1
|
Staples SCR, Yin H, Sutherland FSK, Prescott EK, Tinney D, Hamilton DW, Goldman D, Poepping TL, Ellis CG, Pickering JG. Intussusceptive angiogenesis-on-a-chip: Evidence for transluminal vascular bridging by endothelial delamination. Proc Natl Acad Sci U S A 2025; 122:e2423700122. [PMID: 40244661 PMCID: PMC12036988 DOI: 10.1073/pnas.2423700122] [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: 12/07/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Intussusceptive angiogenesis is an increasingly recognized vessel duplication process that generates and reshapes microvascular beds. However, the mechanism by which a vessel splits into two is poorly understood. Particularly vexing is formation of the hallmark transluminal endothelial cell bridge. How an endothelial cell comes to cross a flowing lumen rather than line it is enigmatic. To elucidate this, we used a microvessel-on-a-chip strategy, creating a microconduit coherently lined with flow-sensitive endothelial cells but in which transluminal bridges also formed. Bridge morphologies ranged from filamentous strand to multicellular columns with a central extracellular matrix-containing core. These bridge architectures were found to recapitulate those in microvessels in embryos, tumors, diseased organs, and the dermis of patients with limb-threatening ischemia. Time-lapse, multiplane, three-dimensional (3D) microscopy of the microphysiologic conduit revealed that bridges arose from endothelial cells oriented orthogonal to flow that partially released from the wall while retaining attachments at the ends. This delamination process was blocked by hyperactivation of Rho and augmented by interventions that weaken cell-substrate interactions, including inhibiting nonmuscle myosin II and blocking α5ß1 integrin. Thus, endothelial cells can leave their monolayer and transect a flowing lumen through controlled delamination. This previously unrecognized lumen entry program could explain the launch of intussusceptive angiogenesis and opens a framework for intervening.
Collapse
Affiliation(s)
- Sabrina C. R. Staples
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Frances S. K. Sutherland
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Emma K. Prescott
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Dylan Tinney
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Douglas W. Hamilton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Daniel Goldman
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Tamie L. Poepping
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Physics and Astronomy, Western University, London, ONN6A 3K7, Canada
| | - Christopher G. Ellis
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - J. Geoffrey Pickering
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- London Health Sciences Centre, London, ONN6A 5A5, Canada
| |
Collapse
|
2
|
Belyaev IB, Griaznova OY, Yaremenko AV, Deyev SM, Zelepukin IV. Beyond the EPR effect: Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv Drug Deliv Rev 2025; 219:115550. [PMID: 40021012 DOI: 10.1016/j.addr.2025.115550] [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: 10/31/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Delivery of nanoparticles (NPs) to solid tumors has long relied on enhanced permeability and retention (EPR) effect, involving permeation of NPs through a leaky vasculature with prolonged retention by reduced lymphatic drainage in tumor. Recent research studies and clinical data challenge EPR concept, revealing alternative pathways and approaches of NP delivery. The area was significantly impacted by the implementation of intravital optical microscopy, unraveling delivery mechanisms at cellular level in vivo. This review presents analysis of the reasons for EPR heterogeneity in tumors and describes non-EPR based concepts for drug delivery, which can supplement the current paradigm. One of the approaches is targeting tumor endothelium by NPs with subsequent intravascular drug release and gradient-driven drug transport to tumor interstitium. Others exploit various immune cells for tumor infiltration and breaking endothelial barriers. Finally, we discuss the involvement of active transcytosis through endothelial cells in NP delivery. This review aims to inspire further understanding of the process of NP extravasation in tumors and provide insights for developing next-generation nanomedicines with improved delivery.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75123, Sweden.
| |
Collapse
|
3
|
Wu X, Fei W, Shen T, Ye L, Li C, Chu S, Liu M, Cheng X, Qin J. Unveiling the potential of biomechanics in pioneering innovative strategies for cancer therapy. Theranostics 2025; 15:2903-2932. [PMID: 40083943 PMCID: PMC11898300 DOI: 10.7150/thno.108605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/02/2025] [Indexed: 03/16/2025] Open
Abstract
Mechanical force transmission is pivotal in tumor biology, profoundly affecting cancer cell behaviors such as proliferation, metastasis, and resistance to therapy. To explore novel biomechanical-based therapeutic strategies for cancer treatment, this paper deciphers the advances in biomechanical measurement approaches and the impact of biomechanical signals on fundamental oncological processes such as tumor microenvironment remodeling, angiogenesis, metastasis, and drug resistance. Then, the mechanisms of biomechanical signal transduction of tumor cells are demonstrated to identify novel targets for tumor therapy. Additionally, this study proposes a novel tumor treatment strategy, the biomechanical regulation tumor nanotherapeutics, including smart biomaterials designed to disturb mechanical signaling pathways and innovative nanodrugs that interfere transduction of biomechanical signals to improve tumor therapeutic outcomes. These methods mark a departure from conventional pharmacological therapies to novel strategies that utilize mechanical forces to impede tumor progression and enhance tumor responsiveness to treatment. In general, this review highlights the critical role of biomechanical signals in cancer biology from a holistic perspective and underscores the potential of biomechanical interventions as a transformative class of therapeutics. By integrating mechanobiology into the development of cancer treatments, this paper paves the way for more precise and effective strategies that leverage the inherent physical properties of the tumor microenvironment.
Collapse
Affiliation(s)
- Xiaodong Wu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Weidong Fei
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Tao Shen
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lei Ye
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Chaoqun Li
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Siran Chu
- Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Mingqi Liu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiaodong Cheng
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou, 310006, China
| | - Jiale Qin
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou, 310006, China
| |
Collapse
|
4
|
Hadjigeorgiou AG, Stylianopoulos T. Hybrid model of tumor growth, angiogenesis and immune response yields strategies to improve antiangiogenic therapy. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2024; 1:4. [PMID: 39759959 PMCID: PMC11698377 DOI: 10.1038/s44341-024-00002-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/31/2024] [Indexed: 01/07/2025]
Abstract
Solid tumors harbor a complex and dynamic microenvironment that hinders the delivery and efficacy of therapeutic interventions. In this study, we developed and utilized a hybrid, discrete-continuous mathematical model to explore the interplay between solid tumor growth, immune response, tumor-induced angiogenesis, and antiangiogenic drugs. By integrating published data with anti-angiogenic drugs, we elucidate three primary mechanisms by which anti-angiogenesis influences tumor progression and treatment outcomes: reduction in tumor growth rate by mitigating and temporally delaying angiogenesis, normalization of blood vessel structure and function, and improving immune cell extravasation and activation. Our results indicate a significant increase in functional blood vessels and perfusion following anti-angiogenic treatment, which in turn improves the intratumoral distribution of immune cells. The normalization window, or optimal time frame for anti-angiogenic drug administration, and the dose of the drug arise naturally in the model and are highlighted as crucial factors in maximizing treatment benefits. Prolonged anti-angiogenic treatment triggers cancer cell migration into healthy tissue and induces immunosuppression due to hypoxia, potentially leading to negative effects because these cancer cells will rapidly proliferate upon treatment termination. In conclusion, the positive contribution of anti-angiogenic treatment must balance the possible negative effects by choosing a proper treatment protocol as well as combining it with proper anti-cancer treatment. Our findings provide valuable insights and a framework for the design of protocols with anti-angiogenic treatment, targeted immunotherapy, and non-targeted anti-cancer therapies.
Collapse
Affiliation(s)
- Andreas G. Hadjigeorgiou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
5
|
Xia M, Stegmeyer RI, Shirakura K, Butz S, Thiriot A, von Andrian UH, Vestweber D. Conditions that promote transcellular neutrophil migration in vivo. Sci Rep 2024; 14:14471. [PMID: 38914623 PMCID: PMC11196655 DOI: 10.1038/s41598-024-65173-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Circulating leukocytes enter tissue either through endothelial junctions (paracellular) or via a pore through the body of endothelial cells (transcellular). We have previously shown that genetically replacing VE-cadherin with a VE-cadherin-α-catenin (VEC-αC) fusion construct-which binds constitutively to actin-obstructs junctions, and blocks leukocyte extravasation in lung, skin and postcapillary venules of cremaster muscle. However, neutrophil recruitment into the inflamed peritoneal cavity was unimpaired. Investigating reasons for this, here, we visualized neutrophil diapedesis by 3D intravital video microscopy in the cremaster muscle and omentum, the major site of neutrophil recruitment into the peritoneal cavity. We found that 80% of neutrophil-extravasation occurred through HEVs in the omentum, which was unimpaired by VEC-αC. In addition, in larger venules (60-85 µm) of both tissues, less than 15% of neutrophils extravasated transcellularly in WT mice. However, in VEC-α-C mice, transcellular diapedesis increased severalfold in the omentum, but not in the cremaster. In line with this, omental venules expressed higher levels of ICAM-1 and atypical chemokine receptor 1. Furthermore, only in the omentum, VEC-αC expression caused reduced elongation of venular endothelium in flow-direction, suggesting different biomechanical properties. Collectively, VEC-αC does not inhibit paracellular transmigration in all types of venules and can modulate the diapedesis route.
Collapse
Affiliation(s)
- Min Xia
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Rebekka I Stegmeyer
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Keisuke Shirakura
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Stefan Butz
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Aude Thiriot
- Department of Immunology and Center for Immune Imaging, Harvard Medical School, Boston, MA, USA
| | - Ulrich H von Andrian
- Department of Immunology and Center for Immune Imaging, Harvard Medical School, Boston, MA, USA
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany.
| |
Collapse
|
6
|
Yang J, Cherin E, Yin J, Dayton PA, Foster FS, Demore CEM. Superharmonic and Microultrasound Imaging With Plane Wave Beamforming Techniques. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2023; 70:1442-1456. [PMID: 37713228 PMCID: PMC10712286 DOI: 10.1109/tuffc.2023.3316120] [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] [Indexed: 09/16/2023]
Abstract
Superharmonic contrast imaging (SpHI) suppresses tissue clutter and allows high-contrast visualization of the vasculature. An array-based dual-frequency (DF) probe has been developed for SpHI, integrating a 21-MHz, 256-element microultrasound imaging array with a 2-MHz, 32-element array to take advantage of the broadband nonlinear responses from microbubble (MB) contrast agents. In this work, ultrafast imaging with plane waves was implemented for SpHI to increase the acquisition frame rate. Ultrafast imaging was also implemented for microultrasound B-mode imaging (HFPW B-mode) to enable high-resolution visualization of the tissue structure. Coherent compounding was demonstrated in vitro and in vivo in both imaging modes. Acquisition frame rates of 4.5 kHz and 187 Hz in HFPW B-mode imaging were achieved for imaging up to 21 mm with one and 25 angles, respectively, and 3.5 kHz and 396 Hz in the SpHI mode with one and nine coherently compounded angles, respectively. SpHI images showed suppression of tissue clutter prior to and after the introduction of MBs in vitro and in vivo. The nine-angle coherently compounded 2-D SpHI images of contrast-filled flow channel showed a contrast-to-tissue ratio (CTR) of 26.0 dB, a 2.5-dB improvement relative to images reconstructed from 0° steering. Consistent with in vitro imaging, the nine-angle compounded 2-D SpHI of a Lewis lung cancer tumor showed a 2.6-dB improvement in contrast enhancement, relative to 0° steering, and additionally revealed a region of nonviable tissue. The 3-D display of the volumetric SpHI data acquired from a xenograft mouse tumor using both 0° steering and nine-angle compounding allowed the visualization of the tumor vasculature. A small vessel visible in the compounded SpHI image, measuring around [Formula: see text], is not visualized in the 0° steering SpHI image, demonstrating the superiority of the latter in detecting fine structures within the tumor.
