1
|
Spring BQ, Watanabe K, Ichikawa M, Mallidi S, Matsudaira T, Timerman D, Swain JWR, Mai Z, Wakimoto H, Hasan T. Red light-activated depletion of drug-refractory glioblastoma stem cells and chemosensitization of an acquired-resistant mesenchymal phenotype. Photochem Photobiol 2024. [PMID: 38922889 DOI: 10.1111/php.13985] [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: 05/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024]
Abstract
Glioblastoma stem cells (GSCs) are potent tumor initiators resistant to radiochemotherapy, and this subpopulation is hypothesized to re-populate the tumor milieu due to selection following conventional therapies. Here, we show that 5-aminolevulinic acid (ALA) treatment-a pro-fluorophore used for fluorescence-guided cancer surgery-leads to elevated levels of fluorophore conversion in patient-derived GSC cultures, and subsequent red light-activation induces apoptosis in both intrinsically temozolomide chemotherapy-sensitive and -resistant GSC phenotypes. Red light irradiation of ALA-treated cultures also exhibits the ability to target mesenchymal GSCs (Mes-GSCs) with induced temozolomide resistance. Furthermore, sub-lethal light doses restore Mes-GSC sensitivity to temozolomide, abrogating GSC-acquired chemoresistance. These results suggest that ALA is not only useful for fluorescence-guided glioblastoma tumor resection, but that it also facilitates a GSC drug-resistance agnostic, red light-activated modality to mop up the surgical margins and prime subsequent chemotherapy.
Collapse
Affiliation(s)
- Bryan Q Spring
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Physics, Northeastern University, Boston, Massachusetts, USA
| | - Kohei Watanabe
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Healthcare Optics Research Laboratory, Canon USA, Inc., Cambridge, Massachusetts, USA
| | - Megumi Ichikawa
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Tatsuyuki Matsudaira
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dmitriy Timerman
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph W R Swain
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Zhiming Mai
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hiroaki Wakimoto
- Brain Tumor Research Center and Molecular Neurosurgery Laboratory, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
2
|
Panic A, Moore J, Gallego-Perez D. Single clonal tracking on biomimetic microtextured platforms for real-time guided migration analysis of myeloid-derived suppressor cell dissemination characteristics ex vivo. Methods Cell Biol 2024; 184:97-103. [PMID: 38555161 DOI: 10.1016/bs.mcb.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Current strategies to undermine the deleterious influence of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment (TME) are lacking effective clinical solutions, in large part, due to insufficient knowledge on susceptible cellular and molecular targets. We describe here the application of biomimetic microfabricated platforms designed to analyze migratory phenotypes of MDSCs in the tumor niche ex vivo, which may enable accelerated therapeutic discovery. By mimicking the guided structural cues present in the physiological architecture of the TME, aligned microtopography substrates can elucidate potential interventions on migratory phenotypes of MDSCs at the single clonal level. Coupled with cellular and molecular biology analysis tools, our approach employs real-time tracking analysis of cell motility to probe the dissemination characteristics of MDSCs under guided migration conditions. These methods allow us to identify cellular subpopulations of interest based on their disseminative and suppressive capabilities. By doing so, we illustrate the potential of applying microscale engineering tools, in concert with dynamic live cell imaging and bioanalysis methods to uncover novel exploitable motility targets for advancing cancer therapy discovery. The inherent simplicity and extended application to a variety of contexts in tumor-associated cell migration render this method widely accessible to existing biological laboratory conditions and interests.
