1
|
Agrawal N, Afzal M, Khan NH, Ganesan S, Kumari M, Sunitha S, Dash A, Goyal K, Kushwaha B, Rekha A, Rana M, Ali H. The role of VEGF in vascular dementia: impact of aging and cellular senescence. Biogerontology 2025; 26:77. [PMID: 40119956 DOI: 10.1007/s10522-025-10219-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Vascular Endothelial Growth Factor (VEGF) is a critical element in vascular dementia (VD) pathogenesis and therapeutic development while remaining strongly influenced by aging processes and cellular aging mechanisms. VEGF's multiple effects comprise neuroprotective functions, its role in vascular development, and its ability to regulate brain blood flow systems, all leading to cognitive preservation. The prefrontal cortex exhibits elevated VEGF gene levels, which directly matches the advancement of cognitive deficits in patients with Alzheimer's disease and VD. These patients exhibit higher VEGF levels in their CSF fluid, demonstrating that disease pathology includes multiple inseparable factors. Aging dramatically worsens VEGF regulation because endothelial dysfunction combines with chronic inflammation and oxidative stress to generate adverse vascular symptoms that include atherosclerosis and stroke. Cellular senescence convolutes these processes by causing damaging inflammatory reactions alongside impaired vascular healing abilities. The secretion of pro-inflammatory cytokines from senescent cells (SCs) disrupts VEGF signaling and produces harmful consequences for both vascular health and cognitive well-being. The neuroprotective properties of VEGF-A165a and VEGF-A165b variants demonstrate their ability to lessen β-amyloid and tau protein toxicity. The protective mechanisms of VEGF depend heavily on VEGF expression levels and receptor functionality, both of which decrease with aging. The combination of approaches that modulate VEGF signaling and SC accumulation shows potential for designing treatments against VD. People can sustain BBB functionality over a longer period through Mediterranean diet consumption together with aerobic exercise along with developing therapies, including senolytics and senomorphics, which delay neurodegenerative progression. Future investigative efforts must improve VEGF delivery methods while studying cellular senescence mechanisms and developing advanced methods to detect SC cells. A three-dimensional healthcare approach combining VEGF-targeted treatments with anti-ageing interventions and detailed diagnostic techniques shows the potential for effective VD management to achieve better patient results.
Collapse
Affiliation(s)
- Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - S Sunitha
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Aniruddh Dash
- Department of Orthopaedics, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
| | - Brajgopal Kushwaha
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, 462044, India
| | - A Rekha
- Hospital and Research Centre, Dr. D. Y. Patil Medical College, Pimpri, Pune, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College,, Saveetha University, Chennai, India
| |
Collapse
|
2
|
Ballato M, Germanà E, Ricciardi G, Giordano WG, Tralongo P, Buccarelli M, Castellani G, Ricci-Vitiani L, D’Alessandris QG, Giuffrè G, Pizzimenti C, Fiorentino V, Zuccalà V, Ieni A, Caffo M, Fadda G, Martini M. Understanding Neovascularization in Glioblastoma: Insights from the Current Literature. Int J Mol Sci 2025; 26:2763. [PMID: 40141406 PMCID: PMC11943220 DOI: 10.3390/ijms26062763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastomas (GBMs), among the most aggressive and resilient brain tumors, characteristically exhibit high angiogenic potential, leading to the formation of a dense yet aberrant vasculature, both morphologically and functionally. With these premises, numerous expectations were initially placed on anti-angiogenic therapies, soon dashed by their limited efficacy in concretely improving patient outcomes. Neovascularization in GBM soon emerged as a complex, dynamic, and heterogeneous process, hard to manage with the classical standard of care. Growing evidence has revealed the existence of numerous non-canonical strategies of angiogenesis, variously exploited by GBM to meet its ever-increasing metabolic demand and differently involved in tumor progression, recurrence, and escape from treatments. In this review, we provide an accurate description of each neovascularization mode encountered in GBM tumors to date, highlighting the molecular players and signaling cascades primarily involved. We also detail the key architectural and functional aspects characteristic of the GBM vascular compartment because of an intricate crosstalk between the different angiogenic networks. Additionally, we explore the repertoire of emerging therapies against GBM that are currently under study, concluding with a question: faced with such a challenging scenario, could combined therapies, tailored to the patient's genetic signatures, represent an effective game changer?
Collapse
Affiliation(s)
- Mariagiovanna Ballato
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Emanuela Germanà
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Gabriele Ricciardi
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
- Istituto Clinico Polispecialistico C.O.T. Cure Ortopediche Traumatologiche s.pa., 98124 Messina, Italy
| | - Walter Giuseppe Giordano
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Pietro Tralongo
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.)
| | - Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.)
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.)
| | | | - Giuseppe Giuffrè
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | | | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Valeria Zuccalà
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Maria Caffo
- Biomedical and Dental Sciences and Morphofunctional Imaging, Unit of Neurosurgery, University of Messina, 98122 Messina, Italy;
| | - Guido Fadda
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| |
Collapse
|
3
|
Zhang Z, Ding Y, Yuan H, Rui C, Fan P, Ji Y, Xiao Y, Dai J, Li L. A multiple-crosslinked injectable hydrogel for modulating tissue microenvironment and accelerating infected diabetic wound repair. J Nanobiotechnology 2025; 23:218. [PMID: 40102884 PMCID: PMC11917161 DOI: 10.1186/s12951-025-03285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/02/2025] [Indexed: 03/20/2025] Open
Abstract
Elevated oxidative stress and inflammation, bacterial infections, and vascular impairment undoubtedly impede the normal diabetic wound healing process, which has encouraged the development of high-performance dressings for wound management. Herein, a new type of multiple-crosslinked injectable hydrogel, GCP, was developed via the radical polymerization of propenyl groups and the formation of copper‒polyphenol coordination bonds and Schiff base bonds. The copper‒polyphenol coordination and Schiff base bonds in the GCP hydrogel were disrupted in the acidic microenvironment of diabetic wound, resulting in the release of copper ions and protocatechualdehyde (PA) to scavenge reactive oxygen species (ROS), promote angiogenesis and cell migration, and exert antibacterial and anti-inflammatory activities via the CuPA complexes. Consequently, markedly accelerated infected diabetic wounds healing was achieved through this tissue microenvironment remodeling strategy. Moreover, the underlying mechanism of the antibacterial properties was investigated by 16S rRNA sequencing. The results indicated that the CuPA complexes can clearly inhibit the growth and reproduction of S. aureus by downregulating specific genes associated with ABC transporters, hindering bacterial protein synthesis, and enhancing oxidoreductase activity. This innovative hydrogel platform for wound management may inspire new methods for the preparation of high-performance biomedical materials and the treatment of other clinical diseases.
Collapse
Affiliation(s)
- Zhengduo Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yuanyuan Ding
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Huipu Yuan
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Chen Rui
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Pengfei Fan
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yinwen Ji
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Ying Xiao
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China.
| | - Jiayong Dai
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, China.
| | - Lei Li
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China.
| |
Collapse
|
4
|
Aksan B, Mauceri D. Beyond vessels: unraveling the impact of VEGFs on neuronal functions and structure. J Biomed Sci 2025; 32:33. [PMID: 40050849 PMCID: PMC11884128 DOI: 10.1186/s12929-025-01128-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/21/2025] [Indexed: 03/10/2025] Open
Abstract
Neurons rely on the bloodstream for essential nutrients and oxygen, which is facilitated by an intricate coupling of the neuronal and vascular systems. Central to this neurovascular interaction is the vascular endothelial growth factor (VEGF) family, a group of secreted growth factors traditionally known for their roles in promoting endothelial cell proliferation, migration, and survival in the cardiovascular and lymphatic systems. However, emerging evidence shows that VEGFs also play indispensable roles in the nervous system, extending beyond their canonical angiogenic and lymphangiogenic functions. Over the past two decades, VEGFs have been found to exert direct effects on neurons, influencing key aspects of neuronal function independently of their actions on vascular cells. In particular, it has become increasingly evident that VEGFs also play crucial functions in the development, regulation, and maintenance of neuronal morphology. Understanding the roles of VEGFs in neuronal development is of high scientific and clinical interest because of the significance of precise neuronal morphology for neural connectivity and network function, as well as the association of morphological abnormalities with neurological and neurodegenerative disorders. This review begins with an overview of the VEGF family members, their structural characteristics, receptors, and established roles in vasculature. However, it then highlights and focuses on the exciting variety of neuronal functions of VEGFs, especially their crucial role in the development, regulation, and maintenance of neuronal morphology.
Collapse
Affiliation(s)
- Bahar Aksan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany.
- Institute of Anatomy and Cell Biology, Dept. Molecular and Cellular Neuroscience, University of Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
5
|
Dias ES, Freitas LC, Miyazawa M, Nogueira DA, Oliveira CE, Pereira AA, Hanemann JA. Vascular endothelial growth factor immunoexpression in oral paracoccidioidomycosis. Med Oral Patol Oral Cir Bucal 2025; 30:e240-e246. [PMID: 39864089 PMCID: PMC11972650 DOI: 10.4317/medoral.26886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Paracoccidioidomycosis (PCM) is a systemic mycosis endemic and limited to Latin America. Brazil is responsible for more than 80% of diagnosed cases in the world. Since PCM is not a notifiable disease, there are still no accurate data on its incidence in Brazil. Vascular endothelial growth factor (VEGF) is considered the main vascular endothelial growth factor expressed in the process of angiogenesis, both under physiological and pathological conditions. To date, there are not studies in the literature that evaluated the expression of VEGF in oral PCM lesions. Therefore, the objective of this study was to evaluate the prevalence of oral lesions of PCM diagnosed from 1998 to 2020; to analyze the immunoexpression of VEGF in oral lesions of PCM; and to compare the VEGF immunostaining with the clinical and microscopic aspects of these lesions. MATERIAL AND METHODS Clinical data of 98 cases of patients with oral PCM were evaluated. A total of 41 selected cases were quantitatively and qualitatively analysed by immunohistochemistry for VEGF. RESULTS Our results showed that oral PCM preferentially affects white males, with mean age of 50.2 years, and the gingiva and the alveolar ridge. It was not possible to correlate VEGF immunoexpression with clinical and microscopic variables. CONCLUSIONS Oral PCM is a relatively uncommon pathological condition and that, in our sample, the immunoexpression of VEGF was mild and observed in a reduced number of cases.
Collapse
Affiliation(s)
- E-S Dias
- School of Dentistry. Federal University of Alfenas Rua Gabriel Monteiro da Silva, 700. Centro 37130.001. Alfenas, Minas Gerais, Brazil
| | | | | | | | | | | | | |
Collapse
|
6
|
Liu J, Wu S, Zhang L, Zhang M, Wu H, Shi M, Weng Y, Ye W, Gu K. A novel electrochemical immunosensor for the ultrasensitive detection of vascular endothelial growth factor based on MB@Cu-MOFs/MWCNTs composite. Mikrochim Acta 2024; 192:37. [PMID: 39730872 DOI: 10.1007/s00604-024-06884-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 12/06/2024] [Indexed: 12/29/2024]
Abstract
A novel proposal is introduced with an unlabeled electrochemical immunosensor for the detection of tumor broad-spectrum biomarker vascular endothelial growth factor (VEGF165) Copper-based metal organic frameworks (Cu MOFs)-carbon nanotubes (MWCNTs) were employed as its substrates, functionalized with methylene blue (MB) for signal enhancement. Cu-MOFs-MWCNTs nanocomposites were synthesized successfully via a solvothermal method and were then deposited on the surface of a glassy carbon electrode (GCE), with the addition of methylene blue to amplify the signal. Due to the expansive specific surface area provided by the carbon nanotubes and the amino groups facilitated by the metal-organic framework nanomaterials, the anti-VEGF165 monoclonal antibody was immobilized on the electrochemical immunosensor through covalent bonding, which could bind specifically to VEGF165, thereby causing a detectable change in the current. The developed immunosensor exhibited a linear detection range for VEGF165 spanning from 1 to 100,000 pg/ml, with the R2 value of 0.997 by differential pulse voltammetry (DPV) method, and the limit of detection was as low as 0.9686. Additionally, the electrochemical immunosensor presented commendable selectivity, availability, and reproducibility, along with an effective capacity for detecting VEGF165 in clinical samples. Consequently, the immunosensor is expected to revolutionize early screening for VEGF165-related diseases, which offer a promising, cost-effective, and precise diagnostic tool for clinical utility.