Collapse
|
7
|
Bao L, Kong H, Ja Y, Wang C, Qin L, Sun H, Dai S. The relationship between cancer and biomechanics. Front Oncol 2023; 13:1273154. [PMID: 37901315 PMCID: PMC10602664 DOI: 10.3389/fonc.2023.1273154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
The onset, development, diagnosis, and treatment of cancer involve intricate interactions among various factors, spanning the realms of mechanics, physics, chemistry, and biology. Within our bodies, cells are subject to a variety of forces such as gravity, magnetism, tension, compression, shear stress, and biological static force/hydrostatic pressure. These forces are perceived by mechanoreceptors as mechanical signals, which are then transmitted to cells through a process known as mechanical transduction. During tumor development, invasion and metastasis, there are significant biomechanical influences on various aspects such as tumor angiogenesis, interactions between tumor cells and the extracellular matrix (ECM), interactions between tumor cells and other cells, and interactions between tumor cells and the circulatory system and vasculature. The tumor microenvironment comprises a complex interplay of cells, ECM and vasculature, with the ECM, comprising collagen, fibronectins, integrins, laminins and matrix metalloproteinases, acting as a critical mediator of mechanical properties and a key component within the mechanical signaling pathway. The vasculature exerts appropriate shear forces on tumor cells, enabling their escape from immune surveillance, facilitating their dissemination in the bloodstream, dictating the trajectory of circulating tumor cells (CTCs) and playing a pivotal role in regulating adhesion to the vessel wall. Tumor biomechanics plays a critical role in tumor progression and metastasis, as alterations in biomechanical properties throughout the malignant transformation process trigger a cascade of changes in cellular behavior and the tumor microenvironment, ultimately culminating in the malignant biological behavior of the tumor.
Collapse
Affiliation(s)
- Liqi Bao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongru Kong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yang Ja
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengchao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Qin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongwei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shengjie Dai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
8
|
Kim G, Park HS, Shin P, Eom T, Yoon JH, Jeong Y, Oh WY. Direct Blood Cell Flow Imaging in Microvascular Networks. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302244. [PMID: 37309282 DOI: 10.1002/smll.202302244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/19/2023] [Indexed: 06/14/2023]
Abstract
Blood flow dynamics in microvascular networks are intimately related to the health of tissues and organs. While numerous imaging modalities and techniques have been developed to assess blood flow dynamics for various applications, their utilization has been hampered by limited imaging speed and indirect quantification of blood flow dynamics. Here, direct blood cell flow imaging (DBFI) is demonstrated that provides visualization of individual motions of blood cells over a field of 0.71 mm × 1.42 mm with a time resolution of 0.69 ms (1450 frames s-1 ) without using any exogenous agents. DBFI enables precise dynamic analysis of blood cell flow velocities and fluxes in various vessels over a large field, from capillaries to arteries and veins, with unprecedented time resolution. Three exemplary applications of DBFI, quantification of blood flow dynamics of 3D vascular networks, analysis of heartbeat induced blood flow dynamics, and analysis of blood flow dynamics of neurovascular coupling, illustrate the potential of this new imaging technology.
Collapse
Affiliation(s)
- Gyounghwan Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Hyun-Sang Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Paul Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Taeguk Eom
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Jin-Hui Yoon
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Yong Jeong
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Wang-Yuhl Oh
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| |
Collapse
|
9
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
10
|
Lin Q, Choyke PL, Sato N. Visualizing vasculature and its response to therapy in the tumor microenvironment. Theranostics 2023; 13:5223-5246. [PMID: 37908739 PMCID: PMC10614675 DOI: 10.7150/thno.84947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/30/2023] [Indexed: 11/02/2023] Open
Abstract
Tumor vasculature plays a critical role in the progression and metastasis of tumors, antitumor immunity, drug delivery, and resistance to therapies. The morphological and functional changes of tumor vasculature in response to therapy take place in a spatiotemporal-dependent manner, which can be predictive of treatment outcomes. Dynamic monitoring of intratumor vasculature contributes to an improved understanding of the mechanisms of action of specific therapies or reasons for treatment failure, leading to therapy optimization. There is a rich history of methods used to image the vasculature. This review describes recent advances in imaging technologies to visualize the tumor vasculature, with a focus on enhanced intravital imaging techniques and tumor window models. We summarize new insights on spatial-temporal vascular responses to various therapies, including changes in vascular perfusion and permeability and immune-vascular crosstalk, obtained from intravital imaging. Finally, we briefly discuss the clinical applications of intravital imaging techniques.
Collapse
Affiliation(s)
| | | | - Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
11
|
Meng L, Huang M, Feng S, Wang Y, Lu J, Li P. Optical Flow-Based Full-Field Quantitative Blood-Flow Velocimetry Using Temporal Direction Filtering and Peak Interpolation. Int J Mol Sci 2023; 24:12048. [PMID: 37569421 PMCID: PMC10419297 DOI: 10.3390/ijms241512048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/15/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
The quantitative measurement of the microvascular blood-flow velocity is critical to the early diagnosis of microvascular dysfunction, yet there are several challenges with the current quantitative flow velocity imaging techniques for the microvasculature. Optical flow analysis allows for the quantitative imaging of the blood-flow velocity with a high spatial resolution, using the variation in pixel brightness between consecutive frames to trace the motion of red blood cells. However, the traditional optical flow algorithm usually suffers from strong noise from the background tissue, and a significant underestimation of the blood-flow speed in blood vessels, due to the errors in detecting the feature points in optical images. Here, we propose a temporal direction filtering and peak interpolation optical flow method (TPIOF) to suppress the background noise, and improve the accuracy of the blood-flow velocity estimation. In vitro phantom experiments and in vivo animal experiments were performed to validate the improvements in our new method.
Collapse
Affiliation(s)
- Liangwei Meng
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Mange Huang
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Shijie Feng
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Yiqian Wang
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Jinling Lu
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Pengcheng Li
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
- Department of Biomedical Engineering, Hainan University, Haikou 570228, China
| |
Collapse
|
12
|
Guo Y, Wang L, Luo Z, Zhu Y, Gao X, Weng X, Wang Y, Yan W, Qu J. Dynamic Volumetric Imaging of Mouse Cerebral Blood Vessels In Vivo with an Ultralong Anti-Diffracting Beam. Molecules 2023; 28:4936. [PMID: 37446598 DOI: 10.3390/molecules28134936] [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: 05/21/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Volumetric imaging of a mouse brain in vivo with one-photon and two-photon ultralong anti-diffracting (UAD) beam illumination was performed. The three-dimensional (3D) structure of blood vessels in the mouse brain were mapped to a two-dimensional (2D) image. The speed of volumetric imaging was significantly improved due to the long focal length of the UAD beam. Comparing one-photon and two-photon UAD beam volumetric imaging, we found that the imaging depth of two-photon volumetric imaging (80 μm) is better than that of one-photon volumetric imaging (60 μm), and the signal-to-background ratio (SBR) of two-photon volumetric imaging is two times that of one-photon volumetric imaging. Therefore, we used two-photon UAD volumetric imaging to perform dynamic volumetric imaging of mouse brain blood vessels in vivo, and obtained the blood flow velocity.
Collapse
Affiliation(s)
- Yong Guo
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Luwei Wang
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Ziyi Luo
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Yinru Zhu
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Xinwei Gao
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Xiaoyu Weng
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Yiping Wang
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Wei Yan
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Junle Qu
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
13
|
Deán-Ben XL, Robin J, Nozdriukhin D, Ni R, Zhao J, Glück C, Droux J, Sendón-Lago J, Chen Z, Zhou Q, Weber B, Wegener S, Vidal A, Arand M, El Amki M, Razansky D. Deep optoacoustic localization microangiography of ischemic stroke in mice. Nat Commun 2023; 14:3584. [PMID: 37328490 PMCID: PMC10275987 DOI: 10.1038/s41467-023-39069-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 05/24/2023] [Indexed: 06/18/2023] Open
Abstract
Super-resolution optoacoustic imaging of microvascular structures deep in mammalian tissues has so far been impeded by strong absorption from densely-packed red blood cells. Here we devised 5 µm biocompatible dichloromethane-based microdroplets exhibiting several orders of magnitude higher optical absorption than red blood cells at near-infrared wavelengths, thus enabling single-particle detection in vivo. We demonstrate non-invasive three-dimensional microangiography of the mouse brain beyond the acoustic diffraction limit (<20 µm resolution). Blood flow velocity quantification in microvascular networks and light fluence mapping was also accomplished. In mice affected by acute ischemic stroke, the multi-parametric multi-scale observations enabled by super-resolution and spectroscopic optoacoustic imaging revealed significant differences in microvascular density, flow and oxygen saturation in ipsi- and contra-lateral brain hemispheres. Given the sensitivity of optoacoustics to functional, metabolic and molecular events in living tissues, the new approach paves the way for non-invasive microscopic observations with unrivaled resolution, contrast and speed.
Collapse
Affiliation(s)
- Xosé Luís Deán-Ben
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich, Switzerland.
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland.
| | - Justine Robin
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Daniil Nozdriukhin
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Ruiqing Ni
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
- Zurich Neuroscience Center, Zurich, Switzerland
| | - Jim Zhao
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Chaim Glück
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Jeanne Droux
- Zurich Neuroscience Center, Zurich, Switzerland
- Department of Neurology, University Hospital and University of Zurich and University of Zurich, Zurich, Switzerland
| | - Juan Sendón-Lago
- Experimental Biomedicine Centre (CEBEGA), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Zhenyue Chen
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Quanyu Zhou
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Bruno Weber
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Susanne Wegener
- Zurich Neuroscience Center, Zurich, Switzerland
- Department of Neurology, University Hospital and University of Zurich and University of Zurich, Zurich, Switzerland
| | - Anxo Vidal
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael Arand
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Mohamad El Amki
- Zurich Neuroscience Center, Zurich, Switzerland
- Department of Neurology, University Hospital and University of Zurich and University of Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich, Switzerland.
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland.