Collapse
Affiliation(s)
- Ana Panic
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| | - Jordan Moore
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States; Department of Surgery, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
3
|
Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, Cai H, Di Vizio D, Driedonks TAP, Erdbrügger U, Falcon‐Perez JM, Fu Q, Hill AF, Lenassi M, Lim SK, Mahoney MG, Mohanty S, Möller A, Nieuwland R, Ochiya T, Sahoo S, Torrecilhas AC, Zheng L, Zijlstra A, Abuelreich S, Bagabas R, Bergese P, Bridges EM, Brucale M, Burger D, Carney RP, Cocucci E, Colombo F, Crescitelli R, Hanser E, Harris AL, Haughey NJ, Hendrix A, Ivanov AR, Jovanovic‐Talisman T, Kruh‐Garcia NA, Ku'ulei‐Lyn Faustino V, Kyburz D, Lässer C, Lennon KM, Lötvall J, Maddox AL, Martens‐Uzunova ES, Mizenko RR, Newman LA, Ridolfi A, Rohde E, Rojalin T, Rowland A, Saftics A, Sandau US, Saugstad JA, Shekari F, Swift S, Ter‐Ovanesyan D, Tosar JP, Useckaite Z, Valle F, Varga Z, van der Pol E, van Herwijnen MJC, Wauben MHM, Wehman AM, Williams S, Zendrini A, Zimmerman AJ, MISEV Consortium, Théry C, Witwer KW. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J Extracell Vesicles 2024; 13:e12404. [PMID: 38326288 PMCID: PMC10850029 DOI: 10.1002/jev2.12404] [Citation(s) in RCA: 318] [Impact Index Per Article: 318.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/09/2024] Open
Abstract
Extracellular vesicles (EVs), through their complex cargo, can reflect the state of their cell of origin and change the functions and phenotypes of other cells. These features indicate strong biomarker and therapeutic potential and have generated broad interest, as evidenced by the steady year-on-year increase in the numbers of scientific publications about EVs. Important advances have been made in EV metrology and in understanding and applying EV biology. However, hurdles remain to realising the potential of EVs in domains ranging from basic biology to clinical applications due to challenges in EV nomenclature, separation from non-vesicular extracellular particles, characterisation and functional studies. To address the challenges and opportunities in this rapidly evolving field, the International Society for Extracellular Vesicles (ISEV) updates its 'Minimal Information for Studies of Extracellular Vesicles', which was first published in 2014 and then in 2018 as MISEV2014 and MISEV2018, respectively. The goal of the current document, MISEV2023, is to provide researchers with an updated snapshot of available approaches and their advantages and limitations for production, separation and characterisation of EVs from multiple sources, including cell culture, body fluids and solid tissues. In addition to presenting the latest state of the art in basic principles of EV research, this document also covers advanced techniques and approaches that are currently expanding the boundaries of the field. MISEV2023 also includes new sections on EV release and uptake and a brief discussion of in vivo approaches to study EVs. Compiling feedback from ISEV expert task forces and more than 1000 researchers, this document conveys the current state of EV research to facilitate robust scientific discoveries and move the field forward even more rapidly.
Collapse
Affiliation(s)
- Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of PathologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Deborah C. I. Goberdhan
- Nuffield Department of Women's and Reproductive HealthUniversity of Oxford, Women's Centre, John Radcliffe HospitalOxfordUK
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublinIreland
- Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
- Trinity St. James's Cancer InstituteTrinity College DublinDublinIreland
| | - Edit I. Buzas
- Department of Genetics, Cell‐ and ImmunobiologySemmelweis UniversityBudapestHungary
- HCEMM‐SU Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
- HUN‐REN‐SU Translational Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
| | - Cherie Blenkiron
- Faculty of Medical and Health SciencesThe University of AucklandAucklandNew Zealand
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | | | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tom A. P. Driedonks
- Department CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Uta Erdbrügger
- University of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Juan M. Falcon‐Perez
- Exosomes Laboratory, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- Metabolomics Platform, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| | - Qing‐Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Andrew F. Hill
- Institute for Health and SportVictoria UniversityMelbourneAustralia
| | - Metka Lenassi
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology (IMCB)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
- Paracrine Therapeutics Pte. Ltd.SingaporeSingapore
- Department of Surgery, YLL School of MedicineNational University SingaporeSingaporeSingapore
| | - Mỹ G. Mahoney
- Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Sujata Mohanty
- Stem Cell FacilityAll India Institute of Medical SciencesNew DelhiIndia
| | - Andreas Möller
- Chinese University of Hong KongHong KongHong Kong S.A.R.