Collapse
Affiliation(s)
- Junjie Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Shengxi Wu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
| | - Lu Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Mingjun Zhang
- Chongqing Jiulongpo District People's Hospital, Chongqing, 400050, China
| | - Haotian Wu
- Department of Physics and Energy, Chongqing University of Technology, Chongqing, 400054, China
| | - Miwan Shi
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yuan Weng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Wenqiao Ye
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Kui Gu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
| |
Collapse
|
7
|
Zhang J, Wang S, Sun Q, Zhang J, Shi X, Yao M, Chen J, Huang Q, Zhang G, Huang Q, Ai K, Bai Y. Peroxynitrite-Free Nitric Oxide-Embedded Nanoparticles Maintain Nitric Oxide Homeostasis for Effective Revascularization of Myocardial Infarcts. ACS NANO 2024; 18:32650-32671. [PMID: 39545833 DOI: 10.1021/acsnano.4c10118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Revascularization is crucial for treating myocardial infarction (MI). Nitric oxide (NO), at an appropriate concentration, is recognized as an ideal and potent pro-angiogenic factor. However, the application of NO in the treatment of MI is limited. Improper NO supplementation is harmful to revascularization because NO is converted into harmful peroxynitrite (ONOO-) in MI tissues with high reactive oxygen species (ROS) levels. We overcome these obstacles by embedding biliverdin and NO into Prussian blue (PB) nanolattices to obtain an ONOO--free NO-embedded nanomedicine (OFEN). Unlike previous NO donors, OFEN provides NO stably and spontaneously for a longer time (>7 days), which makes it possible to maintain a stable concentration of NO, suitable for angiogenesis, through dose optimization. More importantly, based on the synergy between PB and biliverdin, OFEN converts ROS into beneficial O2 and inhibits the production of ONOO- from the source. OFEN specifically targets MI tissues and achieves sustained and stable NO delivery at the MI site. OFEN effectively promotes revascularization in the MI tissue, significantly reduces myocardial death and fibrosis, and ultimately promotes the complete recovery of cardiac function. Our strategy provides a promising approach for the treatment of myocardial and other ischemic diseases.
Collapse
Affiliation(s)
- Jiaxiong Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Shuya Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Quan Sun
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jian Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Xiaojing Shi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Meilian Yao
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jing Chen
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Guogang Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, PR China
| | - Qun Huang
- Department of Child Health Care, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, PR China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Yongping Bai
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| |
Collapse
|
8
|
Yin W, Chen Y, Wang W, Guo M, Tong L, Zhang M, Wang Z, Yuan H. Macrophage-mediated heart repair and remodeling: A promising therapeutic target for post-myocardial infarction heart failure. J Cell Physiol 2024; 239:e31372. [PMID: 39014935 DOI: 10.1002/jcp.31372] [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: 03/04/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Heart failure (HF) remains prevalent in patients who survived myocardial infarction (MI). Despite the accessibility of the primary percutaneous coronary intervention and medications that alleviate ventricular remodeling with functional improvement, there is an urgent need for clinicians and basic scientists to further reveal the mechanisms behind post-MI HF as well as investigate earlier and more efficient treatment after MI. Growing numbers of studies have highlighted the crucial role of macrophages in cardiac repair and remodeling following MI, and timely intervention targeting the immune response via macrophages may represent a promising therapeutic avenue. Recently, technology such as single-cell sequencing has provided us with an updated and in-depth understanding of the role of macrophages in MI. Meanwhile, the development of biomaterials has made it possible for macrophage-targeted therapy. Thus, an overall and thorough understanding of the role of macrophages in post-MI HF and the current development status of macrophage-based therapy will assist in the further study and development of macrophage-targeted treatment for post-infarction cardiac remodeling. This review synthesizes the spatiotemporal dynamics, function, mechanism and signaling of macrophages in the process of HF after MI, as well as discusses the emerging bio-materials and possible therapeutic agents targeting macrophages for post-MI HF.
Collapse
Affiliation(s)
- Wenchao Yin
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yong Chen
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wenjun Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mengqi Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lingjun Tong
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zhaoyang Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
9
|
Kreiner P, Eggenhofer E, Schneider L, Rejas C, Goetz M, Bogovic N, Brunner SM, Evert K, Schlitt HJ, Geissler EK, Junger H. Extrahepatic Bile Duct Organoids as a Model to Study Ischemia/Reperfusion Injury During Liver Transplantation. Transpl Int 2024; 37:13212. [PMID: 39323909 PMCID: PMC11422091 DOI: 10.3389/ti.2024.13212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
Biliary complications are still a major cause for morbidity and mortality after liver transplantation (LT). Ischemia/reperfusion injury (IRI) leads to disruption of the biliary epithelium. We introduce a novel model to study the effect of IRI on human cholangiocytes using extrahepatic cholangiocyte organoids (ECOs). Extrahepatic bile duct tissue was collected during LT at static cold storage and after reperfusion (n = 15); gallbladder tissue was used for controls (n = 5). ECOs (n = 9) were cultured from extrahepatic biliary tissue, with IRI induced in an atmosphere of 95% air (nitrogen), 1% O2 and 5% CO2for 48 h, followed by 24 h of reoxygenation. Qualitative and quantitative histology and qRT-PCR were performed to discern phenotype, markers of hypoxia, programmed cell death and proliferation. ECOs self-organized into circular structures resembling biliary architecture containing cholangiocytes that expressed EpCAM, CK19, LGR5 and SOX-9. After hypoxia, ECOs showed increased expression of VEGF A (p < 0.0001), SLC2A1 (p < 0.0001) and ACSL4 (p < 0.0001) to indicate response to hypoxic damage and subsequent programmed cell death. Increase in cyclin D1 (p < 0.0001) after reoxygenation indicated proliferative activity in ECOs. Therefore, ECO structure and response to IRI are comparable to that found in-vivo, providing a suitable model to study IRI of the bile duct in-vitro.
Collapse
Affiliation(s)
- P Kreiner
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - E Eggenhofer
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - L Schneider
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - C Rejas
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - M Goetz
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - N Bogovic
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - S M Brunner
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - K Evert
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - H J Schlitt
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - E K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - H Junger
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
10
|
Libby JR, Royce H, Walker SR, Li L. The role of extracellular matrix in angiogenesis: Beyond adhesion and structure. BIOMATERIALS AND BIOSYSTEMS 2024; 15:100097. [PMID: 39129826 PMCID: PMC11315062 DOI: 10.1016/j.bbiosy.2024.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/19/2024] [Accepted: 07/06/2024] [Indexed: 08/13/2024] Open
Abstract
While the extracellular matrix (ECM) has long been recognized for its structural contributions, anchoring cells for adhesion, providing mechanical support, and maintaining tissue integrity, recent efforts have elucidated its dynamic, reciprocal, and diverse properties on angiogenesis. The ECM modulates angiogenic signaling and mechanical transduction, influences the extent and degree of receptor activation, controls cellular behaviors, and serves as a reservoir for bioactive macromolecules. Collectively, these factors guide the formation, maturation, and stabilization of a functional vascular network. This review aims to shed light on the versatile roles of the ECM in angiogenesis, transcending its traditional functions as a mere structural material. We will explore its engagement and synergy in signaling modulation, interactions with various angiogenic factors, and highlight its importance in both health and disease. By capturing the essence of the ECM's diverse functionalities, we highlight the significance in the broader context of vascular biology, enabling the design of novel biomaterials to engineer vascularized tissues and their potential therapeutic implications.
Collapse
Affiliation(s)
- Jaxson R. Libby
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Haley Royce
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
| | - Sarah R. Walker
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Linqing Li
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
- Department of Chemistry, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
11
|
Toftdal MS, Christensen NP, Kadumudi FB, Dolatshahi-Pirouz A, Grunnet LG, Chen M. Mechanically reinforced hydrogel vehicle delivering angiogenic factor for beta cell therapy. J Colloid Interface Sci 2024; 667:54-63. [PMID: 38615623 DOI: 10.1016/j.jcis.2024.04.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/18/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic disease affecting millions worldwide. Insulin therapy is currently the golden standard for treating T1DM; however, it does not restore the normal glycaemic balance entirely, which increases the risk of secondary complications. Beta-cell therapy may be a possible way of curing T1DM and has already shown promising results in the clinic. However, low retention rates, poor cell survival, and limited therapeutic potential are ongoing challenges, thus increasing the need for better cell encapsulation devices. This study aimed to develop a mechanically reinforced vascular endothelial growth factor (VEGF)-delivering encapsulation device suitable for beta cell encapsulation and transplantation. Poly(l-lactide-co-ε-caprolactone) (PLCL)/gelatin methacryloyl (GelMA)/alginate coaxial nanofibres were produced using electrospinning and embedded in an alginate hydrogel. The encapsulation device was physically and biologically characterised and was found to be suitable for INS-1E beta cell encapsulation, vascularization, and transplantation in terms of its biocompatibility, porosity, swelling ratio and mechanical properties. Lastly, VEGF was incorporated into the hydrogel and the release kinetics and functional studies revealed a sustained release of bioactive VEGF for at least 14 days, making the modified alginate system a promising candidate for improving the beta cell survival after transplantation.
Collapse
Affiliation(s)
- Mette Steen Toftdal
- Department of Biological and Chemical Engineering, Aarhus University, 8000 Aarhus C, Denmark; Department of Cell Formulation and Delivery, Novo Nordisk A/S, 2760 Måløv, Denmark
| | | | - Firoz Babu Kadumudi
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | | | - Lars Groth Grunnet
- Department of Cell Formulation and Delivery, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Menglin Chen
- Department of Biological and Chemical Engineering, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
12
|
Arteel GE. Hepatic Extracellular Matrix and Its Role in the Regulation of Liver Phenotype. Semin Liver Dis 2024; 44:343-355. [PMID: 39191427 PMCID: PMC12057067 DOI: 10.1055/a-2404-7973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
The hepatic extracellular matrix (ECM) is most accurately depicted as a dynamic compartment that comprises a diverse range of players that work bidirectionally with hepatic cells to regulate overall homeostasis. Although the classic meaning of the ECM referred to only proteins directly involved in generating the ECM structure, such as collagens, proteoglycans, and glycoproteins, the definition of the ECM is now broader and includes all components associated with this compartment. The ECM is critical in mediating phenotype at the cellular, organ, and even organismal levels. The purpose of this review is to summarize the prevailing mechanisms by which ECM mediates hepatic phenotype and discuss the potential or established role of this compartment in the response to hepatic injury in the context of steatotic liver disease.
Collapse
Affiliation(s)
- Gavin E. Arteel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
13
|
Chiang H, Chung CA. Simulation of Soluble and Bound VEGF-stimulated in vitro Capillary-like Network Formation on Deformed Substrate. PLoS Comput Biol 2024; 20:e1012281. [PMID: 39038038 PMCID: PMC11262697 DOI: 10.1371/journal.pcbi.1012281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/26/2024] [Indexed: 07/24/2024] Open
Abstract
Capillary plexus cultivation is crucial in tissue engineering and regenerative medicine. Theoretical simulations have been conducted to supplement the expensive experimental works. However, the mechanisms connecting mechanical and chemical stimuli remained undefined, and the functions of the different VEGF forms in the culture environment were still unclear. In this paper, we developed a hybrid model for simulating short-term in vitro capillary incubations. We used the Cellular Potts model to predict individual cell migration, morphology change, and continuum mechanics to quantify biogel deformation and VEGF transport dynamics. By bridging the mechanical regulation and chemical stimulation in the model, the results showed good agreement between the predicted network topology and experiments, in which elongated cells connected, forming the network cords and round cells gathered, creating cobblestone-like aggregates. The results revealed that the capillary-like networks could develop in high integrity only when the mechanical and chemical couplings worked adequately, with the cell morphology and haptotaxis driven by the soluble and bound forms of VEGF, respectively, functioning simultaneously.