- Zurich Neuroscience Center, Zurich, Switzerland.
| |
Collapse
|
14
|
Ye S, Zhu Y, Zhong D, Song X, Li J, Xiao F, Huang Z, Zhang W, Wu M, Zhang K, Xiang FL, Xu J. G protein-coupled receptor GPR68 inhibits lymphocyte infiltration and contributes to gender-dependent melanoma growth. Front Oncol 2023; 13:1202750. [PMID: 37350933 PMCID: PMC10282648 DOI: 10.3389/fonc.2023.1202750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/15/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction Melanoma is a common and aggressive type of skin cancer with rising incidence rate globally. Gender is one of the determining factors, and overall males have a higher risk of developing melanoma as well as worse prognosis. Emerging evidence show that GPR68, a G protein-coupled receptor that is sensitive to acid and mechanical stimulations for cellular microenvironment, plays an important role in tumor biology. However, whether GPR68 is involved in gender-dependent regulation of tumor growth is unclear. Methods We established a syngeneic melanoma model in Gpr68-deficient mice and investigated tumor growth in males and females. The GPR68 activation-induced cellular responses of melanocytes, including intracellular calcium dynamics, proliferation and migration were measured. The landscape of tumor-infiltrating immune cells were analyzed by flow cytometry and the expression various cytokines were checked by qRT-PCR. Results GPR68 is required for melanoma growth in males but dispensable in females. GPR68 is expressed and functional in B16-F10 melanocytes, but the activity of the receptor does not directly contribute to proliferation and migration of the cells. GPR68 inhibits infiltration of CD45+ lymphocytes, CD8+ T cells and NK cells in melanoma in male mice, but has no apparent effect in females. Furthermore, GPR68 functionally inhibits the expression of IFNγ in the tumor infiltrating CD8+ T cells and NK cells as well as the inflammatory cytokine expression in the spleen in male mice but not in females. Our results show the gender-dependent modulatory effect of GPR68 on tumor-infiltrating immune cells and their tumor-killing capacity. Discussion GPR68 is sensor for acid and mechanical stimulations, which are two important factors in the microenvironment associated with tumor growth and metastasis. Our results suggest a prominent role of the receptor molecules in tumor biology in a gender-dependent manner. Since GPCRs are more feasible to develop small molecule drugs compared to transcription factors, our study demonstrates the potential of GPR68 as a novel druggable therapeutic target for melanoma in male patients.
Collapse
Affiliation(s)
- Shangmei Ye
- Institute of Precision Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yunfeng Zhu
- Institute of Precision Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dongmei Zhong
- Institute of Precision Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaodong Song
- Department of Critical Care Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jialin Li
- Department of Critical Care Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fang Xiao
- Department of Critical Care Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhilei Huang
- Institute of Precision Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenjie Zhang
- Institute of Precision Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mingyue Wu
- Institute of Precision Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kangdi Zhang
- Institute of Precision Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fu-li Xiang
- Institute of Precision Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jie Xu
- Institute of Precision Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Kurma K, Alix-Panabières C. Mechanobiology and survival strategies of circulating tumor cells: a process towards the invasive and metastatic phenotype. Front Cell Dev Biol 2023; 11:1188499. [PMID: 37215087 PMCID: PMC10196185 DOI: 10.3389/fcell.2023.1188499] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Metastatic progression is the deadliest feature of cancer. Cancer cell growth, invasion, intravasation, circulation, arrest/adhesion and extravasation require specific mechanical properties to allow cell survival and the completion of the metastatic cascade. Circulating tumor cells (CTCs) come into contact with the capillary bed during extravasation/intravasation at the beginning of the metastatic cascade. However, CTC mechanobiology and survival strategies in the bloodstream, and specifically in the microcirculation, are not well known. A fraction of CTCs can extravasate and colonize distant areas despite the biomechanical constriction forces that are exerted by the microcirculation and that strongly decrease tumor cell survival. Furthermore, accumulating evidence shows that several CTC adaptations, via molecular factors and interactions with blood components (e.g., immune cells and platelets inside capillaries), may promote metastasis formation. To better understand CTC journey in the microcirculation as part of the metastatic cascade, we reviewed how CTC mechanobiology and interaction with other cell types in the bloodstream help them to survive the harsh conditions in the circulatory system and to metastasize in distant organs.
Collapse
Affiliation(s)
- Keerthi Kurma
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (E LBS), Hamburg, Germany
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (E LBS), Hamburg, Germany
| |
Collapse
|
16
|
Giblin JT, Park SW, Jiang J, Kılıç K, Kura S, Tang J, Boas DA, Chen IA. Measuring capillary flow dynamics using interlaced two-photon volumetric scanning. J Cereb Blood Flow Metab 2023; 43:595-609. [PMID: 36495178 PMCID: PMC10063827 DOI: 10.1177/0271678x221145091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Two photon microscopy and optical coherence tomography (OCT) are two standard methods for measuring flow speeds of red blood cells in microvessels, particularly in animal models. However, traditional two photon microscopy lacks the depth of field to adequately capture the full volumetric complexity of the cerebral microvasculature and OCT lacks the specificity offered by fluorescent labeling. In addition, the traditional raster scanning technique utilized in both modalities requires a balance of image frame rate and field of view, which severely limits the study of RBC velocities in the microvascular network. Here, we overcome this by using a custom two photon system with an axicon based Bessel beam to obtain volumetric images of the microvascular network with fluorescent specificity. We combine this with a novel scan pattern that generates pairs of frames with short time delay sufficient for tracking red blood cell flow in capillaries. We track RBC flow speeds in 10 or more capillaries simultaneously at 1 Hz in a 237 µm × 237 µm × 120 µm volume and quantified both their spatial and temporal variability in speed. We also demonstrate the ability to track flow speed changes around stalls in capillary flow and measure to 300 µm in depth.
Collapse
Affiliation(s)
- John T Giblin
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Seong-Wook Park
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - John Jiang
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Kıvılcım Kılıç
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sreekanth Kura
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Jianbo Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - David A Boas
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Ichun A Chen
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| |
Collapse
|
17
|
Qu H, Wang K, Lin Z, Li S, Tang C, Yin C. Cellulose nanocrystal as an enhancing core for antitumor polymeric micelles to overcome biological barriers. Int J Biol Macromol 2023; 238:124337. [PMID: 37030467 DOI: 10.1016/j.ijbiomac.2023.124337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/13/2023] [Accepted: 04/02/2023] [Indexed: 04/09/2023]
Abstract
Polymeric micelles are extensively studied nanocarriers to improve the solubility, blood circulation, biodistribution, and adverse effects of chemotherapeutic drugs. However, the antitumor efficacy of polymeric micelles is often restricted due to multiple biological barriers, including blood fluid shear stress (FSS) and limited tumor penetration in vivo. Herein, cellulose nanocrystal (CNC) as a green material with rigidity and rod-shaped structure is developed to be an enhancing core for polymeric micelles to overcome these biological barriers. Doxorubicin (DOX) loaded methoxy poly (ethylene glycol)-block-poly (D, L-lactic acid) (mPEG-PLA, PP) ligated CNC nanoparticles (PPC/DOX NPs) are fabricated via one-pot synthesis. In comparison to the self-assembled DOX loaded mPEG-PLA micelles (PP/DOX NPs), PPC/DOX NPs exhibit remarkable improvements in FSS resistance, cellular internalization, blood circulation, tumor penetration, and antitumor efficacy owing to the unique rigidity and rod-shaped structure of CNC core. Moreover, PPC/DOX NPs present various advantages beyond DOX·HCl and CNC/DOX NPs. The superiority of PPC/DOX NPs in antitumor efficacy reveals the effectiveness of adopting CNC as the enhancing core for polymeric micelles, suggesting that CNC is a promising biomaterial in advancing nanomedicine.
Collapse
Affiliation(s)
- Hongfei Qu
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ke Wang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ziyun Lin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Shengqi Li
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
18
|
Pan Y, Park K, Ren J, Volkow ND, Ling H, Koretsky AP, Du C. Dynamic 3D imaging of cerebral blood flow in awake mice using self-supervised-learning-enhanced optical coherence Doppler tomography. Commun Biol 2023; 6:298. [PMID: 36944712 PMCID: PMC10030663 DOI: 10.1038/s42003-023-04656-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/03/2023] [Indexed: 03/23/2023] Open
Abstract
Cerebral blood flow (CBF) is widely used to assess brain function. However, most preclinical CBF studies have been performed under anesthesia, which confounds findings. High spatiotemporal-resolution CBF imaging of awake animals is challenging due to motion artifacts and background noise, particularly for Doppler-based flow imaging. Here, we report ultrahigh-resolution optical coherence Doppler tomography (µODT) for 3D imaging of CBF velocity (CBFv) dynamics in awake mice by developing self-supervised deep-learning for effective image denoising and motion-artifact removal. We compare cortical CBFv in awake vs. anesthetized mice and their dynamic responses in arteriolar, venular and capillary networks to acute cocaine (1 mg/kg, i.v.), a highly addictive drug associated with neurovascular toxicity. Compared with awake, isoflurane (2-2.5%) induces vasodilation and increases CBFv within 2-4 min, whereas dexmedetomidine (0.025 mg/kg, i.p.) does not change vessel diameters nor flow. Acute cocaine decreases CBFv to the same extent in dexmedetomidine and awake states, whereas decreases are larger under isoflurane, suggesting that isoflurane-induced vasodilation might have facilitated detection of cocaine-induced vasoconstriction. Awake mice after chronic cocaine show severe vasoconstriction, CBFv decreases and vascular adaptations with extended diving arteriolar/venular vessels that prioritize blood supply to deeper cortical capillaries. The 3D imaging platform we present provides a powerful tool to study dynamic changes in vessel diameters and morphology alongside CBFv networks in the brain of awake animals that can advance our understanding of the effects of drugs and disease conditions (ischemia, tumors, wound healing).
Collapse
Affiliation(s)
- Yingtian Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA.
| | - Kicheon Park
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Jiaxiang Ren
- Department of Computer Science, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20857, USA
| | - Haibin Ling
- Department of Computer Science, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Alan P Koretsky
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| |
Collapse
|
19
|
Camargo CP, Muhuri AK, Alapan Y, Sestito LF, Khosla M, Manspeaker MP, Smith AS, Paulos CM, Thomas SN. A dhesion analysis via a tumor vasculature-like microfluidic device identifies CD8 + T cells with enhanced tumor homing to improve cell therapy. Cell Rep 2023; 42:112175. [PMID: 36848287 DOI: 10.1016/j.celrep.2023.112175] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 12/14/2022] [Accepted: 02/13/2023] [Indexed: 02/27/2023] Open
Abstract
CD8+ T cell recruitment to the tumor microenvironment is critical for the success of adoptive cell therapy (ACT). Unfortunately, only a small fraction of transferred cells home to solid tumors. Adhesive ligand-receptor interactions have been implicated in CD8+ T cell homing; however, there is a lack of understanding of how CD8+ T cells interact with tumor vasculature-expressed adhesive ligands under the influence of hemodynamic flow. Here, the capacity of CD8+ T cells to home to melanomas is modeled ex vivo using an engineered microfluidic device that recapitulates the hemodynamic microenvironment of the tumor vasculature. Adoptively transferred CD8+ T cells with enhanced adhesion in flow in vitro and tumor homing in vivo improve tumor control by ACT in combination with immune checkpoint blockade. These results show that engineered microfluidic devices can model the microenvironment of the tumor vasculature to identify subsets of T cells with enhanced tumor infiltrating capabilities, a key limitation in ACT.