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Susmita Sahoo
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ana C. Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP) Campus DiademaDiademaBrazil
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Andries Zijlstra
- Department of PathologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- GenentechSouth San FranciscoCaliforniaUSA
| | - Sarah Abuelreich
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Reem Bagabas
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Paolo Bergese
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
- National Center for Gene Therapy and Drugs based on RNA TechnologyPaduaItaly
| | - Esther M. Bridges
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Marco Brucale
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Dylan Burger
- Kidney Research CentreOttawa Hopsital Research InstituteOttawaCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaCanada
- School of Pharmaceutical SciencesUniversity of OttawaOttawaCanada
| | - Randy P. Carney
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Emanuele Cocucci
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| | - Federico Colombo
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Edveena Hanser
- Department of BiomedicineUniversity Hospital BaselBaselSwitzerland
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | | | - Norman J. Haughey
- Departments of Neurology and PsychiatryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and RepairGhent UniversityGhentBelgium
- Cancer Research Institute GhentGhentBelgium
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Tijana Jovanovic‐Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Nicole A. Kruh‐Garcia
- Bio‐pharmaceutical Manufacturing and Academic Resource Center (BioMARC)Infectious Disease Research Center, Colorado State UniversityFort CollinsColoradoUSA
| | - Vroniqa Ku'ulei‐Lyn Faustino
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Diego Kyburz
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Department of RheumatologyUniversity Hospital BaselBaselSwitzerland
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical NutritionInstitute of Medicine at Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Kathleen M. Lennon
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Adam L. Maddox
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Elena S. Martens‐Uzunova
- Erasmus MC Cancer InstituteUniversity Medical Center Rotterdam, Department of UrologyRotterdamThe Netherlands
| | - Rachel R. Mizenko
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Lauren A. Newman
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andrea Ridolfi
- Department of Physics and Astronomy, and LaserLaB AmsterdamVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Eva Rohde
- Department of Transfusion Medicine, University HospitalSalzburger Landeskliniken GmbH of Paracelsus Medical UniversitySalzburgAustria
- GMP Unit, Paracelsus Medical UniversitySalzburgAustria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies, EV‐TTSalzburgAustria
| | - Tatu Rojalin
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
- Expansion Therapeutics, Structural Biology and BiophysicsJupiterFloridaUSA
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andras Saftics
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Ursula S. Sandau
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Celer DiagnosticsTorontoCanada
| | - Simon Swift
- Waipapa Taumata Rau University of AucklandAucklandNew Zealand
| | - Dmitry Ter‐Ovanesyan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| | - Juan P. Tosar
- Universidad de la RepúblicaMontevideoUruguay
- Institut Pasteur de MontevideoMontevideoUruguay
| | - Zivile Useckaite
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Francesco Valle
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Zoltan Varga
- Biological Nanochemistry Research GroupInstitute of Materials and Environmental Chemistry, Research Centre for Natural SciencesBudapestHungary
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | - Edwin van der Pol
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Martijn J. C. van Herwijnen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Marca H. M. Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | | | - Andrea Zendrini
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
| | - Alan J. Zimmerman
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | | | - Clotilde Théry
- Institut Curie, INSERM U932PSL UniversityParisFrance
- CurieCoreTech Extracellular Vesicles, Institut CurieParisFrance
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
4
|
Kumari S, Kumar P. Identification and characterization of putative biomarkers and therapeutic axis in Glioblastoma multiforme microenvironment. Front Cell Dev Biol 2023; 11:1236271. [PMID: 37538397 PMCID: PMC10395518 DOI: 10.3389/fcell.2023.1236271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/23/2023] [Indexed: 08/05/2023] Open
Abstract
Non-cellular secretory components, including chemokines, cytokines, and growth factors in the tumor microenvironment, are often dysregulated, impacting tumorigenesis in Glioblastoma multiforme (GBM) microenvironment, where the prognostic significance of the current treatment remains unsatisfactory. Recent studies have demonstrated the potential of post-translational modifications (PTM) and their respective enzymes, such as acetylation and ubiquitination in GBM etiology through modulating signaling events. However, the relationship between non-cellular secretory components and post-translational modifications will create a research void in GBM therapeutics. Therefore, we aim to bridge the gap between non-cellular secretory components and PTM modifications through machine learning and computational biology approaches. Herein, we highlighted the importance of BMP1, CTSB, LOX, LOXL1, PLOD1, MMP9, SERPINE1, and SERPING1 in GBM etiology. Further, we demonstrated the positive relationship between the E2 conjugating enzymes (Ube2E1, Ube2H, Ube2J2, Ube2C, Ube2J2, and Ube2S), E3 ligases (VHL and GNB2L1) and substrate (HIF1A). Additionally, we reported the novel HAT1-induced acetylation sites of Ube2S (K211) and Ube2H (K8, K52). Structural and functional characterization of Ube2S (8) and Ube2H (1) have identified their association with protein kinases. Lastly, our results found a putative therapeutic axis HAT1-Ube2S(K211)-GNB2L1-HIF1A and potential predictive biomarkers (CTSB, HAT1, Ube2H, VHL, and GNB2L1) that play a critical role in GBM pathogenesis.
