| | - P J Bártolo
- School of Mechanical, Aerospace and Civil Engineering & Manchester Institute of Biotechnology, University of Manchester, UK
| | - P L Granja
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal; Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal
| |
Collapse
|
44
|
Kryza T, Parent C, Pardessus J, Petit A, Burlaud-Gaillard J, Reverdiau P, Iochmann S, Labas V, Courty Y, Heuzé-Vourc'h N. Human kallikrein-related peptidase 12 stimulates endothelial cell migration by remodeling the fibronectin matrix. Sci Rep 2018; 8:6331. [PMID: 29679011 PMCID: PMC5910384 DOI: 10.1038/s41598-018-24576-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/05/2018] [Indexed: 12/30/2022] Open
Abstract
Kallikrein-related peptidase 12 (KLK12) is a kallikrein family peptidase involved in angiogenesis - a complex biological process in which the sprouting, migration and stabilization of endothelial cells requires extracellular matrix remodeling. To characterize the molecular mechanisms associated with KLK12's proangiogenic activity, we evaluated its ability to hydrolyze various matrix proteins. Our results show that KLK12 efficiently cleaved the human extracellular matrix proteins fibronectin and tenascin, both of which are involved in the regulation of endothelial cell adhesion and migration. For fibronectin, the major proteolytic product generated by KLK12 was a 29 kDa fragment containing the amino-terminal domain and the first five type I fibronectin-domains, which are essential for regulating fibronectin assembly. We also demonstrated that KLK12-mediated fibronectin proteolysis antagonizes fibronectin polymerization and fibronectin fibril formation by endothelial cells, leading to an increase in cell migration. Furthermore, a polyclonal antibody raised against KLK12's proteolytic cleavage site on fibronectin prevented the KLK12-dependent inhibition of fibronectin polymerization and the KLK12-mediated pro-migratory effect on endothelial cells. Taken as a whole, our results indicate that KLK12's proangiogenic effect is mediated through several molecular mechanisms.
Collapse
Affiliation(s)
- T Kryza
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France.,Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Australia
| | - C Parent
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - J Pardessus
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - A Petit
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - J Burlaud-Gaillard
- Université François Rabelais de Tours, F-37032, Tours, France.,Plateforme IBiSA de Microscopie Electronique, Université François Rabelais de Tours, F-37032, Tours, France
| | - P Reverdiau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - S Iochmann
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - V Labas
- PRC, INRA, CNRS, Université François Rabelais de Tours, IFCE, F-37380, Nouzilly, France.,PAIB, CIRE, INRA, CHRU de Tours, Université François Rabelais de Tours, F-37380, Nouzilly, France
| | - Y Courty
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - N Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France. .,Université François Rabelais de Tours, F-37032, Tours, France.
| |
Collapse
|
45
|
Spaans S, Fransen PPKH, Ippel BD, de Bont DFA, Keizer HM, Bax NAM, Bouten CVC, Dankers PYW. Supramolecular surface functionalization via catechols for the improvement of cell-material interactions. Biomater Sci 2018. [PMID: 28636048 DOI: 10.1039/c7bm00407a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Optimization of cell-material interactions is crucial for the success of synthetic biomaterials in guiding tissue regeneration. To do so, catechol chemistry is often used to introduce adhesiveness into biomaterials. Here, a supramolecular approach based on ureido-pyrimidinone (UPy) modified polymers is combined with catechol chemistry in order to achieve improved cellular adhesion onto supramolecular biomaterials. UPy-modified hydrophobic polymers with non-cell adhesive properties are developed that can be bioactivated via a modular approach using UPy-modified catechols. It is shown that successful formulation of the UPy-catechol additive with the UPy-polymer results in surfaces that induce cardiomyocyte progenitor cell adhesion, cell spreading, and preservation of cardiac specific extracellular matrix production. Hence, by functionalizing supramolecular surfaces with catechol functionalities, an adhesive supramolecular biomaterial is developed that allows for the possibility to contribute to biomaterial-based regeneration.
Collapse
Affiliation(s)
- S Spaans
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - P P K H Fransen
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. box 513, 5600 MB Eindhoven, The Netherlands
| | - B D Ippel
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - D F A de Bont
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - H M Keizer
- SyMO-Chem BV, Eindhoven University of Technology, De Zaale, 5612 AZ Eindhoven, The Netherlands
| | - N A M Bax
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - C V C Bouten
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands
| | - P Y W Dankers
- Institute for Complex Molecular Systems, Eindhoven University of Technology, De Zaale, 5612 AJ Eindhoven, The Netherlands. and Department of Biomedical Engineering, Soft Tissue Engineering and Mechanobiology, Eindhoven University of Technology, P.O box 513, 5600 MB Eindhoven, The Netherlands and Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, P.O. box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
46
|
Torres A, Bidarra S, Pinto M, Aguiar P, Silva E, Barrias C. Guiding morphogenesis in cell-instructive microgels for therapeutic angiogenesis. Biomaterials 2018; 154:34-47. [DOI: 10.1016/j.biomaterials.2017.10.051] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 12/31/2022]
|
47
|
Kaessmeyer S, Sehl J, Khiao In M, Merle R, Richardson K, Plendl J. Subcellular Interactions during Vascular Morphogenesis in 3D Cocultures between Endothelial Cells and Fibroblasts. Int J Mol Sci 2017; 18:ijms18122590. [PMID: 29194374 PMCID: PMC5751193 DOI: 10.3390/ijms18122590] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/23/2017] [Accepted: 11/28/2017] [Indexed: 12/28/2022] Open
Abstract
Background: Increasing the complexity of in vitro systems to mimic three-dimensional tissues and the cellular interactions within them will increase the reliability of data that were previously collected with in vitro systems. In vivo vascularization is based on complex and clearly defined cell–matrix and cell–cell interactions, where the extracellular matrix (ECM) seems to play a very important role. The aim of this study was to monitor and visualize the subcellular and molecular interactions between endothelial cells (ECs), fibroblasts, and their surrounding microenvironment during vascular morphogenesis in a three-dimensional coculture model. Methods: Quantitative and qualitative analyses during the generation of a coculture tissue construct consisting of endothelial cells and fibroblasts were done using transmission electron microscopy and immunohistochemistry. Results: Dynamic interactions were found in cocultures between ECs, between fibroblasts (FBs), between ECs and FBs, and between the cells and the ECM. Microvesicles were involved in intercellular information transfer. FBs took an active and physical part in the angiogenesis process. The ECM deposited by the cells triggered endothelial angiogenic activity. Capillary-like tubular structures developed and matured. Moreover, some ECM assembled into a basement membrane (BM) having three different layers equivalent to those seen in vivo. Finally, the three-dimensional in vitro construct mirrored the topography of histological tissue sections. Conclusion: Our results visualize the importance of the physical contact between all cellular and acellular components of the cocultures.