Collapse
Affiliation(s)
- Camila P Camargo
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Abir K Muhuri
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yunus Alapan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Lauren F Sestito
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Megha Khosla
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Margaret P Manspeaker
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Aubrey S Smith
- Winship Cancer Institute, Emory University, Atlanta, GA 30332, USA; Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - Susan N Thomas
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30332, USA.
| |
Collapse
|
20
|
McCall JR, Santibanez F, Belgharbi H, Pinton GF, Dayton PA. Non-invasive transcranial volumetric ultrasound localization microscopy of the rat brain with continuous, high volume-rate acquisition. Theranostics 2023; 13:1235-1246. [PMID: 36923540 PMCID: PMC10008741 DOI: 10.7150/thno.79189] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/24/2022] [Indexed: 02/15/2023] Open
Abstract
Rationale: Structure and function of the microvasculature provides critical information about disease state, can be used to identify local regions of pathology, and has been shown to be an indicator of response to therapy. Improved methods of assessing the microvasculature with non-invasive imaging modalities such as ultrasound will have an impact in biomedical theranostics. Ultrasound localization microscopy (ULM) is a new technology which allows processing of ultrasound data for visualization of microvasculature at a resolution better than allowed by acoustic diffraction with traditional ultrasound systems. Previous application of this modality in brain imaging has required the use of invasive procedures, such as a craniotomy, skull-thinning, or scalp removal, all of which are not feasible for the purpose of longitudinal studies. Methods: The impact of ultrasound localization microscopy is expanded using a 1024 channel matrix array ultrasonic transducer, four synchronized programmable ultrasound systems with customized high-performance hardware and software, and high-performance GPUs for processing. The potential of the imaging hardware and processing approaches are demonstrated in-vivo. Results: Our unique implementation allows asynchronous acquisition and data transfer for uninterrupted data collection at an ultra-high fixed frame rate. Using these methods, the vasculature was imaged using 100,000 volumes continuously at a volume acquisition rate of 500 volumes per second. With ULM, we achieved a resolution of 31 µm, which is a resolution improvement on conventional ultrasound imaging by nearly a factor of ten, in 3-D. This was accomplished while imaging through the intact skull with no scalp removal, which demonstrates the utility of this method for longitudinal studies. Conclusions: The results demonstrate new capabilities to rapidly image and analyze complex vascular networks in 3-D volume space for structural and functional imaging in disease assessment, targeted therapeutic delivery, monitoring response to therapy, and other theranostic applications.
Collapse
Affiliation(s)
- Jacob R. McCall
- The Joint Department of Biomedical Engineering, UNC Chapel Hill and NC State University
- Electrical and Computer Engineering, NC State University
| | - Francisco Santibanez
- The Joint Department of Biomedical Engineering, UNC Chapel Hill and NC State University
| | - Hatim Belgharbi
- The Joint Department of Biomedical Engineering, UNC Chapel Hill and NC State University
| | - Gianmarco F. Pinton
- The Joint Department of Biomedical Engineering, UNC Chapel Hill and NC State University
| | - Paul A. Dayton
- The Joint Department of Biomedical Engineering, UNC Chapel Hill and NC State University
| |
Collapse
|
21
|
Niizawa T, Sakuraba R, Kusaka T, Kurihara Y, Sugashi T, Kawaguchi H, Kanno I, Masamoto K. Spatiotemporal analysis of blood plasma and blood cell flow fluctuations of cerebral microcirculation in anesthetized rats. J Cereb Blood Flow Metab 2023; 43:138-152. [PMID: 36138557 PMCID: PMC9875347 DOI: 10.1177/0271678x221125743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 08/09/2022] [Accepted: 08/13/2022] [Indexed: 01/28/2023]
Abstract
Cerebral hemodynamics fluctuates spontaneously over broad frequency ranges. However, its spatiotemporal coherence of flow oscillations in cerebral microcirculation remains incompletely understood. The objective of this study was to characterize the spatiotemporal fluctuations of red blood cells (RBCs) and plasma flow in the rat cerebral microcirculation by simultaneously imaging their dynamic behaviors. Comparisons of changes in cross-section diameters between RBC and plasma flow showed dissociations in penetrating arterioles. The results indicate that vasomotion has the least effect on the lateral movement of circulating RBCs, resulting in variable changes in plasma layer thickness. Parenchymal capillaries exhibited slow fluctuations in RBC velocity (0.1 to 0.3 Hz), regardless of capillary diameter fluctuations (<0.1 Hz). Temporal fluctuations and the velocity of RBCs decreased significantly at divergent capillary bifurcations. The results indicate that a transit of RBCs generates flow resistance in the capillaries and that slow velocity fluctuations of the RBCs are subject to a number of bifurcations. In conclusion, the high-frequency oscillation of the blood flow is filtered at the bifurcation through the capillary networks. Therefore, a number of bifurcations in the cerebral microcirculation may contribute to the power of low-frequency oscillations.
Collapse
Affiliation(s)
- Tomoya Niizawa
- Faculty of Informatics and Engineering, University of
Electro-Communications, Tokyo, Japan
| | - Ruka Sakuraba
- Faculty of Informatics and Engineering, University of
Electro-Communications, Tokyo, Japan
| | - Tomoya Kusaka
- Faculty of Informatics and Engineering, University of
Electro-Communications, Tokyo, Japan
| | - Yuika Kurihara
- Faculty of Informatics and Engineering, University of
Electro-Communications, Tokyo, Japan
| | - Takuma Sugashi
- Faculty of Informatics and Engineering, University of
Electro-Communications, Tokyo, Japan
- Center for Neuroscience and Biomedical Engineering,
University of Electro-Communications, Tokyo, Japan
| | - Hiroshi Kawaguchi
- Human Informatics and Interaction Research Institute,
National Institute of Advanced Industrial Science and Technology
(AIST), Ibaraki, Japan
| | - Iwao Kanno
- Department of Functional Brain Imaging Research,
National Institute of Radiological Sciences, Chiba, Japan
| | - Kazuto Masamoto
- Faculty of Informatics and Engineering, University of
Electro-Communications, Tokyo, Japan
- Center for Neuroscience and Biomedical Engineering,
University of Electro-Communications, Tokyo, Japan
- Department of Functional Brain Imaging Research,
National Institute of Radiological Sciences, Chiba, Japan
| |
Collapse
|
22
|
Cao H, Gao S, Jogani R, Sugimura R. The Tumor Microenvironment Reprograms Immune Cells. Cell Reprogram 2022; 24:343-352. [PMID: 36301256 DOI: 10.1089/cell.2022.0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Tumor tissue comprises a highly complex network of diverse cell types. The tumor microenvironment (TME) can be mainly subdivided into cancer cells and stromal cell compartments, the latter include different types of immune cells, fibroblasts, endothelial cells, and pericytes. Tumor cells reprogram immune cells and other stromal cells in the TME to constrain their antitumor capacity by creating an immunosuppressive milieu and metabolism competition. Moreover, the reprogramming effect on immune cells is localized not only in the tumor but also at the systemic level. With wide application of single-cell sequencing technology, tumor-specific characteristics of immune cells and other stromal cells in the TME have been dissected. In this review, we mainly focus on how tumor cells reprogram immune cells both within the TME and peripheral blood. This information can further help us to improve the efficiency of current immunotherapy as well as bring up new ideas to combat cancer.
Collapse
Affiliation(s)
- Handi Cao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,Centre for Translational Stem Cell Biology, Science Park, Hong Kong
| | - Sanxing Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Ritika Jogani
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,Centre for Translational Stem Cell Biology, Science Park, Hong Kong
| |
Collapse
|
23
|
Salavati H, Debbaut C, Pullens P, Ceelen W. Interstitial fluid pressure as an emerging biomarker in solid tumors. Biochim Biophys Acta Rev Cancer 2022; 1877:188792. [PMID: 36084861 DOI: 10.1016/j.bbcan.2022.188792] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/12/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022]
Abstract
The physical microenvironment of cancer is characterized by elevated stiffness and tissue pressure, the main component of which is the interstitial fluid pressure (IFP). Elevated IFP is an established negative predictive and prognostic parameter, directly affecting malignant behavior and therapy response. As such, measurement of the IFP would allow to develop strategies aimed at engineering the physical microenvironment of cancer. Traditionally, IFP measurement required the use of invasive methods. Recent progress in dynamic and functional imaging methods such as dynamic contrast enhanced (DCE) magnetic resonance imaging and elastography, combined with numerical models and simulation, allows to comprehensively assess the biomechanical landscape of cancer, and may help to overcome physical barriers to drug delivery and immune cell infiltration. Here, we provide a comprehensive overview of the origin of elevated IFP, and its role in the malignant phenotype. Also, we review the methods used to measure IFP using invasive and imaging based methods, and highlight remaining obstacles and potential areas of progress in order to implement IFP measurement in clinical practice.
Collapse
Affiliation(s)
- Hooman Salavati
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; IBitech- Biommeda, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Charlotte Debbaut
- IBitech- Biommeda, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Pim Pullens
- Department of Radiology, Ghent University Hospital, Ghent, Belgium; Ghent Institute of Functional and Metabolic Imaging (GIFMI), Ghent University, Ghent, Belgium; IBitech- Medisip, Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
24
|
Zhang T, Jia Y, Yu Y, Zhang B, Xu F, Guo H. Targeting the tumor biophysical microenvironment to reduce resistance to immunotherapy. Adv Drug Deliv Rev 2022; 186:114319. [PMID: 35545136 DOI: 10.1016/j.addr.2022.114319] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023]
Abstract
Immunotherapy based on immune checkpoint inhibitors has evolved into a new pillar of cancer treatment in clinics, but dealing with treatment resistance (either primary or acquired) is a major challenge. The tumor microenvironment (TME) has a substantial impact on the pathological behaviors and treatment response of many cancers. The biophysical clues in TME have recently been considered as important characteristics of cancer. Furthermore, there is mounting evidence that biophysical cues in TME play important roles in each step of the cascade of cancer immunotherapy that synergistically contribute to immunotherapy resistance. In this review, we summarize five main biophysical cues in TME that affect resistance to immunotherapy: extracellular matrix (ECM) structure, ECM stiffness, tumor interstitial fluid pressure (IFP), solid stress, and vascular shear stress. First, the biophysical factors involved in anti-tumor immunity and therapeutic antibody delivery processes are reviewed. Then, the causes of these five biophysical cues and how they contribute to immunotherapy resistance are discussed. Finally, the latest treatment strategies that aim to improve immunotherapy efficacy by targeting these biophysical cues are shared. This review highlights the biophysical cues that lead to immunotherapy resistance, also supplements their importance in related technologies for studying TME biophysical cues in vitro and therapeutic strategies targeting biophysical cues to improve the effects of immunotherapy.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuanbo Jia
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yang Yu
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710049, PR China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Hui Guo
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
25
|
Chaigneau E, Charpak S. Measurement of Blood Velocity With Laser Scanning Microscopy: Modeling and Comparison of Line-Scan Image-Processing Algorithms. Front Physiol 2022; 13:848002. [PMID: 35464098 PMCID: PMC9022085 DOI: 10.3389/fphys.2022.848002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
Laser scanning microscopy is widely used to measure blood hemodynamics with line-scans in physiological and pathological vessels. With scans of broken lines, i.e., lines made of several segments with different orientations, it also allows simultaneous monitoring of vessel diameter dynamics or the activity of specific cells. Analysis of red blood cell (RBC) velocity from line-scans requires specific image-processing algorithms, as angle measurements, Line-Scanning Particle Image Velocimetry (LSPIV) or Fourier transformation of line-scan images. The conditions under which these image-processing algorithms give accurate measurements have not been fully characterized although the accuracy of measurements vary according to specific experimental parameters: the vessel type, the RBC velocity, the scanning parameters, and the image signal to noise ratio. Here, we developed mathematical models for the three previously mentioned line-scan image-processing algorithms. Our models predict the experimental conditions in which RBC velocity measurements are accurate. We illustrate the case of different vessel types and give the parameter space available for each of them. Last, we developed a software generating artificial line-scan images and used it to validate our models.