Collapse
|
5
|
Yuile A, Wei JQ, Mohan AA, Hotchkiss KM, Khasraw M. Interdependencies of the Neuronal, Immune and Tumor Microenvironment in Gliomas. Cancers (Basel) 2023; 15:2856. [PMID: 37345193 PMCID: PMC10216320 DOI: 10.3390/cancers15102856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
Gliomas are the most common primary brain malignancy and are universally fatal. Despite significant breakthrough in understanding tumor biology, treatment breakthroughs have been limited. There is a growing appreciation that major limitations on effective treatment are related to the unique and highly complex glioma tumor microenvironment (TME). The TME consists of multiple different cell types, broadly categorized into tumoral, immune and non-tumoral, non-immune cells. Each group provides significant influence on the others, generating a pro-tumor dynamic with significant immunosuppression. In addition, glioma cells are highly heterogenous with various molecular distinctions on the cellular level. These variations, in turn, lead to their own unique influence on the TME. To develop future treatments, an understanding of this complex TME interplay is needed. To this end, we describe the TME in adult gliomas through interactions between its various components and through various glioma molecular phenotypes.
Collapse
Affiliation(s)
- Alexander Yuile
- Department of Medical Oncology, Royal North Shore Hospital, Reserve Road, St Leonards, NSW 2065, Australia
- The Brain Cancer Group, North Shore Private Hospital, 3 Westbourne Street, St Leonards, NSW 2065, Australia
- Sydney Medical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Joe Q. Wei
- Department of Medical Oncology, Royal North Shore Hospital, Reserve Road, St Leonards, NSW 2065, Australia
- Sydney Medical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Aditya A. Mohan
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA
| | - Kelly M. Hotchkiss
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA
| | - Mustafa Khasraw
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA
| |
Collapse
|
6
|
Muralidharan A, Pesch GR, Hubbe H, Rems L, Nouri-Goushki M, Boukany PE. Microtrap array on a chip for localized electroporation and electro-gene transfection. Bioelectrochemistry 2022; 147:108197. [PMID: 35810498 DOI: 10.1016/j.bioelechem.2022.108197] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/09/2022] [Accepted: 06/25/2022] [Indexed: 11/19/2022]
Abstract
We developed a localized single-cell electroporation chip to deliver exogenous biomolecules with high efficiency while maintaining high cell viability. In our microfluidic device, the cells are trapped in a microtrap array by flow, after which target molecules are supplied to the device and electrotransferred to the cells under electric pulses. The system provides the ability to monitor the electrotransfer of exogenous biomolecules in real time. We reveal through numerical simulations that localized electroporation is the mechanism of permeabilization in the microtrap array electroporation device. We demonstrate the simplicity and accuracy of this microtrap technology for electroporation by delivery of both small molecules using propidium iodide and large molecules using plasmid DNA for gene expression, illustrating the potential of this minimally invasive method to be widely used for precise intracellular delivery purposes (from bioprocess engineering to therapeutic applications).