Collapse
Affiliation(s)
- Hsun Chiang
- Department of Mechanical Engineering, National Central University, Taoyuan, Taiwan
| | - Chih-Ang Chung
- Department of Mechanical Engineering, National Central University, Taoyuan, Taiwan
| |
Collapse
|
14
|
Verma M, Asakura Y, Wang X, Zhou K, Ünverdi M, Kann AP, Krauss RS, Asakura A. Endothelial cell signature in muscle stem cells validated by VEGFA-FLT1-AKT1 axis promoting survival of muscle stem cell. eLife 2024; 13:e73592. [PMID: 38842166 PMCID: PMC11216748 DOI: 10.7554/elife.73592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 06/05/2024] [Indexed: 06/07/2024] Open
Abstract
Endothelial and skeletal muscle lineages arise from common embryonic progenitors. Despite their shared developmental origin, adult endothelial cells (ECs) and muscle stem cells (MuSCs; satellite cells) have been thought to possess distinct gene signatures and signaling pathways. Here, we shift this paradigm by uncovering how adult MuSC behavior is affected by the expression of a subset of EC transcripts. We used several computational analyses including single-cell RNA-seq (scRNA-seq) to show that MuSCs express low levels of canonical EC markers in mice. We demonstrate that MuSC survival is regulated by one such prototypic endothelial signaling pathway (VEGFA-FLT1). Using pharmacological and genetic gain- and loss-of-function studies, we identify the FLT1-AKT1 axis as the key effector underlying VEGFA-mediated regulation of MuSC survival. All together, our data support that the VEGFA-FLT1-AKT1 pathway promotes MuSC survival during muscle regeneration, and highlights how the minor expression of select transcripts is sufficient for affecting cell behavior.
Collapse
Affiliation(s)
- Mayank Verma
- Department of Pediatrics & Neurology, Division of Pediatric Neurology, The University of Texas Southwestern Medical CenterDallasUnited States
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Yoko Asakura
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Xuerui Wang
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Kasey Zhou
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Mahmut Ünverdi
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Graduate School of Biomedical Sciencesf, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Graduate School of Biomedical Sciencesf, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Atsushi Asakura
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| |
Collapse
|
15
|
Ohayon S, Taib L, Verma NC, Iarossi M, Bhattacharya I, Marom B, Huttner D, Meller A. Full-Length Single Protein Molecules Tracking and Counting in Thin Silicon Channels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314319. [PMID: 38461367 DOI: 10.1002/adma.202314319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/25/2024] [Indexed: 03/11/2024]
Abstract
Emerging single-molecule protein sensing techniques are ushering in a transformative era in biomedical research. Nevertheless, challenges persist in realizing ultra-fast full-length protein sensing, including loss of molecular integrity due to protein fragmentation, biases introduced by antibodies affinity, identification of proteoforms, and low throughputs. Here, a single-molecule method for parallel protein separation and tracking is introduced, yielding multi-dimensional molecular properties used for their identification. Proteins are tagged by chemo-selective dual amino-acid specific labels and are electrophoretically separated by their mass/charge in custom-designed thin silicon channel with subwavelength height. This approach allows analysis of thousands of individual proteins within a few minutes by tracking their motion during the migration. The power of the method is demonstrated by quantifying a cytokine panel for host-response discrimination between viral and bacterial infections. Moreover, it is shown that two clinically-relevant splice isoforms of Vascular endothelial growth factor (VEGF) can be accurately quantified from human serum samples. Being non-destructive and compatible with full-length intact proteins, this method opens up ways for antibody-free single-protein molecule quantification.
Collapse
Affiliation(s)
- Shilo Ohayon
- Department of Biomedical Engineering, Technion-IIT, Haifa, 3200003, Israel
| | - Liran Taib
- Department of Biomedical Engineering, Technion-IIT, Haifa, 3200003, Israel
| | | | - Marzia Iarossi
- Department of Biomedical Engineering, Technion-IIT, Haifa, 3200003, Israel
| | - Ivy Bhattacharya
- Department of Biomedical Engineering, Technion-IIT, Haifa, 3200003, Israel
| | - Barak Marom
- Department of Biomedical Engineering, Technion-IIT, Haifa, 3200003, Israel
| | - Diana Huttner
- Department of Biomedical Engineering, Technion-IIT, Haifa, 3200003, Israel
| | - Amit Meller
- Department of Biomedical Engineering, Technion-IIT, Haifa, 3200003, Israel
- Russell Berrie Nanotechnology Institute, Technion-IIT, Haifa, 3200003, Israel
| |
Collapse
|
16
|
Miao X, Lin J, Li A, Gao T, Liu T, Shen J, Sun Y, Wei J, Bao B, Zheng X. AAV-mediated VEGFA overexpression promotes angiogenesis and recovery of locomotor function following spinal cord injury via PI3K/Akt signaling. Exp Neurol 2024; 375:114739. [PMID: 38401852 DOI: 10.1016/j.expneurol.2024.114739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/09/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
Spinal cord injury (SCI) is a disorder of the central nervous system resulting from various factors such as trauma, inflammation, tumors, and other etiologies. This condition leads to impairment in motor, sensory, and autonomic functions below the level of injury. Limitations of current therapeutic approaches prompt an investigation into therapeutic angiogenesis through persistent local expression of proangiogenic factors. Here, we investigated whether overexpression of adeno-associated virus (AAV)-mediated vascular endothelial growth factor A (VEGFA) in mouse SCI promoted locomotor function recovery, and whether the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway was mechanistically involved. Three weeks before SCI, AAV-VEGFA was injected at the T10 level to induce VEGFA overexpression. Neurofunctional, histological, and biochemical assessments were done to determine tissue damage and/or recovery of neuromuscular and behavioral impairments. Daily injections of the PI3K/Akt pathway inhibitor LY294002 were made to assess a possible mechanism. AAV-VEGFA overexpression dramatically improved locomotor function and ameliorated pathological injury caused by SCI. Improved motor-evoked potentials in hindlimbs and more spinal CD31-positive microvessels were observed in AAV-VEGFA-overexpressing mice. LY294002 reduced PI3K and Akt phosphorylation levels and attenuated AAV-VEGFA-related improvements. In conclusion, sustained local AAV-mediated VEGFA overexpression in spinal cord can significantly promote angiogenesis and ameliorate locomotor impairment after SCI in a contusion mouse model through activation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Xin Miao
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Junqing Lin
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Ang Li
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Tao Gao
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Tiexin Liu
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Junjie Shen
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Yi Sun
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Jiabao Wei
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Bingbo Bao
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Xianyou Zheng
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China.
| |
Collapse
|
17
|
Huang S, Zhang P, Yin N, Xu Z, Liu X, Wu A, Zhang X, Li Z, Zhang Z, Zhong T, Liu L, Shi Y, Dong J. Glioblastoma stem cell-derived exosomal miR-374b-3p promotes tumor angiogenesis and progression through inducing M2 macrophages polarization. iScience 2024; 27:109270. [PMID: 38487014 PMCID: PMC10937837 DOI: 10.1016/j.isci.2024.109270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/30/2024] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Glioblastoma stem cells (GSCs) reside in hypoxic periarteriolar niches of glioblastoma micro-environment, however, the crosstalk of GSCs with macrophages on regulating tumor angiogenesis and progression are not fully elucidated. GSCs-derived exosomes (GSCs-exos) are essential mediators during tumor immune-microenvironment remodeling initiated by GSCs, resulting in M2 polarization of tumor-associated macrophages (TAMs) as we reported previously. Our data disclosed aberrant upregulation of miR-374b-3p in both clinical glioblastoma specimens and human cell lines of GSCs. MiR-374b-3p level was high in GSCs-exos and can be internalized by macrophages. Mechanistically, GSCs exosomal miR-374b-3p induced M2 polarization of macrophages by downregulating phosphatase and tensin expression, thereby promoting migration and tube formation of vascular endothelial cells after coculture with M2 macrophages. Cumulatively, these data indicated that GSCs exosomal miR-374b-3p can enhance tumor angiogenesis by inducing M2 polarization of macrophages, as well as promote malignant progression of glioblastoma. Targeting exosomal miR-374b-3p may serve as a potential target against glioblastoma.
Collapse
Affiliation(s)
- Shilu Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Peng Zhang
- Department of Neurosurgery, Rugao People’s Hospital, RuGao 226500, China
| | - Nanheng Yin
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zhipeng Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Xinglei Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Anyi Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Xiaopei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zengyang Li
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zhicheng Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Tao Zhong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Shi
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| |
Collapse
|
18
|
Loh KM, Ang LT. Building human artery and vein endothelial cells from pluripotent stem cells, and enduring mysteries surrounding arteriovenous development. Semin Cell Dev Biol 2024; 155:62-75. [PMID: 37393122 DOI: 10.1016/j.semcdb.2023.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/07/2023] [Indexed: 07/03/2023]
Abstract
Owing to their manifold roles in health and disease, there have been intense efforts to synthetically generate blood vessels in vitro from human pluripotent stem cells (hPSCs). However, there are multiple types of blood vessel, including arteries and veins, which are molecularly and functionally different. How can we specifically generate either arterial or venous endothelial cells (ECs) from hPSCs in vitro? Here, we summarize how arterial or venous ECs arise during embryonic development. VEGF and NOTCH arbitrate the bifurcation of arterial vs. venous ECs in vivo. While manipulating these two signaling pathways biases hPSC differentiation towards arterial and venous identities, efficiently generating these two subtypes of ECs has remained challenging until recently. Numerous questions remain to be fully addressed. What is the complete identity, timing and combination of extracellular signals that specify arterial vs. venous identities? How do these extracellular signals intersect with fluid flow to modulate arteriovenous fate? What is a unified definition for endothelial progenitors or angioblasts, and when do arterial vs. venous potentials segregate? How can we regulate hPSC-derived arterial and venous ECs in vitro, and generate organ-specific ECs? In turn, answers to these questions could avail the production of arterial and venous ECs from hPSCs, accelerating vascular research, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Kyle M Loh
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA.
| | - Lay Teng Ang
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
19
|
Jiang H, Li X, Chen T, Liu Y, Wang Q, Wang Z, Jia J. Bioprinted vascular tissue: Assessing functions from cellular, tissue to organ levels. Mater Today Bio 2023; 23:100846. [PMID: 37953757 PMCID: PMC10632537 DOI: 10.1016/j.mtbio.2023.100846] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/21/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023] Open
Abstract
3D bioprinting technology is widely used to fabricate various tissue structures. However, the absence of vessels hampers the ability of bioprinted tissues to receive oxygen and nutrients as well as to remove wastes, leading to a significant reduction in their survival rate. Despite the advancements in bioinks and bioprinting technologies, bioprinted vascular structures continue to be unsuitable for transplantation compared to natural blood vessels. In addition, a complete assessment index system for evaluating the structure and function of bioprinted vessels in vitro has not yet been established. Therefore, in this review, we firstly highlight the significance of selecting suitable bioinks and bioprinting techniques as they two synergize with each other. Subsequently, focusing on both vascular-associated cells and vascular tissues, we provide a relatively thorough assessment of the functions of bioprinted vascular tissue based on the physiological functions that natural blood vessels possess. We end with a review of the applications of vascular models, such as vessel-on-a-chip, in simulating pathological processes and conducting drug screening at the organ level. We believe that the development of fully functional blood vessels will soon make great contributions to tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Haihong Jiang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xueyi Li
- Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| | - Tianhong Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Yang Liu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qian Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Zhimin Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China
| | - Jia Jia
- School of Life Sciences, Shanghai University, Shanghai, China
- Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| |
Collapse
|
20
|
Haba D, Ohmiya T, Sekino M, Qin Q, Takizawa C, Tomida S, Minematsu T, Sanada H, Nakagami G. Efficacy of wearable vibration dressings on full-thickness wound healing in a hyperglycemic rat model. Wound Repair Regen 2023; 31:816-826. [PMID: 37950849 DOI: 10.1111/wrr.13129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/13/2023]
Abstract
Local low-frequency vibration promotes blood flow and wound healing in hard-to-heal diabetic foot ulcers (DFUs). However, vibration treatment is challenging in patients with DFUs due to wound management difficulties and low adherence. Consequently, developing wearable self-care devices becomes imperative for effective wound healing. This study introduces a wearable vibration dressing and assesses its impact on wound healing in hyperglycemic rats. Low-frequency vibration at 52 Hz was applied to the wound for 40 min/day in awake rats. Relative wound areas on post-wounding days (PWDs) 4-7 were significantly smaller and the wound closure rate was significantly higher in the vibration group than in the control group (p < 0.05, respectively). The total haemoglobin at baseline and after vibration on post-wounding day 7 was significantly larger in the vibration group than in the control group (p < 0.05). On PWD 7, the thickness of the granulation tissue was significantly higher in the vibration group than in the control group (p < 0.05). Moreover, the number of blood vessels at the wound site and vascular endothelial growth factor A protein expression were significantly higher in the vibration group than in the control group (p < 0.05, respectively). The ratio of (CD68+ /iNOS+ )/(CD163+ ) macrophages in the vibration group was significantly lower than that in the control group (p < 0.05). These results indicate the potential of wearable vibration dressings as new self-care devices that can promote angiogenesis and blood flow, improve inflammation, and enhance wound healing in DFUs.