Collapse
Affiliation(s)
- Emmanuelle Chaigneau
- Institut de la Vision, INSERM U968, Paris, France
- Institut de la Vision, CNRS UMR 7210, Paris, France
- Institut de la Vision, Sorbonne Université, Paris, France
- *Correspondence: Emmanuelle Chaigneau,
| | - Serge Charpak
- Institut de la Vision, INSERM U968, Paris, France
- Institut de la Vision, CNRS UMR 7210, Paris, France
- Institut de la Vision, Sorbonne Université, Paris, France
- Serge Charpak,
| |
Collapse
|
26
|
Park M, Le TA, Yoon J. Offline Programming Guidance for Swarm Steering of Micro-/Nano Magnetic Particles in a Dynamic Multichannel Vascular Model. IEEE Robot Autom Lett 2022. [DOI: 10.1109/lra.2022.3148789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
27
|
Vaghi C, Fanciullino R, Benzekry S, Poignard C. Macro-scale models for fluid flow in tumour tissues: impact of microstructure properties. J Math Biol 2022; 84:27. [DOI: 10.1007/s00285-022-01719-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/04/2022] [Accepted: 01/20/2022] [Indexed: 11/28/2022]
|
28
|
Barak T, Ristori E, Ercan-Sencicek AG, Miyagishima DF, Nelson-Williams C, Dong W, Jin SC, Prendergast A, Armero W, Henegariu O, Erson-Omay EZ, Harmancı AS, Guy M, Gültekin B, Kilic D, Rai DK, Goc N, Aguilera SM, Gülez B, Altinok S, Ozcan K, Yarman Y, Coskun S, Sempou E, Deniz E, Hintzen J, Cox A, Fomchenko E, Jung SW, Ozturk AK, Louvi A, Bilgüvar K, Connolly ES, Khokha MK, Kahle KT, Yasuno K, Lifton RP, Mishra-Gorur K, Nicoli S, Günel M. PPIL4 is essential for brain angiogenesis and implicated in intracranial aneurysms in humans. Nat Med 2021; 27:2165-2175. [PMID: 34887573 PMCID: PMC8768030 DOI: 10.1038/s41591-021-01572-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 10/05/2021] [Indexed: 12/16/2022]
Abstract
Intracranial aneurysm (IA) rupture leads to subarachnoid hemorrhage, a sudden-onset disease that often causes death or severe disability. Although genome-wide association studies have identified common genetic variants that increase IA risk moderately, the contribution of variants with large effect remains poorly defined. Using whole-exome sequencing, we identified significant enrichment of rare, deleterious mutations in PPIL4, encoding peptidyl-prolyl cis-trans isomerase-like 4, in both familial and index IA cases. Ppil4 depletion in vertebrate models causes intracerebral hemorrhage, defects in cerebrovascular morphology and impaired Wnt signaling. Wild-type, but not IA-mutant, PPIL4 potentiates Wnt signaling by binding JMJD6, a known angiogenesis regulator and Wnt activator. These findings identify a novel PPIL4-dependent Wnt signaling mechanism involved in brain-specific angiogenesis and maintenance of cerebrovascular integrity and implicate PPIL4 gene mutations in the pathogenesis of IA.
Collapse
Affiliation(s)
- Tanyeri Barak
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | - Emma Ristori
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale School of Medicine, New Haven, CT, USA
| | - A Gulhan Ercan-Sencicek
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | - Danielle F Miyagishima
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Sheng Chih Jin
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew Prendergast
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale School of Medicine, New Haven, CT, USA
| | - William Armero
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale School of Medicine, New Haven, CT, USA
| | - Octavian Henegariu
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | - E Zeynep Erson-Omay
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | - Akdes Serin Harmancı
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | - Mikhael Guy
- Yale Center for Research Computing, Yale University, New Haven, CT, USA
| | - Batur Gültekin
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Deniz Kilic
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Devendra K Rai
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | - Nükte Goc
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Burcu Gülez
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Selin Altinok
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Kent Ozcan
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Yanki Yarman
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Süleyman Coskun
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | - Emily Sempou
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Engin Deniz
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Jared Hintzen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale School of Medicine, New Haven, CT, USA
| | - Andrew Cox
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Elena Fomchenko
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Su Woong Jung
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Ali Kemal Ozturk
- Department of Neurosurgery, Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Angeliki Louvi
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | - Kaya Bilgüvar
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | - E Sander Connolly
- Department of Neurosurgery, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Mustafa K Khokha
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Katsuhito Yasuno
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA
| | - Richard P Lifton
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Ketu Mishra-Gorur
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA.
| | - Stefania Nicoli
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale School of Medicine, New Haven, CT, USA.
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA.
| | - Murat Günel
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
29
|
Arpino JM, Yin H, Prescott EK, Staples SCR, Nong Z, Li F, Chevalier J, Balint B, O’Neil C, Mortuza R, Milkovich S, Lee JJ, Lorusso D, Sandig M, Hamilton DW, Holdsworth DW, Poepping TL, Ellis CG, Pickering JG. Low-flow intussusception and metastable VEGFR2 signaling launch angiogenesis in ischemic muscle. SCIENCE ADVANCES 2021; 7:eabg9509. [PMID: 34826235 PMCID: PMC8626079 DOI: 10.1126/sciadv.abg9509] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Efforts to promote sprouting angiogenesis in skeletal muscles of individuals with peripheral artery disease have not been clinically successful. We discovered that, contrary to the prevailing view, angiogenesis following ischemic muscle injury in mice was not driven by endothelial sprouting. Instead, real-time imaging revealed the emergence of wide-caliber, primordial conduits with ultralow flow that rapidly transformed into a hierarchical neocirculation by transluminal bridging and intussusception. This process was accelerated by inhibiting vascular endothelial growth factor receptor-2 (VEGFR2). We probed this response by developing the first live-cell model of transluminal endothelial bridging using microfluidics. Endothelial cells subjected to ultralow shear stress could reposition inside the flowing lumen as pillars. Moreover, the low-flow lumen proved to be a privileged location for endothelial cells with reduced VEGFR2 signaling capacity, as VEGFR2 mechanosignals were boosted. These findings redefine regenerative angiogenesis in muscle as an intussusceptive process and uncover a basis for its launch.
Collapse
Affiliation(s)
- John-Michael Arpino
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Hao Yin
- Robarts Research Institute, Western University, London, Canada
| | - Emma K. Prescott
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Sabrina C. R. Staples
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Zengxuan Nong
- Robarts Research Institute, Western University, London, Canada
| | - Fuyan Li
- Robarts Research Institute, Western University, London, Canada
| | - Jacqueline Chevalier
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Brittany Balint
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Caroline O’Neil
- Robarts Research Institute, Western University, London, Canada
| | | | - Stephanie Milkovich
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Jason J. Lee
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medicine, Western University, London, Canada
| | - Daniel Lorusso
- Robarts Research Institute, Western University, London, Canada
| | - Martin Sandig
- Department of Anatomy and Cell Biology, Western University, London, Canada
| | | | - David W. Holdsworth
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Tamie L. Poepping
- Department of Physics and Astronomy, Western University, London, Canada
| | - Christopher G. Ellis
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medicine, Western University, London, Canada
| | - J. Geoffrey Pickering
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medicine, Western University, London, Canada
- Department of Biochemistry, Western University, London, Canada
- Corresponding author.
| |
Collapse
|
30
|
Anzabi M, Li B, Wang H, Kura S, Sakadžić S, Boas D, Østergaard L, Ayata C. Optical coherence tomography of arteriolar diameter and capillary perfusion during spreading depolarizations. J Cereb Blood Flow Metab 2021; 41:2256-2263. [PMID: 33593116 PMCID: PMC8393288 DOI: 10.1177/0271678x21994013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/21/2020] [Accepted: 01/08/2021] [Indexed: 11/17/2022]
Abstract
Spreading depolarization (SD) is associated with profound oligemia and reduced oxygen availability in the mouse cortex during the depolarization phase. Coincident pial arteriolar constriction has been implicated as the primary mechanism for the oligemia. However, where in the vascular bed the hemodynamic response starts has been unclear. To resolve the origin of the hemodynamic response, we used optical coherence tomography (OCT) to simultaneously monitor changes in the vascular tree from capillary bed to pial arteries in mice during two consecutive SDs 15 minutes apart. We found that capillary flow dropped several seconds before pial arteriolar constriction. Moreover, penetrating arterioles constricted before pial arteries suggesting upstream propagation of constriction. Smaller caliber distal pial arteries constricted stronger than larger caliber proximal arterioles, suggesting that the farther the constriction propagates, the weaker it gets. Altogether, our data indicate that the hemodynamic response to cortical SD originates in the capillary bed.
Collapse
Affiliation(s)
- Maryam Anzabi
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
- Center of Functionally Integrative Neuroscience (CFIN) and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Baoqiang Li
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences
| | - Hui Wang
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Sreekanth Kura
- Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Sava Sakadžić
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - David Boas
- Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience (CFIN) and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, USA
| |
Collapse
|
31
|
Akbari E, Spychalski GB, Menyhert MM, Rangharajan KK, Tinapple JW, Prakash S, Song JW. Endothelial barrier function is co-regulated at vessel bifurcations by fluid forces and sphingosine-1-phosphate. BIOMATERIALS AND BIOSYSTEMS 2021; 3:100020. [PMID: 35317095 PMCID: PMC8936769 DOI: 10.1016/j.bbiosy.2021.100020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/12/2021] [Accepted: 05/29/2021] [Indexed: 12/31/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid mediator of endothelial barrier function. Prior studies have implicated mechanical stimulation due to intravascular laminar shear stress in co-regulating S1P signaling in endothelial cells (ECs). Yet, vascular networks in vivo consist of vessel bifurcations, and this geometry generates hemodynamic forces at the bifurcation point distinct from laminar shear stress. However, the role of these forces at vessel bifurcations in regulating S1P-dependent endothelial barrier function is not known. In this study, we implemented a microfluidic platform that recapitulates the flow dynamics of vessel bifurcations with in situ quantification of the permeability of microvessel analogues. Co-application of S1P with impinging bifurcated fluid flow, which is characterized by approximately zero shear stress and 38 dyn•cm-2 stagnation pressure at the vessel bifurcation point, promotes vessel stabilization. Similarly, co-treatment of S1P with 3 dyn•cm-2 laminar shear stress is also protective of endothelial barrier function. Moreover, it is shown that vessel stabilization due to bifurcated fluid flow and laminar shear stress is dependent on S1P receptor 1 or 2 signaling. Collectively, these findings demonstrate the endothelium-protective function of fluid forces at vessel bifurcations and their involvement in coordinating S1P-dependent regulation of vessel permeability.