Collapse
Affiliation(s)
- Aswin Muralidharan
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands.
| | - Georg R Pesch
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands
| | - Hendrik Hubbe
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands
| | - Lea Rems
- Faculty of Electrical Engineering, University of Ljubljana, Trzaska 25, 1000 Ljubljana, Slovenia
| | - Mahdiyeh Nouri-Goushki
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands.
| |
Collapse
|
7
|
Zhang A, Fang J, Li X, Wang J, Chen M, Chen HJ, He G, Xie X. Cellular nanointerface of vertical nanostructure arrays and its applications. NANOSCALE ADVANCES 2022; 4:1844-1867. [PMID: 36133409 PMCID: PMC9419580 DOI: 10.1039/d1na00775k] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/28/2021] [Indexed: 06/16/2023]
Abstract
Vertically standing nanostructures with various morphologies have been developed with the emergence of the micro-/nanofabrication technology. When cells are cultured on them, various bio-nano interfaces between cells and vertical nanostructures would impact the cellular activities, depending on the shape, density, and height of nanostructures. Many cellular pathway activation processes involving a series of intracellular molecules (proteins, RNA, DNA, enzymes, etc.) would be triggered by the cell morphological changes induced by nanostructures, affecting the cell proliferation, apoptosis, differentiation, immune activation, cell adhesion, cell migration, and other behaviors. In addition, the highly localized cellular nanointerface enhances coupled stimulation on cells. Therefore, understanding the mechanism of the cellular nanointerface can not only provide innovative tools for regulating specific cell functions but also offers new aspects to understand the fundamental cellular activities that could facilitate the precise monitoring and treatment of diseases in the future. This review mainly describes the fabrication technology of vertical nanostructures, analyzing the formation of cellular nanointerfaces and the effects of cellular nanointerfaces on cells' fates and functions. At last, the applications of cellular nanointerfaces based on various nanostructures are summarized.
Collapse
Affiliation(s)
- Aihua Zhang
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
| | - Jiaru Fang
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
| | - Xiangling Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
- School of Biomedical Engineering, Sun Yat-Sen University Guangzhou 510006 China
| | - Ji Wang
- The First Affiliated Hospital of Sun Yat-Sen University Guangzhou 510080 China
| | - Meiwan Chen
- Institute of Chinese Medical Sciences, University of Macau Taipa Macau SAR China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
- Key Laboratory of Molecular Target & Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
- The First Affiliated Hospital of Sun Yat-Sen University Guangzhou 510080 China
| |
Collapse
|
8
|
Shen S, Lin S, Chen Y, Zhang Y, He Y, Xu X, Feng Y, Lu Y, Mo R. Combating Cancer Stem-Like Cell-Derived Resistance to Anticancer Protein by Liposome-Mediated Acclimatization Strategy. NANO LETTERS 2022; 22:2419-2428. [PMID: 35254834 DOI: 10.1021/acs.nanolett.2c00004] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Antibody-based therapeutics, which induce apoptosis of malignant cells by selectively binding to their receptors, hold tremendous promise for clinical cancer therapy. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has received considerable interest due to its favorable capability of activating apoptosis in cancer cells by interacting with death receptors (DRs). However, cancer stem-like cells (CSCs) show deficient or lower DR and are highly resistant to TRAIL-mediated apoptosis limiting the therapeutic efficacy. Here, we report a liposome-mediated acclimatization strategy to overcome the CSC-emanated TRAIL resistance. The liposomal assemblies coencapsulating plasmid DNA encoding TRAIL and salinomycin enable cancer cells as protein generators to express TRAIL, and more importantly, can acclimatize resistant CSCs to be sensitized to the TRAIL-triggered apoptosis by salinomycin-induced upregulation of DR expression on CSCs. This programmable liposome-based drug codelivery system shows the potential to efficiently eliminate CSCs and inhibit CSC-enriched tumor growth in the orthotopic colon tumor mouse model.