Collapse
Affiliation(s)
- Daijiro Haba
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takafumi Ohmiya
- Department of Electrical Engineering and Information Systems, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Masaki Sekino
- Department of Electrical Engineering and Information Systems, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Qi Qin
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Chihiro Takizawa
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sanai Tomida
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeo Minematsu
- Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Skincare Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Ishikawa Prefectural Nursing University, Ishikawa, Japan
| | - Hiromi Sanada
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Ishikawa Prefectural Nursing University, Ishikawa, Japan
| | - Gojiro Nakagami
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
21
|
Chyła-Danił G, Sałaga-Zaleska K, Kreft E, Stumski O, Krzesińska A, Sakowicz-Burkiewicz M, Kuchta A, Jankowski M. Long-Term Effects of Suramin on Renal Function in Streptozotocin-Induced Diabetes in Rats. Int J Mol Sci 2023; 24:14671. [PMID: 37834118 PMCID: PMC10572378 DOI: 10.3390/ijms241914671] [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: 08/30/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
In short-term diabetes (3 weeks), suramin, a drug used clinically, affects renal function and the expression of vascular endothelial growth factor A (VEGF-A), which may be involved in the pathogenesis of diabetic nephropathy, the main cause of end-stage renal disease. In the present study, we evaluated the long-term (11 weeks) effects of suramin (10 mg/kg, i.p., once-weekly) in diabetic rats. Concentrations of VEGF-A, albumin, soluble adhesive molecules (sICAM-1, sVCAM-1), nucleosomes, and thrombin-antithrombin complex (TAT) were measured by ELISA, total protein was measured using a biuret reagent. Glomerular expression of VEGF-A was evaluated by Western blot, mRNA for VEGF-A receptors in the renal cortex by RT-PCR. The vasoreactivity of the interlobar arteries to acetylcholine was assessed by wire myography. Long-term diabetes led to an increased concentration of VEGF-A, TAT, and urinary excretion of total protein and albumin, and a decrease in the concentration of sVCAM-1. We have shown that suramin in diabetes reduces total urinary protein excretion and restores the relaxing properties of acetylcholine relaxation properties to non-diabetic levels. Suramin had no effect on glomerular expression VEGF-A expression and specific receptors, and on sICAM-1 and nucleosomes concentrations in diabetic rats. In conclusion, the long-term effect of suramin on the kidneys in diabetes, expressed in the reduction of proteinuria and the restoration of endothelium-dependent relaxation of the renal arteries, can be considered as potentially contributing to the reduction/slowing down of the development of diabetic nephropathy.
Collapse
Affiliation(s)
- Gabriela Chyła-Danił
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland; (G.C.-D.)
| | - Kornelia Sałaga-Zaleska
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland; (G.C.-D.)
| | - Ewelina Kreft
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland; (G.C.-D.)
| | - Olaf Stumski
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland; (G.C.-D.)
| | - Aleksandra Krzesińska
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland; (G.C.-D.)
| | | | - Agnieszka Kuchta
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland; (G.C.-D.)
| | - Maciej Jankowski
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland; (G.C.-D.)
| |
Collapse
|
22
|
Farrugia BL, Melrose J. The Glycosaminoglycan Side Chains and Modular Core Proteins of Heparan Sulphate Proteoglycans and the Varied Ways They Provide Tissue Protection by Regulating Physiological Processes and Cellular Behaviour. Int J Mol Sci 2023; 24:14101. [PMID: 37762403 PMCID: PMC10531531 DOI: 10.3390/ijms241814101] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
This review examines the roles of HS-proteoglycans (HS-PGs) in general, and, in particular, perlecan and syndecan as representative examples and their interactive ligands, which regulate physiological processes and cellular behavior in health and disease. HS-PGs are essential for the functional properties of tissues both in development and in the extracellular matrix (ECM) remodeling that occurs in response to trauma or disease. HS-PGs interact with a biodiverse range of chemokines, chemokine receptors, protease inhibitors, and growth factors in immune regulation, inflammation, ECM stabilization, and tissue protection. Some cell regulatory proteoglycan receptors are dually modified hybrid HS/CS proteoglycans (betaglycan, CD47). Neurexins provide synaptic stabilization, plasticity, and specificity of interaction, promoting neurotransduction, neurogenesis, and differentiation. Ternary complexes of glypican-1 and Robbo-Slit neuroregulatory proteins direct axonogenesis and neural network formation. Specific neurexin-neuroligin complexes stabilize synaptic interactions and neural activity. Disruption in these interactions leads to neurological deficits in disorders of functional cognitive decline. Interactions with HS-PGs also promote or inhibit tumor development. Thus, HS-PGs have complex and diverse regulatory roles in the physiological processes that regulate cellular behavior and the functional properties of normal and pathological tissues. Specialized HS-PGs, such as the neurexins, pikachurin, and Eyes-shut, provide synaptic stabilization and specificity of neural transduction and also stabilize the axenome primary cilium of phototoreceptors and ribbon synapse interactions with bipolar neurons of retinal neural networks, which are essential in ocular vision. Pikachurin and Eyes-Shut interactions with an α-dystroglycan stabilize the photoreceptor synapse. Novel regulatory roles for HS-PGs controlling cell behavior and tissue function are expected to continue to be uncovered in this fascinating class of proteoglycan.
Collapse
Affiliation(s)
- Brooke L. Farrugia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Raymond Purves Laboratory of Bone and Joint Research, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Sydney Medical School (Northern), University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
23
|
Liu Z, Rütten S, Buhl EM, Zhang M, Liu J, Rojas-González DM, Mela P. Development of a Silk Fibroin-Small Intestinal Submucosa Small-Diameter Vascular Graft with Sequential VEGF and TGF-β1 Inhibitor Delivery for In Situ Tissue Engineering. Macromol Biosci 2023; 23:e2300184. [PMID: 37262314 DOI: 10.1002/mabi.202300184] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/23/2023] [Indexed: 06/03/2023]
Abstract
Proper endothelialization and limited collagen deposition on the luminal surface after graft implantation plays a crucial role to prevent the occurrence of stenosis. To achieve these conditions, a biodegradable graft with adequate mechanical properties and the ability to sequentially deliver therapeutic agents isfabricated. In this study, a dual-release system is constructed through coaxial electrospinning by incorporating recombinant human vascular endothelial growth factor (VEGF) and transforming growth factor β1 (TGF-β1) inhibitor into silk fibroin (SF) nanofibers to form a bioactive membrane. The functionalized SF membrane as the inner layer of the graft is characterized by the release profile, cell proliferation and protein expression. It presents excellent biocompatibility and biodegradation, facilitating cell attachment, proliferation, and infiltration. The core-shell structure enables rapid VEGF release within 10 days and sustained plasmid delivery for 21 days. A 2.0-mm-diameter vascular graft is fabricated by integrating the SF membrane with decellularized porcine small intestinal submucosa (SIS), aiming to facilitate the integration process under a stable extracellular matrix structure. The bioengineered graft is functionalized with the sequential administration of VEGF and TGF-β1, and with the reinforced and compatible mechanical properties, thereby offers an orchestrated solution for stenosis with potential for in situ vascular tissue engineering applications.
Collapse
Affiliation(s)
- Zhengni Liu
- Department of Biohybrid & Medical Textiles (BioTex) at AME-Institute of Applied Medical Engineering, Helmholtz Institute-CBMS, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
- Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, Jimo road 150, Shanghai, 200120, PR China
| | - Stephan Rütten
- Electron Microscopy Facility, Uniklinik RWTH Aachen, Pauwelsstrasse, 30, 52074, Aachen, Germany
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Uniklinik RWTH Aachen, Pauwelsstrasse, 30, 52074, Aachen, Germany
| | - Minjun Zhang
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju road 639, Shanghai, 200011, PR China
| | - Jiajie Liu
- Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, Jimo road 150, Shanghai, 200120, PR China
| | - Diana M Rojas-González
- Department of Biohybrid & Medical Textiles (BioTex) at AME-Institute of Applied Medical Engineering, Helmholtz Institute-CBMS, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Petra Mela
- Department of Biohybrid & Medical Textiles (BioTex) at AME-Institute of Applied Medical Engineering, Helmholtz Institute-CBMS, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| |
Collapse
|
24
|
Turner CT, Zeglinski MR, Boivin W, Zhao H, Pawluk MA, Richardson KC, Chandrabalan A, Bird P, Ramachandran R, Sehmi R, Lima H, Gauvreau G, Granville DJ. Granzyme K contributes to endothelial microvascular damage and leakage during skin inflammation. Br J Dermatol 2023; 189:279-291. [PMID: 36652225 DOI: 10.1093/bjd/ljac017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 06/06/2022] [Accepted: 09/13/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Granzyme K (GzmK) is a serine protease with minimal presence in healthy tissues while abundant in inflamed tissues. Initially thought to play an exclusive role in immune-mediated cell death, extracellular GzmK can also promote inflammation. OBJECTIVES To evaluate the role of GzmK in the pathogenesis of atopic dermatitis (AD), the most common inflammatory skin disease. METHODS A panel of human AD and control samples was analysed to determine if GzmK is elevated. Next, to determine a pathological role for GzmK in AD-like skin inflammation, oxazolone-induced dermatitis was induced in GzmK-/- and wild-type (WT) mice. RESULTS In human lesional AD samples, there was an increase in the number of GzmK+ cells compared with healthy controls. GzmK-/- mice exhibited reduced overall disease severity characterized by reductions in scaling, erosions and erythema. Surprisingly, the presence of GzmK did not notably increase the overall pro-inflammatory response or epidermal barrier permeability in WT mice; rather, GzmK impaired angiogenesis, increased microvascular damage and microhaemorrhage. Mechanistically, GzmK contributed to vessel damage through cleavage of syndecan-1, a key structural component of the glycocalyx, which coats the luminal surface of vascular endothelia. CONCLUSIONS GzmK may provide a potential therapeutic target for skin conditions associated with persistent inflammation, vasculitis and pathological angiogenesis.
Collapse
Affiliation(s)
- Christopher T Turner
- International Collaboration On Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia, Vancouver, BC, Canada
| | - Matthew R Zeglinski
- International Collaboration On Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia, Vancouver, BC, Canada
| | - Wendy Boivin
- Department of Pathology and Laboratory Medicine; University of British Columbia, Vancouver, BC, Canada
| | - Hongyan Zhao
- International Collaboration On Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia, Vancouver, BC, Canada
| | - Megan A Pawluk
- International Collaboration On Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia, Vancouver, BC, Canada
| | - Katlyn C Richardson
- International Collaboration On Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia, Vancouver, BC, Canada
| | - Arundhasa Chandrabalan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Phillip Bird
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Roma Sehmi
- Division of Respirology, Department of Medicine, McMaster University, Ontario, L8S 4K1, Canada
| | - Hermenio Lima
- Division of Dermatology, Department of Medicine, McMaster University, Ontario, L8S 4K1, Canada
| | - Gail Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Ontario, L8S 4K1, Canada
| | - David J Granville
- International Collaboration On Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia, Vancouver, BC, Canada
- British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, BC, Canada
| |
Collapse
|
25
|
Rauff A, Manning JC, Hoying JB, LaBelle SA, Strobel HA, Stoddard GJ, Weiss JA. Dynamic Biophysical Cues Near the Tip Cell Microenvironment Provide Distinct Guidance Signals to Angiogenic Neovessels. Ann Biomed Eng 2023; 51:1835-1846. [PMID: 37149511 DOI: 10.1007/s10439-023-03202-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/01/2023] [Indexed: 05/08/2023]
Abstract
The formation of new vascular networks via angiogenesis is a crucial biological mechanism to balance tissue metabolic needs, yet the coordination of factors that influence the guidance of growing neovessels remain unclear. This study investigated the influence of extracellular cues within the immediate environment of sprouting tips over multiple hours and obtained quantitative relationships describing their effects on the growth trajectories of angiogenic neovessels. Three distinct microenvironmental cues-fibril tracks, ECM density, and the presence of nearby cell bodies-were extracted from 3D time series image data. The prominence of each cue was quantified along potential sprout trajectories to predict the response to multiple microenvironmental factors simultaneously. Sprout trajectories significantly correlated with the identified microenvironmental cues. Specifically, ECM density and nearby cellular bodies were the strongest predictors of the trajectories taken by neovessels (p < 0.001 and p = 0.016). Notwithstanding, direction changing trajectories, deviating from the initial neovessel orientation, were significantly correlated with fibril tracks (p = 0.003). Direction changes also occurred more frequently with strong microenvironmental cues. This provides evidence for the first time that local matrix fibril alignment influences changes in sprout trajectories but does not materially contribute to persistent sprouting. Together, our results suggest the microenvironmental cues significantly contribute to guidance of sprouting trajectories. Further, the presented methods quantitatively distinguish the influence of individual microenvironmental stimuli during guidance.