Collapse
Affiliation(s)
- Ehsan Akbari
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Griffin B. Spychalski
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Miles M. Menyhert
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Kaushik K. Rangharajan
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Joseph W. Tinapple
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Shaurya Prakash
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States, 43210
| | - Jonathan W. Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States, 43210
| |
Collapse
|
32
|
Compressed vessels bias red blood cell partitioning at bifurcations in a hematocrit-dependent manner: Implications in tumor blood flow. Proc Natl Acad Sci U S A 2021; 118:2025236118. [PMID: 34140409 DOI: 10.1073/pnas.2025236118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The tumor microenvironment is abnormal and associated with tumor tissue hypoxia, immunosuppression, and poor response to treatment. One important abnormality present in tumors is vessel compression. Vessel decompression has been shown to increase survival rates in animal models via enhanced and more homogeneous oxygenation. However, our knowledge of the biophysical mechanisms linking tumor decompression to improved tumor oxygenation is limited. In this study, we propose a computational model to investigate the impact of vessel compression on red blood cell (RBC) dynamics in tumor vascular networks. Our results demonstrate that vessel compression can alter RBC partitioning at bifurcations in a hematocrit-dependent and flow rate-independent manner. We identify RBC focusing due to cross-streamline migration as the mechanism responsible and characterize the spatiotemporal recovery dynamics controlling downstream partitioning. Based on this knowledge, we formulate a reduced-order model that will help future research to elucidate how these effects propagate at a whole vascular network level. These findings contribute to the mechanistic understanding of hemodilution in tumor vascular networks and oxygen homogenization following pharmacological solid tumor decompression.
Collapse
|
33
|
Momin A, Bahrampour S, Min HK, Chen X, Wang X, Sun Y, Huang X. Channeling Force in the Brain: Mechanosensitive Ion Channels Choreograph Mechanics and Malignancies. Trends Pharmacol Sci 2021; 42:367-384. [PMID: 33752907 DOI: 10.1016/j.tips.2021.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/15/2021] [Accepted: 02/26/2021] [Indexed: 12/15/2022]
Abstract
Force is everywhere. Through cell-intrinsic activities and interactions with the microenvironment, cells generate, transmit, and sense mechanical forces, such as compression, tension, and shear stress. These forces shape the mechanical properties of cells and tissues. Akin to how balanced biochemical signaling safeguards physiological processes, a mechanical optimum is required for homeostasis. The brain constructs a mechanical optimum from its cellular and extracellular constituents. However, in brain cancer, the mechanical properties are disrupted: tumor and nontumoral cells experience dysregulated solid and fluid stress, while tumor tissue develops altered stiffness. Mechanosensitive (MS) ion channels perceive mechanical cues to govern ion flux and cellular signaling. In this review, we describe the mechanical properties of the brain in healthy and cancer states and illustrate MS ion channels as sensors of mechanical cues to regulate malignant growth. Targeting MS ion channels offers disease insights at the interface of cancer, neuroscience, and mechanobiology to reveal therapeutic opportunities in brain tumors.
Collapse
Affiliation(s)
- Ali Momin
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ONT, M5S 3E1, Canada.
| | - Shahrzad Bahrampour
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Hyun-Kee Min
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ONT, M5S 3E1, Canada
| | - Xin Chen
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada
| | - Xian Wang
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ONT, M5S 3G8, Canada
| | - Xi Huang
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ONT, M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ONT, M5S 3E1, Canada.
| |
Collapse
|
34
|
Tang J, Kilic K, Szabo TL, Boas DA. Improved Color Doppler for Cerebral Blood Flow Axial Velocity Imaging. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:758-764. [PMID: 33156785 PMCID: PMC8098776 DOI: 10.1109/tmi.2020.3036468] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Conventional color Doppler ultrasound imaging suffers from mutual frequency cancellation when applied to quantify axial blood flow velocities in the rodent brain where inverse flows exist within an ultrasound measurement voxel. Here, we report an improved color Doppler-based functional ultrasound imaging method (iCD-fUS) for axial blood flow velocity imaging of the rodent brain. By applying a directional filter and high frequency noise thresholding, iCD-fUS is able to accurately quantify blood flow velocities within the brain as validated with the ultrasound localization microscopy velocimetry method. We show that iCD-fUS is able to image and resolve the directional axial blood flow velocity throughout the entire coronal section of the brain at a temporal frame rate of up to 10 Hz with a spatial resolution of ~100 [Formula: see text]. We further applied iCD-fUS to image the axial blood flow velocity change in response to whisker stimulation in an awake mouse, showing its potential for studying brain activation. With these capabilities, iCD-fUS provides a powerful, quantitative tool for in vivo chronic research.
Collapse
|
35
|
Imaging and optogenetic modulation of vascular mural cells in the live brain. Nat Protoc 2020; 16:472-496. [PMID: 33299155 DOI: 10.1038/s41596-020-00425-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/24/2020] [Indexed: 12/17/2022]
Abstract
Mural cells (smooth muscle cells and pericytes) are integral components of brain blood vessels that play important roles in vascular formation, blood-brain barrier maintenance, and regulation of regional cerebral blood flow (rCBF). These cells are implicated in conditions ranging from developmental vascular disorders to age-related neurodegenerative diseases. Here we present complementary tools for cell labeling with transgenic mice and organic dyes that allow high-resolution intravital imaging of the different mural cell subtypes. We also provide detailed methodologies for imaging of spontaneous and neural activity-evoked calcium transients in mural cells. In addition, we describe strategies for single- and two-photon optogenetics that allow manipulation of the activity of individual and small clusters of mural cells. Together with measurements of diameter and flow in individual brain microvessels, calcium imaging and optogenetics allow the investigation of pericyte and smooth muscle cell physiology and their role in regulating rCBF. We also demonstrate the utility of these tools to investigate mural cells in the context of Alzheimer's disease and cerebral ischemia mouse models. Thus, these methods can be used to reveal the functional and structural heterogeneity of mural cells in vivo, and allow detailed cellular studies of the normal function and pathophysiology of mural cells in a variety of disease models. The implementation of this protocol can take from several hours to days depending on the intended applications.
Collapse
|
36
|
Abnormal morphology biases hematocrit distribution in tumor vasculature and contributes to heterogeneity in tissue oxygenation. Proc Natl Acad Sci U S A 2020; 117:27811-27819. [PMID: 33109723 PMCID: PMC7668105 DOI: 10.1073/pnas.2007770117] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Oxygen heterogeneity in solid tumors is recognized as a limiting factor for therapeutic efficacy. This heterogeneity arises from the abnormal tumor vascular structure. We investigate the role that anomalies in red blood cell transport plays in establishing oxygen heterogeneity in tumor tissue. We introduce a metric to characterize tumor vasculature (mean vessel length-to-diameter ratio, λ) and demonstrate how it predicts tissue-oxygen heterogeneity. We also report an increase in λ following treatment with the antiangiogenic agent DC101. Together, we propose λ as an effective way of monitoring the action of antiangiogenic agents and a proxy measure of oxygen heterogeneity in tumor tissue. Unraveling the causal relationship between tumor vascular structure and tissue oxygenation will pave the way for new personalized therapeutic approaches. Oxygen heterogeneity in solid tumors is recognized as a limiting factor for therapeutic efficacy. This heterogeneity arises from the abnormal vascular structure of the tumor, but the precise mechanisms linking abnormal structure and compromised oxygen transport are only partially understood. In this paper, we investigate the role that red blood cell (RBC) transport plays in establishing oxygen heterogeneity in tumor tissue. We focus on heterogeneity driven by network effects, which are challenging to observe experimentally due to the reduced fields of view typically considered. Motivated by our findings of abnormal vascular patterns linked to deviations from current RBC transport theory, we calculated average vessel lengths L¯ and diameters d¯ from tumor allografts of three cancer cell lines and observed a substantial reduction in the ratio λ=L¯/d¯ compared to physiological conditions. Mathematical modeling reveals that small values of the ratio λ (i.e., λ<6) can bias hematocrit distribution in tumor vascular networks and drive heterogeneous oxygenation of tumor tissue. Finally, we show an increase in the value of λ in tumor vascular networks following treatment with the antiangiogenic cancer agent DC101. Based on our findings, we propose λ as an effective way of monitoring the efficacy of antiangiogenic agents and as a proxy measure of perfusion and oxygenation in tumor tissue undergoing antiangiogenic treatment.
Collapse
|
37
|
Dogra P, Butner JD, Ramirez JR, Cristini V, Wang Z. Investigating the Effect of Aging on the Pharmacokinetics and Tumor Delivery of Nanomaterials using Mathematical Modeling. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:2447-2450. [PMID: 33018501 DOI: 10.1109/embc44109.2020.9175322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The application of nanomedicine for diagnosis and treatment of cancer has immense potential, but has witnessed only limited clinical success, in part due to insufficient understanding of the role of nanomaterial properties and physiological variables in governing nanoparticle (NP) pharmacology. Here, we present a multiscale mathematical model to examine the effects of physiological changes associated with patient age on the pharmacokinetics and tumor delivery efficiency of NPs. We show that physiological changes due to aging prolong the residence of NPs in the systemic circulation, thereby improving passive accumulation of NPs in tumors.Clinical Relevance - Understanding the effect of inter-individual variability on the pharmacological behavior of nanomaterials will improve their clinical translatability.
Collapse
|
38
|
Nia HT, Datta M, Seano G, Zhang S, Ho WW, Roberge S, Huang P, Munn LL, Jain RK. In vivo compression and imaging in mouse brain to measure the effects of solid stress. Nat Protoc 2020; 15:2321-2340. [PMID: 32681151 PMCID: PMC11412114 DOI: 10.1038/s41596-020-0328-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 04/06/2020] [Indexed: 02/07/2023]
Abstract
We recently developed an in vivo compression device that simulates the solid mechanical forces exerted by a growing tumor on the surrounding brain tissue and delineates the physical versus biological effects of a tumor. This device, to our knowledge the first of its kind, can recapitulate the compressive forces on the cerebellar cortex from primary (e.g., glioblastoma) and metastatic (e.g., breast cancer) tumors, as well as on the cerebellum from tumors such as medulloblastoma and ependymoma. We adapted standard transparent cranial windows normally used for intravital imaging studies in mice to include a turnable screw for controlled compression (acute or chronic) and decompression of the cerebral cortex. The device enables longitudinal imaging of the compressed brain tissue over several weeks or months as the screw is progressively extended against the brain tissue to recapitulate tumor growth-induced solid stress. The cranial window can be simply installed on the mouse skull according to previously established methods, and the screw mechanism can be readily manufactured in-house. The total time for construction and implantation of the in vivo compressive cranial window is <1 h (per mouse). This technique can also be used to study a variety of other diseases or disorders that present with abnormal solid masses in the brain, including cysts and benign growths.
Collapse
Affiliation(s)
- Hadi T Nia
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Meenal Datta
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Giorgio Seano
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Tumor Microenvironment Laboratory, Institut Curie Research Center, Paris-Saclay University, PSL Research University, Inserm U1021, CNRS UMR3347, Orsay, France
| | - Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - William W Ho
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sylvie Roberge
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peigen Huang
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Liu C, Liang Y, Wang L. Single-shot photoacoustic microscopy of hemoglobin concentration, oxygen saturation, and blood flow in sub-microseconds. PHOTOACOUSTICS 2020; 17:100156. [PMID: 31956486 PMCID: PMC6957791 DOI: 10.1016/j.pacs.2019.100156] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/28/2019] [Accepted: 12/05/2019] [Indexed: 05/04/2023]
Abstract
We present fast functional optical-resolution photoacoustic microscopy (OR-PAM) that can simultaneously image hemoglobin concentration, blood flow speed, and oxygen saturation with three-pulse excitation. To instantaneously determine the blood flow speed, dual-pulse photoacoustic flowmetry is developed to determine the blood flow speed from photoacoustic signal decay in sub-microseconds. Grueneisen relaxation effect is compensated for in the oxygen saturation calculation. The blood flow imaging is validated in phantom and in vivo experiments. The results show that the flow speed can be measured accurately in sub-microseconds by comparing the dual-pulse flowmetric method with photoacoustic Doppler flowmetry. Wide-field OR-PAM of hemoglobin concentration, blood flow speed, and oxygen saturation are demonstrated in the mouse ear. This technical advance enables more biomedical applications for fast functional OR-PAM.