Collapse
Affiliation(s)
- Sabine Kaessmeyer
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universitaet Berlin, Koserstraße 20, 14195 Berlin, Germany.
| | - Julia Sehl
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universitaet Berlin, Koserstraße 20, 14195 Berlin, Germany.
| | - Maneenooch Khiao In
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universitaet Berlin, Koserstraße 20, 14195 Berlin, Germany.
| | - Roswitha Merle
- Department of Veterinary Medicine, Institute of Veterinary Epidemiology and Biostatistics, Freie Universitaet Berlin, Koenigsweg 67, 14163 Berlin, Germany.
| | - Ken Richardson
- College of Veterinary Medicine, School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia.
| | - Johanna Plendl
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universitaet Berlin, Koserstraße 20, 14195 Berlin, Germany.
| |
Collapse
|
48
|
5-Aminolevulinic Acid Guided Sampling of Glioblastoma Microenvironments Identifies Pro-Survival Signaling at Infiltrative Margins. Sci Rep 2017; 7:15593. [PMID: 29142297 PMCID: PMC5688093 DOI: 10.1038/s41598-017-15849-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/31/2017] [Indexed: 11/14/2022] Open
Abstract
Glioblastoma (GBM) contains diverse microenvironments with uneven distributions of oncogenic alterations and signaling networks. The diffusely infiltrative properties of GBM result in residual tumor at neurosurgical resection margins, representing the source of relapse in nearly all cases and suggesting that therapeutic efforts should be focused there. To identify signaling networks and potential druggable targets across tumor microenvironments (TMEs), we utilized 5-ALA fluorescence-guided neurosurgical resection and sampling, followed by proteomic analysis of specific TMEs. Reverse phase protein array (RPPA) was performed on 205 proteins isolated from the tumor margin, tumor bulk, and perinecrotic regions of 13 previously untreated, clinically-annotated and genetically-defined high grade gliomas. Differential protein and pathway signatures were established and then validated using western blotting, immunohistochemistry, and comparable TCGA RPPA datasets. We identified 37 proteins differentially expressed across high-grade glioma TMEs. We demonstrate that tumor margins were characterized by pro-survival and anti-apoptotic proteins, whereas perinecrotic regions were enriched for pro-coagulant and DNA damage response proteins. In both our patient cohort and TCGA cases, the data suggest that TMEs possess distinct protein expression profiles that are biologically and therapeutically relevant.
Collapse
|
49
|
Increased synthesis and deposition of extracellular matrix proteins leads to endoplasmic reticulum stress in the trabecular meshwork. Sci Rep 2017; 7:14951. [PMID: 29097767 PMCID: PMC5668243 DOI: 10.1038/s41598-017-14938-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/18/2017] [Indexed: 12/15/2022] Open
Abstract
Increased synthesis and deposition of extracellular matrix (ECM) proteins in the trabecular meshwork (TM) is associated with TM dysfunction and intraocular pressure (IOP) elevation in glaucoma. However, it is not understood how ECM accumulation leads to TM dysfunction and IOP elevation. Using a mouse model of glucocorticoid (GC)-induced glaucoma, primary human TM cells and human post-mortem TM tissues, we show that increased ECM accumulation leads to endoplasmic reticulum (ER) stress in the TM. The potent GC, dexamethasone (Dex) increased the secretory protein load of ECM proteins in the ER of TM cells, inducing ER stress. Reduction of fibronectin, a major regulator of ECM structure, prevented ER stress in Dex-treated TM cells. Overexpression of fibronectin via treatment with cellular fibronectin also induced chronic ER stress in primary human TM cells. Primary human TM cells grown on ECM derived from Dex-treated TM cells induced ER stress markers. TM cells were more prone to ER stress from ECM accumulation compared to other ocular cell types. Moreover, increased co-localization of ECM proteins with ER stress markers was observed in human post-mortem glaucomatous TM tissues. These data indicate that ER stress is associated with increased ECM accumulation in mouse and human glaucomatous TM tissues.
Collapse
|
50
|
Dias J, Baptista-Silva S, Oliveira CD, Sousa A, Oliveira A, Bártolo P, Granja P. In situ crosslinked electrospun gelatin nanofibers for skin regeneration. Eur Polym J 2017. [DOI: 10.1016/j.eurpolymj.2017.08.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|