Collapse
Affiliation(s)
- Adam Rauff
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive, Rm. 3100, Salt Lake City, UT, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Jason C Manning
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive, Rm. 3100, Salt Lake City, UT, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Steven A LaBelle
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive, Rm. 3100, Salt Lake City, UT, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Gregory J Stoddard
- Study Design and Biostatistics Center, University of Utah, Salt Lake City, UT, USA
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive, Rm. 3100, Salt Lake City, UT, USA.
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
26
|
Wang X, Sun Q, Wang W, Liu B, Gu Y, Chen L. Decoding key cell sub-populations and molecular alterations in glioblastoma at recurrence by single-cell analysis. Acta Neuropathol Commun 2023; 11:125. [PMID: 37525259 PMCID: PMC10391841 DOI: 10.1186/s40478-023-01613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/01/2023] [Indexed: 08/02/2023] Open
Abstract
Glioblastoma (GBM) is the most frequent malignant brain tumor, the relapse of which is unavoidable following standard treatment. However, the effective treatment for recurrent GBM is lacking, necessitating the understanding of key mechanisms driving tumor recurrence and the identification of new targets for intervention. Here, we integrated single-cell RNA-sequencing data spanning 36 patient-matched primary and recurrent GBM (pGBM and rGBM) specimens, with 6 longitudinal GBM spatial transcriptomics to explore molecular alterations at recurrence, with each cell type characterized in parallel. Genes involved in extracellular matrix (ECM) organization are preferentially enriched in rGBM cells, and MAFK is highlighted as a potential regulator. Notably, we uncover a unique subpopulation of GBM cells that is much less detected in pGBM and highly expresses ECM and mesenchyme related genes, suggesting it may contribute to the molecular transition of rGBM. Further regulatory network analysis reveals that transcription factors, such as NFATC4 and activator protein 1 members, may function as hub regulators. All non-tumor cells alter their specific sets of genes as well and certain subgroups of myeloid cells appear to be physically associated with the mesenchyme-like GBM subpopulation. Altogether, our study provides new insights into the molecular understanding of GBM relapse and candidate targets for rGBM treatment.
Collapse
Affiliation(s)
- Xin Wang
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
- BGI Research, Hangzhou, 310030 China
| | - Qian Sun
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| | - Weiwen Wang
- China National GeneBank, BGI Research, Shenzhen, 518120 China
| | - Baohui Liu
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| | - Ying Gu
- BGI Research, Hangzhou, 310030 China
- BGI Research, Shenzhen, 518083 China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083 China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Liang Chen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
27
|
Chyła-Danił G, Sałaga-Zaleska K, Kreft E, Krzesińska A, Herman S, Kuchta A, Sakowicz-Burkiewicz M, Lenartowicz M, Jankowski M. Suramin Affects the Renal VEGF-A/VEGFR Axis in Short-Term Streptozotocin-Induced Diabetes. Pharmaceuticals (Basel) 2023; 16:ph16030470. [PMID: 36986570 PMCID: PMC10053825 DOI: 10.3390/ph16030470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Diabetic nephropathy (DN) accounts for approximately 50% of end-stage renal diseases. Vascular endothelial growth factor A (VEGF-A) is thought to be a critical mediator of vascular dysfunction in DN, but its role is unclear. The lack of pharmacological tools to modify renal concentrations further hinders the understanding of its role in DN. In this study, rats were evaluated after 3 weeks of streptozotocin-induced diabetes and two suramin treatments (10 mg/kg, ip). Vascular endothelial growth factor A expression was evaluated by western blot of glomeruli and immunofluorescence of the renal cortex. RT-PCR for receptors Vegfr1 mRNA and Vegfr2 mRNA quantitation was performed. The soluble adhesive molecules (sICAM-1, sVCAM-1) in blood were measured by ELISA and the vasoreactivity of interlobar arteries to acetylcholine was evaluated using wire myography. Suramin administration reduced the expression and intraglomerular localisation of VEGF-A. Increased VEGFR-2 expression in diabetes was reduced by suramin to non-diabetic levels. Diabetes reduced the sVCAM-1 concentrations. Suramin in diabetes restored acetylcholine relaxation properties to non-diabetic levels. In conclusion, suramin affects the renal VEGF-A/VEGF receptors axis and has a beneficial impact on endothelium-dependent relaxation of renal arteries. Thus, suramin may be used as a pharmacological agent to investigate the potential role of VEGF-A in the pathogenesis of renal vascular complications in short-term diabetes.
Collapse
Affiliation(s)
- Gabriela Chyła-Danił
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland
| | - Kornelia Sałaga-Zaleska
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland
| | - Ewelina Kreft
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland
| | - Aleksandra Krzesińska
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland
| | - Sylwia Herman
- Laboratory of Genetics and Evolutionism, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland
| | - Agnieszka Kuchta
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland
| | | | - Małgorzata Lenartowicz
- Laboratory of Genetics and Evolutionism, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland
| | - Maciej Jankowski
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-210 Gdańsk, Poland
| |
Collapse
|
28
|
Cadamuro F, Nicotra F, Russo L. 3D printed tissue models: From hydrogels to biomedical applications. J Control Release 2023; 354:726-745. [PMID: 36682728 DOI: 10.1016/j.jconrel.2023.01.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
The development of new advanced constructs resembling structural and functional properties of human organs and tissues requires a deep knowledge of the morphological and biochemical properties of the extracellular matrices (ECM), and the capacity to reproduce them. Manufacturing technologies like 3D printing and bioprinting represent valuable tools for this purpose. This review will describe how morphological and biochemical properties of ECM change in different tissues, organs, healthy and pathological states, and how ECM mimics with the required properties can be generated by 3D printing and bioprinting. The review describes and classifies the polymeric materials of natural and synthetic origin exploited to generate the hydrogels acting as "inks" in the 3D printing process, with particular emphasis on their functionalization allowing crosslinking and conjugation with signaling molecules to develop bio-responsive and bio-instructive ECM mimics.
Collapse
Affiliation(s)
- Francesca Cadamuro
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy
| | - Francesco Nicotra
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy
| | - Laura Russo
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy; CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY Galway, Ireland.
| |
Collapse
|
29
|
Elomaa L, Lindner M, Leben R, Niesner R, Weinhart M. In vitro vascularization of hydrogel-based tissue constructs via a combined approach of cell sheet engineering and dynamic perfusion cell culture. Biofabrication 2023; 15. [DOI: 10.1088/1758-5090/ac9433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/22/2022] [Indexed: 11/11/2022]
Abstract
Abstract
The bioengineering of artificial tissue constructs requires special attention to their fast vascularization to provide cells with sufficient nutrients and oxygen. We addressed the challenge of in vitro vascularization by employing a combined approach of cell sheet engineering, 3D printing, and cellular self-organization in dynamic maturation culture. A confluent cell sheet of human umbilical vein endothelial cells (HUVECs) was detached from a thermoresponsive cell culture substrate and transferred onto a 3D-printed, perfusable tubular scaffold using a custom-made cell sheet rolling device. Under indirect co-culture conditions with human dermal fibroblasts (HDFs), the cell sheet-covered vessel mimic embedded in a collagen gel together with additional singularized HUVECs started sprouting into the surrounding gel, while the suspended cells around the tube self-organized and formed a dense lumen-containing 3D vascular network throughout the gel. The HDFs cultured below the HUVEC-containing cell culture insert provided angiogenic support to the HUVECs via molecular crosstalk without competing for space with the HUVECs or inducing rapid collagen matrix remodeling. The resulting vascular network remained viable under these conditions throughout the 3 week cell culture period. This static indirect co-culture setup was further transferred to dynamic flow conditions, where the medium perfusion was enabled via two independently addressable perfusion circuits equipped with two different cell culture chambers, one hosting the HDFs and the other hosting the HUVEC-laden collagen gel. Using this system, we successfully connected the collagen-embedded HUVEC culture to a dynamic medium flow, and within 1 week of the dynamic cell culture, we detected angiogenic sprouting and dense microvascular network formation via HUVEC self-organization in the hydrogel. Our approach of combining a 3D-printed and cell sheet-covered vascular precursor that retained its sprouting capacity together with the self-assembling HUVECs in a dynamic perfusion culture resulted in a vascular-like 3D network, which is a critical step toward the long-term vascularization of bioengineered in vitro tissue constructs.
Collapse
|
30
|
Fontemaggi G. Non-coding RNA regulatory networks in post-transcriptional regulation of VEGFA in cancer. IUBMB Life 2023; 75:30-39. [PMID: 35467790 PMCID: PMC10084289 DOI: 10.1002/iub.2620] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/10/2022] [Indexed: 12/29/2022]
Abstract
The switch from the normal quiescent vasculature to angiogenesis in tumors is induced by a variety of growth factors, released from cancer and stromal cells upon oxygen and nutrients deprivation. Vascular endothelial growth factor A (VEGF-A) is a potent-secreted mitogen and the only growth factor specific to endothelial cells that is observed almost ubiquitously at sites of angiogenesis. Expression of VEGF-A in cancer cells is controlled through transcriptional and post-transcriptional mechanisms. Post-transcriptional regulation of VEGF-A occurs at multiple levels, through the control of splicing, mRNA stability and translation rate, enabling a fine-tuned expression and release of VEGF-A. Mounting evidence is highlighting the important role played by microRNAs (miRNAs) in the control of VEGF-A mRNA stability and translation in cancer. Moreover, non-coding RNAs, as long non-coding RNAs and circular RNAs, are emerging as crucial modulators of VEGF-A-targeting miRNAs, with consequent ability to modulate VEGF-A expression. This review discusses the recent progress on the ncRNA-related networks controlling VEGF-A expression in cancer cells and provides insights into the complexity of VEGF-A post-transcriptional regulation.
Collapse
Affiliation(s)
- Giulia Fontemaggi
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
31
|
Zhang C, Li T, Yin S, Gao M, He H, Li Y, Jiang D, Shi M, Wang J, Yu L. Monocytes deposit migrasomes to promote embryonic angiogenesis. Nat Cell Biol 2022; 24:1726-1738. [PMID: 36443426 DOI: 10.1038/s41556-022-01026-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/12/2022] [Indexed: 11/30/2022]
Abstract
Pro-angiogenic factors are key regulators of angiogenesis. Here we report that highly migratory cells patrol the area of capillary formation in chick embryo chorioallantoic membrane. These cells deposit migrasomes on their migration tracks, creating migrasome-enriched areas. Single-cell sequencing identified these cells as monocytes. Depletion of monocytes impairs capillary formation. Quantitative mass spectrometry analysis reveals that monocyte migrasomes are enriched with pro-angiogenic factors. Purified migrasomes promote capillary formation and monocyte recruitment in vivo, and endothelial cell tube formation and monocyte chemotaxis in vitro. Knockdown or knockout of TSPAN4 reduces migrasome formation and impairs capillary formation and monocyte recruitment. Mechanistically, monocytes promote angiogenesis via VEGFA and CXCL12 in migrasomes. In summary, monocytes deposit migrasomes enriched in pro-angiogenic factors to promote angiogenesis.