Collapse
Affiliation(s)
- Chao Liu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave., Kowloon, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Yuexing Yi Dao, Nanshan District, Shenzhen, Guang Dong, 518057, China
| | - Yizhi Liang
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou, 510632, China
- Corresponding author.
| | - Lidai Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave., Kowloon, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Yuexing Yi Dao, Nanshan District, Shenzhen, Guang Dong, 518057, China
- Corresponding author at: Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave., Kowloon, Hong Kong, China.
| |
Collapse
|
40
|
Dogra P, Butner JD, Ruiz Ramírez J, Chuang YL, Noureddine A, Jeffrey Brinker C, Cristini V, Wang Z. A mathematical model to predict nanomedicine pharmacokinetics and tumor delivery. Comput Struct Biotechnol J 2020; 18:518-531. [PMID: 32206211 PMCID: PMC7078505 DOI: 10.1016/j.csbj.2020.02.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/14/2020] [Accepted: 02/22/2020] [Indexed: 02/07/2023] Open
Abstract
Towards clinical translation of cancer nanomedicine, it is important to systematically investigate the various parameters related to nanoparticle (NP) physicochemical properties, tumor characteristics, and inter-individual variability that affect the tumor delivery efficiency of therapeutic nanomaterials. Comprehensive investigation of these parameters using traditional experimental approaches is impractical due to the vast parameter space; mathematical models provide a more tractable approach to navigate through such a multidimensional space. To this end, we have developed a predictive mathematical model of whole-body NP pharmacokinetics and their tumor delivery in vivo, and have conducted local and global sensitivity analyses to identify the factors that result in low tumor delivery efficiency and high off-target accumulation of NPs. Our analyses reveal that NP degradation rate, tumor blood viscosity, NP size, tumor vascular fraction, and tumor vascular porosity are the key parameters in governing NP kinetics in the tumor interstitium. The impact of these parameters on tumor delivery efficiency of NPs is discussed, and optimal values for maximizing NP delivery are presented.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Joseph D. Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yao-li Chuang
- Department of Mathematics, California State University, Northridge, CA 91330, USA
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87106, USA
| | - C. Jeffrey Brinker
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87106, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87102, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Corresponding author at: Mathematics in Medicine Program, The Houston Methodist Research Institute, HMRI R8-122, 6670 Bertner Ave, Houston, TX 77030, USA.
| |
Collapse
|
41
|
HemoSYS: A Toolkit for Image-based Systems Biology of Tumor Hemodynamics. Sci Rep 2020; 10:2372. [PMID: 32047171 PMCID: PMC7012876 DOI: 10.1038/s41598-020-58918-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/19/2020] [Indexed: 11/09/2022] Open
Abstract
Abnormal tumor hemodynamics are a critical determinant of a tumor’s microenvironment (TME), and profoundly affect drug delivery, therapeutic efficacy and the emergence of drug and radio-resistance. Since multiple hemodynamic variables can simultaneously exhibit transient and spatiotemporally heterogeneous behavior, there is an exigent need for analysis tools that employ multiple variables to characterize the anomalous hemodynamics within the TME. To address this, we developed a new toolkit called HemoSYS for quantifying the hemodynamic landscape within angiogenic microenvironments. It employs multivariable time-series data such as in vivo tumor blood flow (BF), blood volume (BV) and intravascular oxygen saturation (Hbsat) acquired concurrently using a wide-field multicontrast optical imaging system. The HemoSYS toolkit consists of propagation, clustering, coupling, perturbation and Fourier analysis modules. We demonstrate the utility of each module for characterizing the in vivo hemodynamic landscape of an orthotropic breast cancer model. With HemoSYS, we successfully described: (i) the propagation dynamics of acute hypoxia; (ii) the initiation and dissolution of distinct hemodynamic niches; (iii) tumor blood flow regulation via local vasomotion; (iv) the hemodynamic response to a systemic perturbation with carbogen gas; and (v) frequency domain analysis of hemodynamic heterogeneity in the TME. HemoSYS (freely downloadable via the internet) enables vascular phenotyping from multicontrast in vivo optical imaging data. Its modular design also enables characterization of non-tumor hemodynamics (e.g. brain), other preclinical disease models (e.g. stroke), vascular-targeted therapeutics, and hemodynamic data from other imaging modalities (e.g. MRI).
Collapse
|
42
|
Nashimoto Y, Okada R, Hanada S, Arima Y, Nishiyama K, Miura T, Yokokawa R. Vascularized cancer on a chip: The effect of perfusion on growth and drug delivery of tumor spheroid. Biomaterials 2019; 229:119547. [PMID: 31710953 DOI: 10.1016/j.biomaterials.2019.119547] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 12/16/2022]
Abstract
Tumor vasculature creates a hostile tumor microenvironment (TME) in vivo and nourishes cancers, resulting in cancer progression and drug resistance. To mimic the biochemical and biomechanical environments of tumors in vitro, several models integrated with a vascular network have been reported. However, the tumor responses to biochemical and biomechanical stimuli were evaluated under static conditions and failed to incorporate the effects of blood flow to tumors. In this study, we present a tumor-on-a-chip platform that enables the evaluation of tumor activities with intraluminal flow in an engineered tumor vascular network. The fibroblasts in the tumor spheroid induced angiogenic sprouts, which constructed a perfusable vascular network in a tumor spheroid. The perfusability of the engineered vascular network was preserved during the culture. Moreover, perfusion for over 24 h significantly increased the proliferation activities of tumor cells and decreased cell death in the spheroid. Drug administration under perfusion condition did not show the dose-dependent effects of anticancer drugs on tumor activities in contrast to the results under static conditions. Our results demonstrate the importance of flow in a vascular network for the evaluation of tumor activities in a drug screening platform.
Collapse
Affiliation(s)
- Yuji Nashimoto
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan; Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Miyagi, 980-8578, Japan; Graduate School of Engineering, Tohoku University, Miyagi, 980-8579, Japan
| | - Ryu Okada
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Sanshiro Hanada
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, 860-0811, Japan
| | - Yuichiro Arima
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, 860-0811, Japan
| | - Koichi Nishiyama
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, 860-0811, Japan
| | - Takashi Miura
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan.
| |
Collapse
|
43
|
Chen H, Cai Y, Chen Q, Li Z. Multiscale modeling of solid stress and tumor cell invasion in response to dynamic mechanical microenvironment. Biomech Model Mechanobiol 2019; 19:577-590. [PMID: 31571083 DOI: 10.1007/s10237-019-01231-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/23/2019] [Indexed: 12/16/2022]
Abstract
Mathematical models can provide a quantitatively sophisticated description of tumor cell (TC) behaviors under mechanical microenvironment and help us better understand the role of specific biophysical factors based on their influences on the TC behaviors. To this end, we propose an off-lattice cell-based multiscale mathematical model to describe the dynamic growth-induced solid stress during tumor progression and investigate the influence of the mechanical microenvironment on TC invasion. At the cellular level, cell-cell and cell-matrix interactive forces depend on the mechanical properties of the cells and the cancer-associated fibroblasts in the stroma, respectively. The constitutive relationship between the interactive forces and cell migrations obeys the Hooke's law and damping effects. At the tissue level, the integrated growth-induced forces caused by proliferating cells within the simulation region are balanced by the external forces applied by the surrounding host tissues. Then, the cell movements are calculated according to the Newton's second law of motion, and the morphology of TC invasion is updated. The simulation results reveal the continuous changes of the macroscopic mechanical forces due to the interactions among the structural components and the microscopic environmental factors. Moreover, the simulation results demonstrate the adverse effect of the stiffness of tumor tissue on tumor growth and invasion. A decrease in the stiffness of tumor and matrix can promote TCs to proliferate at a much faster rate and invade into the surrounding healthy tissue more easily, whereas an increase in the stiffness can lead to an aggressive morphology of tumor invasion. We envision that the proposed model can be served as a quantitative theoretical platform to study the underlying biophysical role of the mechanical microenvironmental factors during tumor invasion and metastasis.
Collapse
Affiliation(s)
- H Chen
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.,School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology, Brisbane, QLD, 4001, Australia
| | - Y Cai
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Q Chen
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Z Li
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China. .,School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology, Brisbane, QLD, 4001, Australia.
| |
Collapse
|
44
|
Zanotelli MR, Reinhart-King CA. Mechanical Forces in Tumor Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1092:91-112. [PMID: 30368750 PMCID: PMC6986816 DOI: 10.1007/978-3-319-95294-9_6] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A defining hallmark of cancer and cancer development is upregulated angiogenesis. The vasculature formed in tumors is structurally abnormal, not organized in the conventional hierarchical arrangement, and more permeable than normal vasculature. These features contribute to leaky, tortuous, and dilated blood vessels, which act to create heterogeneous blood flow, compression of vessels, and elevated interstitial fluid pressure. As such, abnormalities in the tumor vasculature not only affect the delivery of nutrients and oxygen to the tumor, but also contribute to creating an abnormal tumor microenvironment that further promotes tumorigenesis. The role of chemical signaling events in mediating tumor angiogenesis has been well researched; however, the relative contribution of physical cues and mechanical regulation of tumor angiogenesis is less understood. Growing research indicates that the physical microenvironment plays a significant role in tumor progression and promoting abnormal tumor vasculature. Here, we review how mechanical cues found in the tumor microenvironment promote aberrant tumor angiogenesis. Specifically, we discuss the influence of matrix stiffness and mechanical stresses in tumor tissue on tumor vasculature, as well as the mechanosensory pathways utilized by endothelial cells to respond to the physical cues found in the tumor microenvironment. We also discuss the impact of the resulting aberrant tumor vasculature on tumor progression and therapeutic treatment.
Collapse
Affiliation(s)
- Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Cynthia A Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
45
|
Mignemi NA, McClatchey PM, Kilchrist KV, Williams IM, Millis BA, Syring KE, Duvall CL, Wasserman DH, McGuinness OP. Rapid changes in the microvascular circulation of skeletal muscle impair insulin delivery during sepsis. Am J Physiol Endocrinol Metab 2019; 316:E1012-E1023. [PMID: 30860883 PMCID: PMC6620574 DOI: 10.1152/ajpendo.00501.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/21/2019] [Accepted: 03/07/2019] [Indexed: 01/07/2023]
Abstract
Sepsis costs the healthcare system $23 billion annually and has a mortality rate between 10 and 40%. An early indication of sepsis is the onset of hyperglycemia, which is the result of sepsis-induced insulin resistance in skeletal muscle. Previous investigations have focused on events in the myocyte (e.g., insulin signaling and glucose transport and subsequent metabolism) as the causes for this insulin-resistant state. However, the delivery of insulin to the skeletal muscle is also an important determinant of insulin action. Skeletal muscle microvascular blood flow, which delivers the insulin to the muscle, is known to be decreased during sepsis. Here we test whether the reduced capillary blood flow to skeletal muscle belies the sepsis-induced insulin resistance by reducing insulin delivery to the myocyte. We hypothesize that decreased capillary flow and consequent decrease in insulin delivery is an early event that precedes gross cardiovascular alterations seen with sepsis. This hypothesis was examined in mice treated with either lipopolysaccharide (LPS) or polymicrobial sepsis followed by intravital microscopy of the skeletal muscle microcirculation. We calculated insulin delivery to the myocyte using two independent methods and found that LPS and sepsis rapidly reduce insulin delivery to the skeletal muscle by ~50%; this was driven by decreases in capillary flow velocity and the number of perfused capillaries. Furthermore, the changes in skeletal muscle microcirculation occur before changes in both cardiac output and arterial blood pressure. These data suggest that a rapid reduction in skeletal muscle insulin delivery contributes to the induction of insulin resistance during sepsis.