Collapse
Affiliation(s)
- Cuifang Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Tianqi Li
- School of Life Science, Tsinghua University, Beijing, China
| | - Shuyao Yin
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Mingyi Gao
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Helen He
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Li
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Dong Jiang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Minghui Shi
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jianbin Wang
- School of Life Science, Tsinghua University, Beijing, China
| | - Li Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
32
|
Zafer D, Adams T, Olson E, Stenman L, Taparli O, Eickhoff J, Cengiz P, Mezu-Ndubuisi OJ. Retinal vascular recovery revealed by retinal imaging following neonatal hypoxia ischemia in mice: Is there a role for tyrosine kinase receptor modulation? Brain Res 2022; 1796:148093. [PMID: 36116486 PMCID: PMC10013450 DOI: 10.1016/j.brainres.2022.148093] [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: 03/12/2022] [Revised: 09/06/2022] [Accepted: 09/11/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Hypoxic ischemic encephalopathy (HIE) secondary to perinatal asphyxia leads to long-term visual disabilities. Dilated retinal exams in human newborns with HIE is an emerging diagnostic tool, but phenotypes of hypoxia ischemia (HI) related retinal vascular injury are unclear. 7,8-Dihydroxyflavone (7,8-DHF) is a TrkB agonist with protective effects on HI-related brain damage. We studied retinal vessels in a mouse model of neonatal HIE and the efficacy of 7,8-DHF in ameliorating HI-related retinal vascular injury. METHODS C57BL6/J mice at post-natal day (P) 9 received unilateral left carotid artery ligation followed by exposure to 10 % oxygen for 50 min. Phosphate buffered saline or 7,8-DHF (5 mg/kg) were administered daily for 7 days intraperitoneally. Control groups of naïve or carotid artery ligation only mice were studied. Fluorescein angiography was performed in acute (two weeks post-exposure) and chronic (four weeks post-exposure) time points. Retinal artery width, retinal vein width, and collateral vessel length were quantified. RESULTS Ligation of the common carotid artery alone caused retinal artery dilation in acute and chronic time points, but had no effect on retinal veins. At acute time point, HI caused increased retinal artery vasodilation, but was reversed by 7,8-DHF. HI caused short collateral vessel formation in ipsilateral eyes, rescued by 7,8-DHF treatment. CONCLUSION Retinal artery vasodilation and collateral vessel formation due to HI were rescued by 7,8-DHF treatment. Retinal and collateral vessel monitoring could be diagnostic biomarkers for HI severity. Studies to elucidate mechanisms of 7,8-DHF action on retinal vessels could aid development of therapies for neonatal HI.
Collapse
Affiliation(s)
- Dila Zafer
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA; Waisman Center, University of Wisconsin, Madison, WI, USA
| | - Thao Adams
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Ellie Olson
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Lauren Stenman
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA.
| | - Onur Taparli
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA; Waisman Center, University of Wisconsin, Madison, WI, USA.
| | - Jens Eickhoff
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA.
| | - Pelin Cengiz
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA; Waisman Center, University of Wisconsin, Madison, WI, USA.
| | - Olachi J Mezu-Ndubuisi
- Department of Pediatrics, University of Rochester, Rochester, NY, USA; Department of Ophthalmology, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
33
|
Anticarcinogenic Effects of Isothiocyanates on Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms232213834. [PMID: 36430307 PMCID: PMC9693344 DOI: 10.3390/ijms232213834] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, accounting for about 90% of cases. Sorafenib, lenvatinib, and the combination of atezolizumab and bevacizumab are considered first-line treatments for advanced HCC. However, clinical application of these drugs has also caused some adverse reactions such as hypertension, elevated aspartate aminotransferases, and proteinuria. At present, natural products and their derivatives have drawn more and more attention due to less side effects as cancer treatments. Isothiocyanates (ITCs) are one type of hydrolysis products from glucosinolates (GLSs), secondary plant metabolites found exclusively in cruciferous vegetables. Accumulating evidence from encouraging in vitro and in vivo animal models has demonstrated that ITCs have multiple biological activities, especially their potentially health-promoting activities (antibacterial, antioxidant, and anticarcinogenic effects). In this review, we aim to comprehensively summarize the chemopreventive, anticancer, and chemosensitizative effects of ITCs on HCC, and explain the underlying molecular mechanisms.
Collapse
|
34
|
Qin Y, Yang J, Liang C, Liu J, Deng Z, Yan B, Fu Y, Luo Y, Li X, Wei X, Li W. Pan-cancer analysis identifies migrasome-related genes as a potential immunotherapeutic target: A bulk omics research and single cell sequencing validation. Front Immunol 2022; 13:994828. [PMID: 36405728 PMCID: PMC9669594 DOI: 10.3389/fimmu.2022.994828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Introduction The migrasome is a newly discovered organelle that resembles extracellular vesicles in structure. However, the function of the migrasome in tumors, particularly in relation to tumor immunity and tumor microenvironment, is unclear. Methods Gene expression data, copy number variation raw data, and methylation data of 33 cancer types were downloaded from The Cancer Genome Atlas database. Immunohistochemistry (IHC) based on 114 case of colorectal cancer was used to validate the expression of the migrasome hub-gene. We analyzed the expression, prognosis, genetic variation, and drug sensitivity profiles of migrasome-related genes (MRGs) in pan-cancer datasets. A migrasome score was constructed based on gene set enrichment analysis, and the correlation of migrasomes with the tumor microenvironment was assessed. The CancerSEA was used to perform a single-cell level functional analysis of the migrasome. Additionally, we also analyzed the correlation between migrasomes and tumor mutational burden (TMB), microsatellite instability (MSI), and tumor immune dysfunction and exclusion scores. Single-cell transcriptome sequencing (scRNA-seq) data was used to assess the activation state of migrasomes in the tumor microenvironment. Results PIGK expression was significantly up-regulated in 22 of 33 tumors, and high expression of migrasome was estimated to have contributed to poor prognosis. Missense mutations are the most common type of mutation in MRGs. We identified piperlongumine as a potential drug targeting migrasomes. The migrasome score was significantly and positively correlated with the tumor immunity score and the stroma score. In most tumors, the abundance of macrophages in the tumor microenvironment was significantly and positively correlated with the migrasome score. Additionally, the migrasome scores were significantly correlated with the immune checkpoint genes in pan-cancer as well as immune checkpoint therapy-related markers including TMB and MSI. According to scRNA-seq analysis, migrasome differed significantly among cells of the tumor microenvironment. IHC confirmed low expression of ITGA5 and PIGK in colorectal cancer. Discussion We performed the first pan-cancer analysis of migrasomes and discovered that they play an important role in tumor development and immune escape. Our study provides new insights into the role of migrasomes in tumor prognosis and immunotherapy.
Collapse
Affiliation(s)
- Yan Qin
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region and Research center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
| | - Jie Yang
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region and Research center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
| | - Cao Liang
- Department of Surgical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Jun Liu
- Department of Surgical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Zhixing Deng
- Department of Surgical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Binli Yan
- Department of Surgical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Ying Fu
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region and Research center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
| | - Yinghua Luo
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region and Research center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
| | - Xiaozhen Li
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region and Research center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
- *Correspondence: Wei Li, ; Xiaoying Wei, ; Xiaozhen Li,
| | - Xiaoying Wei
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region and Research center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
- *Correspondence: Wei Li, ; Xiaoying Wei, ; Xiaozhen Li,
| | - Wei Li
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region and Research center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
- *Correspondence: Wei Li, ; Xiaoying Wei, ; Xiaozhen Li,
| |
Collapse
|
35
|
Berger AA, Kawaler EA, Dao F, Misirlioglu S, Fernandez EA, Olvera N, Van Oudenhove E, DeLair D, Levine DA. The role of CTNNB1 mutations and matrix metalloproteinases (MMPs) in anti-angiogenesis treatment of endometrial carcinoma. Gynecol Oncol 2022; 167:323-333. [PMID: 36150916 DOI: 10.1016/j.ygyno.2022.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/08/2022] [Accepted: 09/10/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Treatment options and associated biomarkers for advanced and recurrent disease are limited. Endometrial cancers (ECs) with CTNNB1 exon 3 mutations appear to have preferential response to bevacizumab, an anti-angiogenesis treatment, though the mechanism of action is unknown. We aim to identify mediators of bevacizumab-responsive endometrial cancers. METHODS We analyzed RNA expression from TCGA and protein expression from CPTAC to identify likely targets for β-catenin overactivity. We then transiently and stably overexpressed β-catenin in EC cells to confirm the results suggested by our in silico analysis. We performed corroborative experiments by silencing CTNNB1 in mutated cell lines to demonstrate functional specificity. We implanted transduced cells into xenograft models to study microvessel density. RESULTS CTNNB1-mutated ECs were associated with increased β-catenin and MMP7 protein abundance (P < 0.001), but not VEGF-A protein abundance. Overexpressing β-catenin in EC cells did not increase VEGF-A abundance but did increase expression and secretion of MMP7 (P < 0.03). Silencing CTNNB1 in CTNNB1-mutated cells decreased MMP7 gene expression in EC (P < 0.0001). Microvessel density was not increased. CONCLUSIONS These data provide a mechanistic understanding for bevacizumab-response in CTNNB1-mutated ECs demonstrated in GOG-86P. We hypothesize that overexpressed and secreted MMP7 potentially digests VEGFR-1, releasing VEGF-A, and increasing its availability. These activities may drive the formation of permeable vessels, which contributes to tumor progression, metastasis, and immune suppression. This mechanism is unique to EC and advocates for further clinical trials evaluating this treatment-related biomarker.
Collapse
Affiliation(s)
- Amnon A Berger
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States of America; Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States of America
| | - Emily A Kawaler
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Fanny Dao
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States of America
| | - Selim Misirlioglu
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States of America
| | | | - Narciso Olvera
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States of America
| | - Elke Van Oudenhove
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States of America
| | - Deborah DeLair
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States of America
| | - Douglas A Levine
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States of America; Merck Research Laboratories, Rahway, NJ, United States of America.
| |
Collapse
|
36
|
Richardson KC, Jung K, Pardo J, Turner CT, Granville DJ. Noncytotoxic Roles of Granzymes in Health and Disease. Physiology (Bethesda) 2022; 37:323-348. [PMID: 35820180 DOI: 10.1152/physiol.00011.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Granzymes are serine proteases previously believed to play exclusive and somewhat redundant roles in lymphocyte-mediated target cell death. However, recent studies have challenged this paradigm. Distinct substrate profiles and functions have since emerged for each granzyme while their dysregulated proteolytic activities have been linked to diverse pathologies.
Collapse
Affiliation(s)
- Katlyn C Richardson
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen Jung
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julian Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), Zaragoza, Spain.,Department of Microbiology, Radiology, Pediatrics and Public Health, University of Zaragoza, Zaragoza, Spain.,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Zaragoza, Spain
| | - Christopher T Turner
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia.,Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
37
|
Savic G, Stevanovic I, Mihajlovic D, Jurisevic M, Gajovic N, Jovanovic I, Ninkovic M. MMP-9/BDNF ratio predicts more severe COVID-19 outcomes. Int J Med Sci 2022; 19:1903-1911. [PMID: 36438922 PMCID: PMC9682503 DOI: 10.7150/ijms.75337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/16/2022] [Indexed: 01/24/2023] Open
Abstract
COVID-19 clinically manifests from asymptomatic to the critical range. Immune response provokes the pro-inflammatory interactions, which lead to the cytokines, reactive oxygen/nitrogen species, peptidases, and arachidonic acid metabolites enlargement and activation of coagulation components. Matrix metalloproteinases (MMPs) contribute to tissue destruction in the development of COVID-19. Due to the endothelial, systemic course of the disease, VEGF A participates actively in COVID-19 development, while neurotrophic and metabolic effects of BDNF recommends for the prediction of complications in COVID-19 patients. Searching for a marker that would improve and simplify the ranking in COVID-19, the study intended to evaluate the relationship of MMP-9 with VEGF A, BDNF, and MMP-8 with the COVID-19 severity. Upon admission to the hospital and before the therapy administration, 77 patients were classified into a mild, moderate, severe, or critical group. Due to the inflammatory stage in COVID-19, a comparison between groups showed related differences in leukocytes, neutrophils, lymphocytes, and platelets counts as anticipated. Only in seriously ill patients, there is a significant increase in the serum concentration of MMP-9, MMP-8, and VEGF A, while BDNF values did not show significant variations between groups. However, all those parameters positively correlated with each other. The ratio of MMP-9/BDNF markedly decreased in the severe and critically patients compared to the mild group. Testing the capability of this ratio to predict the COVID-19 stage by ROC curves, we found the MMP-9/BDNF could be a suitable marker for differentiating stages I/II (AUC 0.7597), stage I/III (AUC 0.9011), and stage I/IV (AUC 0.7727). Presented data describe for the first time the high-level systemic MMP-9/BDNF ratio in patients with COVID-19. This parameter could contribute to a more precise determination of the phase of the disease.
Collapse
Affiliation(s)
- Goran Savic
- Medical Faculty of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia
| | - Ivana Stevanovic
- Medical Faculty of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia
- Institute of Medical Research, Military Medical Academy, Crnotravska 17, Belgrade, Serbia
| | - Dusan Mihajlovic
- Medical Faculty of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia
| | - Milena Jurisevic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Nevena Gajovic
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Ivan Jovanovic
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Milica Ninkovic
- Medical Faculty of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia
- Institute of Medical Research, Military Medical Academy, Crnotravska 17, Belgrade, Serbia
| |
Collapse
|
38
|
Menger MM, Laschke MW, Nussler AK, Menger MD, Histing T. The vascularization paradox of non-union formation. Angiogenesis 2022; 25:279-290. [PMID: 35165821 PMCID: PMC9249698 DOI: 10.1007/s10456-022-09832-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/21/2021] [Indexed: 01/01/2023]
Abstract
Despite major research efforts to elucidate mechanisms of non-union formation, failed fracture healing remains a common complication in orthopedic surgery. Adequate vascularization has been recognized as a crucial factor for successful bone regeneration, as newly formed microvessels guarantee the supply of the callus tissue with vital oxygen, nutrients, and growth factors. Accordingly, a vast number of preclinical studies have focused on the development of vascularization strategies to stimulate fracture repair. However, recent evidence suggests that stimulation of blood vessel formation is an oversimplified approach to support bone regeneration. This review discusses the role of vascularization during bone regeneration and delineates a phenomenon, for which we coin the term "the vascularization paradox of non-union-formation". This view is based on the results of a variety of experimental studies that suggest that the callus tissue of non-unions is indeed densely vascularized and that pro-angiogenic mediators, such as vascular endothelial growth factor, are sufficiently expressed at the facture site. By gaining further insights into the molecular and cellular basis of non-union vascularization, it may be possible to develop more optimized treatment approaches or even prevent the non-union formation in the future.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany.
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany.
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Andreas K Nussler
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| |
Collapse
|
39
|
Characterization of the Expression of Angiogenic Factors in Cutaneous Squamous Cell Carcinoma of Domestic Cats. Vet Sci 2022; 9:vetsci9070375. [PMID: 35878392 PMCID: PMC9351683 DOI: 10.3390/vetsci9070375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is a common malignant skin cancer with a significant impact on health, and it is important to determine the degree of reliance of CSCC on angiogenesis for growth and metastasis. Major regulators of angiogenesis are the vascular endothelial growth factor (VEGF) family and their associated receptors. Alternative pre-mRNA splicing produces multiple isoforms of VEGF-A and PLGF with distinct biological properties. Several studies highlight the function of VEGF-A in CSCC, but there are no studies of the different isoforms of VEGF-A and PLGF for this neoplasm. We characterized the expression of three isoforms of VEGF-A, two isoforms of PLGF, and their receptors in cat CSCC biopsies compared to normal haired skin (NHS). Although our results revealed no significant changes in transcript levels of panVEGF-A or their isoforms, the mRNA levels of PLGF I and the receptors Flt-1 and KDR were downregulated in CSCC compared to NHS. Differences were observed in ligand:receptor mRNA expression ratio, with the expression of VEGF-A relative to its receptor KDR higher in CSCC, which is consistent with our hypothesis and prior human SCC studies. Immunolocalization in tissue showed increased expression of all measured factors and receptors in tumor cells compared to NHS and surrounding vasculature. We conclude that the factors measured may play a pivotal role in CSCC growth, although further studies are needed to clarify the role of angiogenic factors in feline CSCC.
Collapse
|
40
|
González-Zamora J, Hernandez M, Recalde S, Bezunartea J, Montoliu A, Bilbao-Malavé V, Orbe J, Rodríguez JA, Llorente-González S, Fernández-Robredo P, García-Layana A. Matrix Metalloproteinase 10 Contributes to Choroidal Neovascularisation. Biomedicines 2022; 10:biomedicines10071557. [PMID: 35884862 PMCID: PMC9313238 DOI: 10.3390/biomedicines10071557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Age-related macular degeneration (AMD) is currently the main cause of severe visual loss among older adults in developed countries. The pathophysiology has not been clarified, but oxidative stress is believed to play a major role. Matrix metalloproteinases (MMP) may play a prominent role in several steps of the pathophysiology of AMD, especially in its neovascular form; therefore, there is of great interest in understanding their role in choroidal neovascularisation. This study aimed to elucidate the role of MMP10 in the development of choroidal neovascularisation (CNV). We have demonstrated that MMP10 was expressed by retinal pigment epithelium cells and endothelial cells of the neovascular membrane, in cell culture, mouse and human retina. MMP10 expression and activity increased under oxidative stress conditions in ARPE-19 cells. MMP10-/- mice developed smaller laser-induced areas of CNV. Furthermore, to exclude a systemic MMP10 imbalance in these patients, plasma MMP10 concentrations were assessed in an age- and sex-matched sample of 52 control patients and 52 patients with neovascular AMD and no significant differences were found between the groups, demonstrating that MMP10 induction is a local phenomenon. Our findings suggest that MMP10 participates in the development of choroidal neovascularisation and promotes MMP10 as a possible new therapeutic target.
Collapse
Affiliation(s)
- Jorge González-Zamora
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
| | - María Hernandez
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
- Correspondence: (M.H.); (P.F.-R.)
| | - Sergio Recalde
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
| | - Jaione Bezunartea
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
| | - Ana Montoliu
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
| | - Valentina Bilbao-Malavé
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
| | - Josune Orbe
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA-Universidad de Navarra, CIBERCV, 31008 Pamplona, Spain
| | - José A. Rodríguez
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA-Universidad de Navarra, CIBERCV, 31008 Pamplona, Spain
| | - Sara Llorente-González
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
| | - Patricia Fernández-Robredo
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
- Correspondence: (M.H.); (P.F.-R.)
| | - Alfredo García-Layana
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (J.G.-Z.); (S.R.); (J.B.); (A.M.); (V.B.-M.); (S.L.-G.); (A.G.-L.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain; (J.O.); (J.A.R.)
| |
Collapse
|
41
|
Tan S, Chen Z, Mironchik Y, Mori N, Penet MF, Si G, Krishnamachary B, Bhujwalla ZM. VEGF Overexpression Significantly Increases Nanoparticle-Mediated siRNA Delivery and Target-Gene Downregulation. Pharmaceutics 2022; 14:pharmaceutics14061260. [PMID: 35745832 PMCID: PMC9229257 DOI: 10.3390/pharmaceutics14061260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 02/01/2023] Open
Abstract
The availability of nanoparticles (NPs) to deliver small interfering RNA (siRNA) has significantly expanded the specificity and range of ‘druggable’ targets for precision medicine in cancer. This is especially important for cancers such as triple negative breast cancer (TNBC) for which there are no targeted treatments. Our purpose here was to understand the role of tumor vasculature and vascular endothelial growth factor (VEGF) overexpression in a TNBC xenograft in improving the delivery and function of siRNA NPs using in vivo as well as ex vivo imaging. We used triple negative MDA-MB-231 human breast cancer xenografts derived from cells engineered to overexpress VEGF to understand the role of VEGF and vascularization in NP delivery and function. We used polyethylene glycol (PEG) conjugated polyethylenimine (PEI) NPs to deliver siRNA that downregulates choline kinase alpha (Chkα), an enzyme that is associated with malignant transformation and tumor progression. Because Chkα converts choline to phosphocholine, effective delivery of Chkα siRNA NPs resulted in functional changes of a significant decrease in phosphocholine and total choline that was detected with 1H magnetic resonance spectroscopy (MRS). We observed a significant increase in NP delivery and a significant decrease in Chkα and phosphocholine in VEGF overexpressing xenografts. Our results demonstrated the importance of tumor vascularization in achieving effective siRNA delivery and downregulation of the target gene Chkα and its function.
Collapse
Affiliation(s)
- Shanshan Tan
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Zhihang Chen
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Noriko Mori
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21205, USA
| | - Ge Si
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
| | - Zaver M. Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, MD 21205, USA; (S.T.); (Z.C.); (Y.M.); (N.M.); (M.-F.P.); (G.S.); (B.K.)
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21205, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Correspondence:
| |
Collapse
|
42
|
Yavvari P, Laporte A, Elomaa L, Schraufstetter F, Pacharzina I, Daberkow AD, Hoppensack A, Weinhart M. 3D-Cultured Vascular-Like Networks Enable Validation of Vascular Disruption Properties of Drugs In Vitro. Front Bioeng Biotechnol 2022; 10:888492. [PMID: 35769106 PMCID: PMC9234334 DOI: 10.3389/fbioe.2022.888492] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Vascular-disrupting agents are an interesting class of anticancer compounds because of their combined mode of action in preventing new blood vessel formation and disruption of already existing vasculature in the immediate microenvironment of solid tumors. The validation of vascular disruption properties of these drugs in vitro is rarely addressed due to the lack of proper in vitro angiogenesis models comprising mature and long-lived vascular-like networks. We herein report an indirect coculture model of human umbilical vein endothelial cells (HUVECs) and human dermal fibroblasts (HDFs) to form three-dimensional profuse vascular-like networks. HUVECs embedded and sandwiched in the collagen scaffold were cocultured with HDFs located outside the scaffold. The indirect coculture approach with the vascular endothelial growth factor (VEGF) producing HDFs triggered the formation of progressively maturing lumenized vascular-like networks of endothelial cells within less than 7 days, which have proven to be viably maintained in culture beyond day 21. Molecular weight-dependent Texas red-dextran permeability studies indicated high vascular barrier function of the generated networks. Their longevity allowed us to study the dose-dependent response upon treatment with the three known antiangiogenic and/or vascular disrupting agents brivanib, combretastatin A4 phosphate (CA4P), and 6´-sialylgalactose (SG) via semi-quantitative brightfield and qualitative confocal laser scanning microscopic (CLSM) image analysis. Compared to the reported data on in vivo efficacy of these drugs in terms of antiangiogenic and vascular disrupting effects, we observed similar trends with our 3D model, which are not reflected in conventional in vitro angiogenesis assays. High-vascular disruption under continuous treatment of the matured vascular-like network was observed at concentrations ≥3.5 ng·ml−1 for CA4P and ≥300 nM for brivanib. In contrast, SG failed to induce any significant vascular disruption in vitro. This advanced model of a 3D vascular-like network allows for testing single and combinational antiangiogenic and vascular disrupting effects with optimized dosing and may thus bridge the gap between the in vitro and in vivo experiments in validating hits from high-throughput screening. Moreover, the physiological 3D environment mimicking in vitro assay is not only highly relevant to in vivo studies linked to cancer but also to the field of tissue regeneration.