Collapse
Affiliation(s)
- Nicholas A Mignemi
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - P Mason McClatchey
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Kameron V Kilchrist
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| | - Ian M Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Bryan A Millis
- Department of Cell and Developmental Biology, Vanderbilt University , Nashville, Tennessee
- Vanderbilt Biophotonics Center, Vanderbilt University , Nashville, Tennessee
| | - Kristen E Syring
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
- Vanderbilt Mouse Metabolic Phenotyping Center , Nashville, Tennessee
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
- Vanderbilt Mouse Metabolic Phenotyping Center , Nashville, Tennessee
| |
Collapse
|
46
|
Li L, Poloyac SM, Watkins SC, St. Croix CM, Alexander H, Gibson GA, Loughran PA, Kirisci L, Clark RSB, Kochanek PM, Vazquez AL, Manole MD. Cerebral microcirculatory alterations and the no-reflow phenomenon in vivo after experimental pediatric cardiac arrest. J Cereb Blood Flow Metab 2019; 39:913-925. [PMID: 29192562 PMCID: PMC6501505 DOI: 10.1177/0271678x17744717] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/29/2017] [Accepted: 10/25/2017] [Indexed: 01/02/2023]
Abstract
Decreased cerebral blood flow (CBF) after cardiac arrest (CA) contributes to secondary ischemic injury in infants and children. We previously reported cortical hypoperfusion with tissue hypoxia early in a pediatric rat model of asphyxial CA. In order to identify specific alterations as potential therapeutic targets to improve cortical hypoperfusion post-CA, we characterize the CBF alterations at the cortical microvascular level in vivo using multiphoton microscopy. We hypothesize that microvascular constriction and disturbances of capillary red blood cell (RBC) flow contribute to cortical hypoperfusion post-CA. After resuscitation from 9 min asphyxial CA, transient dilation of capillaries and venules at 5 min was followed by pial arteriolar constriction at 30 and 60 min (19.6 ± 1.3, 19.3 ± 1.2 µm at 30, 60 min vs. 22.0 ± 1.2 µm at baseline, p < 0.05). At the capillary level, microcirculatory disturbances were highly heterogeneous, with RBC stasis observed in 25.4% of capillaries at 30 min post-CA. Overall, the capillary plasma mean transit time was increased post-CA by 139.7 ± 51.5%, p < 0.05. In conclusion, pial arteriolar constriction, the no-reflow phenomenon and increased plasma transit time were observed post-CA. Our results detail the microvascular disturbances in a pediatric asphyxial CA model and provide a powerful platform for assessing specific vascular-targeted therapies.
Collapse
Affiliation(s)
- Lingjue Li
- Center of Clinical Pharmaceutical
Sciences,
University
of Pittsburgh, PA, USA
- School of Pharmacy,
University
of Pittsburgh, PA, USA
| | - Samuel M Poloyac
- Center of Clinical Pharmaceutical
Sciences,
University
of Pittsburgh, PA, USA
- School of Pharmacy,
University
of Pittsburgh, PA, USA
| | - Simon C Watkins
- Center for Biologic Imaging,
University
of Pittsburgh, PA, USA
| | | | - Henry Alexander
- Safar Center for Resuscitation Research,
University
of Pittsburgh, PA, USA
| | | | | | | | - Robert SB Clark
- Safar Center for Resuscitation Research,
University
of Pittsburgh, PA, USA
- Department of Critical Care Medicine,
University
of Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research,
University
of Pittsburgh, PA, USA
- Department of Critical Care Medicine,
University
of Pittsburgh, PA, USA
- Department of Pediatrics,
University
of Pittsburgh, PA, USA
| | | | - Mioara D Manole
- Safar Center for Resuscitation Research,
University
of Pittsburgh, PA, USA
- Department of Critical Care Medicine,
University
of Pittsburgh, PA, USA
- Department of Pediatrics,
University
of Pittsburgh, PA, USA
| |
Collapse
|
47
|
Tumor Ensemble-Based Modeling and Visualization of Emergent Angiogenic Heterogeneity in Breast Cancer. Sci Rep 2019; 9:5276. [PMID: 30918274 PMCID: PMC6437174 DOI: 10.1038/s41598-019-40888-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/19/2019] [Indexed: 01/29/2023] Open
Abstract
There is a critical need for new tools to investigate the spatio-temporal heterogeneity and phenotypic alterations that arise in the tumor microenvironment. However, computational investigations of emergent inter- and intra-tumor angiogenic heterogeneity necessitate 3D microvascular data from 'whole-tumors' as well as "ensembles" of tumors. Until recently, technical limitations such as 3D imaging capabilities, computational power and cost precluded the incorporation of whole-tumor microvascular data in computational models. Here, we describe a novel computational approach based on multimodality, 3D whole-tumor imaging data acquired from eight orthotopic breast tumor xenografts (i.e. a tumor 'ensemble'). We assessed the heterogeneous angiogenic landscape from the microvascular to tumor ensemble scale in terms of vascular morphology, emergent hemodynamics and intravascular oxygenation. We demonstrate how the abnormal organization and hemodynamics of the tumor microvasculature give rise to unique microvascular niches within the tumor and contribute to inter- and intra-tumor heterogeneity. These tumor ensemble-based simulations together with unique data visualization approaches establish the foundation of a novel 'cancer atlas' for investigators to develop their own in silico systems biology applications. We expect this hybrid image-based modeling framework to be adaptable for the study of other tissues (e.g. brain, heart) and other vasculature-dependent diseases (e.g. stroke, myocardial infarction).
Collapse
|
48
|
Differential Effects of Ang-2/VEGF-A Inhibiting Antibodies in Combination with Radio- or Chemotherapy in Glioma. Cancers (Basel) 2019; 11:cancers11030314. [PMID: 30845704 PMCID: PMC6468722 DOI: 10.3390/cancers11030314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 12/16/2022] Open
Abstract
Antiangiogenic strategies have not shown striking antitumor activities in the majority of glioma patients so far. It is unclear which antiangiogenic combination regimen with standard therapy is most effective. Therefore, we compared anti-VEGF-A, anti-Ang2, and bispecific anti-Ang-2/VEGF-A antibody treatments, alone and in combination with radio- or temozolomide (TMZ) chemotherapy, in a malignant glioma model using multiparameter two-photon in vivo microscopy in mice. We demonstrate that anti-Ang-2/VEGF-A lead to the strongest vascular changes, including vascular normalization, both as monotherapy and when combined with chemotherapy. The latter was accompanied by the most effective chemotherapy-induced death of cancer cells and diminished tumor growth. This was most probably due to a better tumor distribution of the drug, decreased tumor cell motility, and decreased formation of resistance-associated tumor microtubes. Remarkably, all these parameters where reverted when radiotherapy was chosen as combination partner for anti-Ang-2/VEGF-A. In contrast, the best combination partner for radiotherapy was anti-VEGF-A. In conclusion, while TMZ chemotherapy benefits most from combination with anti-Ang-2/VEGF-A, radiotherapy does from anti-VEGF-A. The findings imply that uninformed combination regimens of antiangiogenic and cytotoxic therapies should be avoided.
Collapse
|
49
|
Clendenon SG, Fu X, Von Hoene RA, Clendenon JL, Sluka JP, Winfree S, Mang H, Martinez M, Filson AJ, Klaunig JE, Glazier JA, Dunn KW. A simple automated method for continuous fieldwise measurement of microvascular hemodynamics. Microvasc Res 2018; 123:7-13. [PMID: 30502365 DOI: 10.1016/j.mvr.2018.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 01/25/2023]
Abstract
Microvascular perfusion dynamics are vital to physiological function and are frequently dysregulated in injury and disease. Typically studies measure microvascular flow in a few selected vascular segments over limited time, failing to capture spatial and temporal variability. To quantify microvascular flow in a more complete and unbiased way we developed STAFF (Spatial Temporal Analysis of Fieldwise Flow), a macro for FIJI open-source image analysis software. Using high-speed microvascular flow movies, STAFF generates kymographs for every time interval for every vascular segment, calculates flow velocities from red blood cell shadow angles, and outputs the data as color-coded velocity map movies and spreadsheets. In untreated mice, analyses demonstrated profound variation even between adjacent sinusoids over seconds. In acetaminophen-treated mice we detected flow reduction localized to pericentral regions. STAFF is a powerful new tool capable of providing novel insights by enabling measurement of the complex spatiotemporal dynamics of microvascular flow.
Collapse
Affiliation(s)
- Sherry G Clendenon
- Biocomplexity Institute, Indiana University, Bloomington, IN, USA; Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Xiao Fu
- Biocomplexity Institute, Indiana University, Bloomington, IN, USA; Department of Physics, Indiana University, Bloomington, IN, USA
| | - Robert A Von Hoene
- Biocomplexity Institute, Indiana University, Bloomington, IN, USA; Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | | | - James P Sluka
- Biocomplexity Institute, Indiana University, Bloomington, IN, USA; Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Seth Winfree
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Henry Mang
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | | | - Adele J Filson
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - James E Klaunig
- School of Public Health, Indiana University, Bloomington, IN, USA
| | - James A Glazier
- Biocomplexity Institute, Indiana University, Bloomington, IN, USA; Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Kenneth W Dunn
- Department of Medicine, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
50
|
VCAM-1 + macrophages guide the homing of HSPCs to a vascular niche. Nature 2018; 564:119-124. [PMID: 30455424 DOI: 10.1038/s41586-018-0709-7] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 09/14/2018] [Indexed: 12/13/2022]
Abstract
Haematopoietic stem and progenitor cells (HSPCs) give rise to all blood lineages that support the entire lifespan of vertebrates1. After HSPCs emerge from endothelial cells within the developing dorsal aorta, homing allows the nascent cells to anchor in their niches for further expansion and differentiation2-5. Unique niche microenvironments, composed of various blood vessels as units of microcirculation and other niche components such as stromal cells, regulate this process6-9. However, the detailed architecture of the microenvironment and the mechanism for the regulation of HSPC homing remain unclear. Here, using advanced live imaging and a cell-labelling system, we perform high-resolution analyses of the HSPC homing in caudal haematopoietic tissue of zebrafish (equivalent to the fetal liver in mammals), and reveal the role of the vascular architecture in the regulation of HSPC retention. We identify a VCAM-1+ macrophage-like niche cell population that patrols the inner surface of the venous plexus, interacts with HSPCs in an ITGA4-dependent manner, and directs HSPC retention. These cells, named 'usher cells', together with caudal venous capillaries and plexus, define retention hotspots within the homing microenvironment. Thus, the study provides insights into the mechanism of HSPC homing and reveals the essential role of a VCAM-1+ macrophage population with patrolling behaviour in HSPC retention.
Collapse
|