Collapse
Affiliation(s)
| | - Anna Laporte
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Inga Pacharzina
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Anke Hoppensack
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Hannover, Germany
- *Correspondence: Marie Weinhart, ,
| |
Collapse
|
43
|
Schumacher A, Roumans N, Rademakers T, Joris V, Eischen-Loges MJ, van Griensven M, LaPointe VL. Enhanced Microvasculature Formation and Patterning in iPSC–Derived Kidney Organoids Cultured in Physiological Hypoxia. Front Bioeng Biotechnol 2022; 10:860138. [PMID: 35782512 PMCID: PMC9240933 DOI: 10.3389/fbioe.2022.860138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/05/2022] [Indexed: 01/10/2023] Open
Abstract
Stem cell–derived kidney organoids have been shown to self-organize from induced pluripotent stem cells into most important renal structures. However, the structures remain immature in culture and contain endothelial networks with low connectivity and limited organoid invasion. Furthermore, the nephrons lose their phenotype after approximately 25 days. To become applicable for future transplantation, further maturation in vitro is essential. Since kidneys in vivo develop in hypoxia, we studied the modulation of oxygen availability in culture. We hypothesized that introducing long-term culture at physiological hypoxia, rather than the normally applied non-physiological, hyperoxic 21% O2, could initiate angiogenesis, lead to enhanced growth factor expression and improve the endothelial patterning. We therefore cultured the kidney organoids at 7% O2 instead of 21% O2 for up to 25 days and evaluated nephrogenesis, growth factor expression such as VEGF-A and vascularization. Whole mount imaging revealed a homogenous morphology of the endothelial network with enhanced sprouting and interconnectivity when the kidney organoids were cultured in hypoxia. Three-dimensional vessel quantification confirmed that the hypoxic culture led to an increased average vessel length, likely due to the observed upregulation of VEGFA-189 and VEGFA-121, and downregulation of the antiangiogenic protein VEGF-A165b measured in hypoxia. This research indicates the importance of optimization of oxygen availability in organoid systems and the potential of hypoxic culture conditions in improving the vascularization of organoids.
Collapse
Affiliation(s)
- Anika Schumacher
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Nadia Roumans
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Timo Rademakers
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Virginie Joris
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Maria José Eischen-Loges
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Martijn van Griensven
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Vanessa L.S. LaPointe
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
- *Correspondence: Vanessa L.S. LaPointe,
| |
Collapse
|
44
|
Lange M, Ohnesorge N, Hoffmann D, Rocha SF, Benedito R, Siekmann AF. Zebrafish mutants in vegfab can affect endothelial cell proliferation without altering ERK phosphorylation and are phenocopied by loss of PI3K signaling. Dev Biol 2022; 486:26-43. [PMID: 35337795 PMCID: PMC11238767 DOI: 10.1016/j.ydbio.2022.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/23/2022]
Abstract
The formation of appropriately patterned blood vessel networks requires endothelial cell migration and proliferation. Signaling through the Vascular Endothelial Growth Factor A (VEGFA) pathway is instrumental in coordinating these processes. mRNA splicing generates short (diffusible) and long (extracellular matrix bound) Vegfa isoforms. The differences between these isoforms in controlling cellular functions are not understood. In zebrafish, vegfaa generates short and long isoforms, while vegfab only generates long isoforms. We found that mutations in vegfaa had an impact on endothelial cell (EC) migration and proliferation. Surprisingly, mutations in vegfab more strongly affected EC proliferation in distinct blood vessels, such as intersegmental blood vessels in the zebrafish trunk and central arteries in the head. Analysis of downstream signaling pathways revealed no change in MAPK (ERK) activation, while inhibiting PI3 kinase signaling phenocopied vegfab mutant phenotypes in affected blood vessels. Together, these results suggest that extracellular matrix bound Vegfa might act through PI3K signaling to control EC proliferation in a distinct set of blood vessels during angiogenesis.
Collapse
Affiliation(s)
- Martin Lange
- Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, D-48149, Muenster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Muenster, Germany
| | - Nils Ohnesorge
- Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, D-48149, Muenster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Muenster, Germany
| | - Dennis Hoffmann
- Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, D-48149, Muenster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Muenster, Germany
| | - Susana F Rocha
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, E28029, Spain
| | - Rui Benedito
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, E28029, Spain
| | - Arndt F Siekmann
- Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, D-48149, Muenster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Muenster, Germany; Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
45
|
Chen F, Zhang H, Wang J. Circular RNA CircSHKBP1 accelerates the proliferation, invasion, angiogenesis, and stem cell-like properties via modulation of microR-766-5p/high mobility group AT-hook 2 axis in laryngeal squamous cell carcinoma. Bioengineered 2022; 13:11551-11563. [PMID: 35502885 PMCID: PMC9275975 DOI: 10.1080/21655979.2022.2068922] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a common malignancy in head and neck. Circular SHKBP1 (circSHKBP) exerts momentous functions in the occurrence of many cancers including LSCC. Thus, we investigated the oncogenic capacities of circSHKBP1 in LSCC, and revealed the underlying mechanism as a competing endogenous RNA. The expression levels of circSHKBP1, miR-766-5p, and high mobility group AT-hook 2 (HMGA2) were examined by quantitative real-time PCR and their influences on the overall survival were measured by Kaplan–Meier method. The correlations between circSHKBP1 and miR-766-5p or HMGA2 were detected by Spearman’s rank correlation analysis. In vitro, the influences of circSHKBP1/miR-766-5p/HMGA2 axis on the tumorigenesis of LSCC were examined by CCK-8, transwell, sphere formation, and angiogenesis assays, respectively. circSHKBP1 expression was up-regulated in the LSCC specimens and cell lines. And elevated circSHKBP1 expression was closely linked to poor prognosis. Silencing circSHKBP1 expression restrained cell proliferation, invasion, angiogenesis, stem cell-like properties and tumor growth. We observed that miR-766-5p was down-regulated and negatively correlated to circSHKBP1 in LSCC samples. However, HMGA2 was highly expressed and positively associated with circSHKBP1 in these specimens. Importantly, the levels of circSHKBP1, miR-766-5p, and HMGA2 were closely associated with patients’ clinical parameters including lymph nodes metastasis and TNM stages. Mechanistic analysis clarified that circSHKBP1 sponged miR-766-5p to regulate HMGA2, the target of miR-766-5p. Moreover, miR-766-5p inhibition and overexpression of HMGA2 rescued the tumor-suppressing roles of circSHKBP1 downregulation in LSCC. In conclusion, circSHKBP1 accelerated the tumorigenesis of LSCC via modulating HMGA2 by targeting miR-766-5p.
Collapse
Affiliation(s)
- Fu Chen
- Department of Radiation Oncology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Haiyan Zhang
- Department of Radiation Oncology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Jie Wang
- Department of Radiation Oncology, Eye & ENT Hospital of Fudan University, Shanghai, China
| |
Collapse
|
46
|
Fibrosis of Peritoneal Membrane as Target of New Therapies in Peritoneal Dialysis. Int J Mol Sci 2022; 23:ijms23094831. [PMID: 35563220 PMCID: PMC9102299 DOI: 10.3390/ijms23094831] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 01/27/2023] Open
Abstract
Peritoneal dialysis (PD) is an efficient renal replacement therapy for patients with end-stage renal disease. Even if it ensures an outcome equivalent to hemodialysis and a better quality of life, in the long-term, PD is associated with the development of peritoneal fibrosis and the consequents patient morbidity and PD technique failure. This unfavorable effect is mostly due to the bio-incompatibility of PD solution (mainly based on high glucose concentration). In the present review, we described the mechanisms and the signaling pathway that governs peritoneal fibrosis, epithelial to mesenchymal transition of mesothelial cells, and angiogenesis. Lastly, we summarize the present and future strategies for developing more biocompatible PD solutions.
Collapse
|
47
|
Eleftheriadou D, Berg M, Phillips JB, Shipley RJ. A combined experimental and computational framework to evaluate the behavior of therapeutic cells for peripheral nerve regeneration. Biotechnol Bioeng 2022; 119:1980-1996. [PMID: 35445744 PMCID: PMC9323509 DOI: 10.1002/bit.28105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/22/2022] [Accepted: 04/08/2022] [Indexed: 11/08/2022]
Abstract
Recent studies have explored the potential of tissue‐mimetic scaffolds in encouraging nerve regeneration. One of the major determinants of the regenerative success of cellular nerve repair constructs (NRCs) is the local microenvironment, particularly native low oxygen conditions which can affect implanted cell survival and functional performance. In vivo, cells reside in a range of environmental conditions due to the spatial gradients of nutrient concentrations that are established. Here we evaluate in vitro the differences in cellular behavior that such conditions induce, including key biological features such as oxygen metabolism, glucose consumption, cell death, and vascular endothelial growth factor secretion. Experimental measurements are used to devise and parameterize a mathematical model that describes the behavior of the cells. The proposed model effectively describes the interactions between cells and their microenvironment and could in the future be extended, allowing researchers to compare the behavior of different therapeutic cells. Such a combinatorial approach could be used to accelerate the clinical translation of NRCs by identifying which critical design features should be optimized when fabricating engineered nerve repair conduits.
Collapse
Affiliation(s)
- D Eleftheriadou
- Centre for Nerve Engineering, University College London, London, WC1E 6B.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX.,Department of Mechanical Engineering, University College London, London, WC1E 7JE
| | - M Berg
- Centre for Nerve Engineering, University College London, London, WC1E 6B.,Department of Mechanical Engineering, University College London, London, WC1E 7JE
| | - J B Phillips
- Centre for Nerve Engineering, University College London, London, WC1E 6B.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX
| | - R J Shipley
- Centre for Nerve Engineering, University College London, London, WC1E 6B.,Department of Mechanical Engineering, University College London, London, WC1E 7JE
| |
Collapse
|
48
|
Ansari MJ, Bokov D, Markov A, Jalil AT, Shalaby MN, Suksatan W, Chupradit S, AL-Ghamdi HS, Shomali N, Zamani A, Mohammadi A, Dadashpour M. Cancer combination therapies by angiogenesis inhibitors; a comprehensive review. Cell Commun Signal 2022; 20:49. [PMID: 35392964 PMCID: PMC8991477 DOI: 10.1186/s12964-022-00838-y] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Abnormal vasculature is one of the most conspicuous traits of tumor tissue, largely contributing to tumor immune evasion. The deregulation mainly arises from the potentiated pro-angiogenic factors secretion and can also target immune cells' biological events, such as migration and activation. Owing to this fact, angiogenesis blockade therapy was established to fight cancer by eliminating the nutrient and oxygen supply to the malignant cells by impairing the vascular network. Given the dominant role of vascular-endothelium growth factor (VEGF) in the angiogenesis process, the well-known anti-angiogenic agents mainly depend on the targeting of its actions. However, cancer cells mainly show resistance to anti-angiogenic agents by several mechanisms, and also potentiated local invasiveness and also distant metastasis have been observed following their administration. Herein, we will focus on clinical developments of angiogenesis blockade therapy, more particular, in combination with other conventional treatments, such as immunotherapy, chemoradiotherapy, targeted therapy, and also cancer vaccines. Video abstract.
Collapse
Affiliation(s)
- Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - Dmitry Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, 119991 Russian Federation
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr., Moscow, 109240 Russian Federation
| | - Alexander Markov
- Tyumen State Medical University, Tyumen, Russian Federation
- Industrial University, Tyumen, Russian Federation
| | - Abduladheem Turki Jalil
- Faculty of Biology and Ecology, Yanka Kupala State University of Grodno, 230023 Grodno, Belarus
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Dentistry, Kut University College, Kut, Wasit 52001 Iraq
| | - Mohammed Nader Shalaby
- Biological Sciences and Sports Health Department, Faculty of Physical Education, Suez Canal University, Ismailia, Egypt
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Hasan S. AL-Ghamdi
- Internal Medicine Department, Division of Dermatology, Albaha University, Al Bahah, Kingdom of Saudi Arabia
| | - Navid Shomali
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zamani
- Shiraz Transplant Center, Abu Ali Sina Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammadi
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Mehdi Dadashpour
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
49
|
ABSTRACTS (BY NUMBER). Tissue Eng Part A 2022. [DOI: 10.1089/ten.tea.2022.29025.abstracts] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
50
|
Mezu-Ndubuisi OJ, Maheshwari A. Role of the Endothelium in Neonatal Diseases. NEWBORN 2022; 1:44-57. [PMID: 35754998 PMCID: PMC9217741 DOI: 10.5005/jp-journals-11002-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In both fetal and neonatal physiologic and pathologic processes in most organs, endothelial cells are known to play critical roles. Although the endothelium is one of the most ubiquitous cell type in the body, the tight adherence to the blood vessel wall has made it difficult to study their diverse function and structure. In this article, we have reviewed endothelial cell origins and explored their heterogeneity in terms of structure, function, developmental changes, and their role in inflammatory and infectious diseases. We have also attempted to evaluate the untapped therapeutic potentials of endothelial cells in neonatal disease. This article comprises various peer-reviewed studies, including ours, and an extensive database literature search from EMBASE, PubMed, and Scopus.
Collapse
Affiliation(s)
- Olachi J Mezu-Ndubuisi
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Akhil Maheshwari
- Global Newborn Society, Clarksville, Maryland, United States of America
| |
Collapse
|