Collapse
Affiliation(s)
- Shiyang Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Shiqi Lin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yuying Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yingjiao He
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Feng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yougong Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
9
|
Duarte-Sanmiguel S, Panic A, Dodd DJ, Salazar-Puerta A, Moore JT, Lawrence WR, Nairon K, Francis C, Zachariah N, McCoy W, Turaga R, Skardal A, Carson WE, Higuita-Castro N, Gallego-Perez D. In Situ Deployment of Engineered Extracellular Vesicles into the Tumor Niche via Myeloid-Derived Suppressor Cells. Adv Healthc Mater 2022; 11:e2101619. [PMID: 34662497 PMCID: PMC8891033 DOI: 10.1002/adhm.202101619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/26/2021] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs) have emerged as a promising carrier system for the delivery of therapeutic payloads in multiple disease models, including cancer. However, effective targeting of EVs to cancerous tissue remains a challenge. Here, it is shown that nonviral transfection of myeloid-derived suppressor cells (MDSCs) can be leveraged to drive targeted release of engineered EVs that can modulate transfer and overexpression of therapeutic anticancer genes in tumor cells and tissue. MDSCs are immature immune cells that exhibit enhanced tropism toward tumor tissue and play a role in modulating tumor progression. Current MDSC research has been mostly focused on mitigating immunosuppression in the tumor niche; however, the tumor homing abilities of these cells present untapped potential to deliver EV therapeutics directly to cancerous tissue. In vivo and ex vivo studies with murine models of breast cancer show that nonviral transfection of MDSCs does not hinder their ability to home to cancerous tissue. Moreover, transfected MDSCs can release engineered EVs and mediate antitumoral responses via paracrine signaling, including decreased invasion/metastatic activity and increased apoptosis/necrosis. Altogether, these findings indicate that MDSCs can be a powerful tool for the deployment of EV-based therapeutics to tumor tissue.
Collapse
Affiliation(s)
| | - Ana Panic
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Daniel J. Dodd
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
- The Ohio State University, Biomedical Sciences Graduate Program, Columbus, OH 43210
| | - Ana Salazar-Puerta
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Jordan T. Moore
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - William R. Lawrence
- The Ohio State University, Biomedical Sciences Graduate Program, Columbus, OH 43210
| | - Kylie Nairon
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Carlie Francis
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Natalie Zachariah
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - William McCoy
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Rithvik Turaga
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Aleksander Skardal
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - William E. Carson
- The Ohio State University, Department of Surgery, Columbus, OH 43210
| | - Natalia Higuita-Castro
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
- The Ohio State University, Department of Surgery, Columbus, OH 43210
- The Ohio State University, Biophysics Program, OH 43210
| | - Daniel Gallego-Perez
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
- The Ohio State University, Department of Surgery, Columbus, OH 43210
| |
Collapse
|
10
|
The Advances in Glioblastoma On-a-Chip for Therapy Approaches. Cancers (Basel) 2022; 14:cancers14040869. [PMID: 35205617 PMCID: PMC8870462 DOI: 10.3390/cancers14040869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary This systematic review showed different therapeutic approaches to glioblastoma on-a-chip with varying levels of complexity, answering, from the simplest question to the most sophisticated questions, in a biological system integrated in an efficient way. With advances in manufacturing protocols, soft lithography in PDMS material was the most used in the studies, applying different strategy geometrics in device construction. The microenvironment showed the relevant elaborations in co-culture between mainly human tumor cells and support cells involved in the collagen type I matrix; remaining an adequate way to assess the therapeutic approach. The most complex devices showed efficient intersection between different systems, allowing in vitro studies with major human genetic similarity, reproducibility, and low cost, on a highly customizable platform. Abstract This systematic review aimed to verify the use of microfluidic devices in the process of implementing and evaluating the effectiveness of therapeutic approaches in glioblastoma on-a-chip, providing a broad view of advances to date in the use of this technology and their perspectives. We searched studies with the variations of the keywords “Glioblastoma”, “microfluidic devices”, “organ-on-a-chip” and “therapy” of the last ten years in PubMed and Scopus databases. Of 446 articles identified, only 22 articles were selected for analysis according to the inclusion and exclusion criteria. The microfluidic devices were mainly produced by soft lithography technology, using the PDMS material (72%). In the microenvironment, the main extracellular matrix used was collagen type I. Most studies used U87-MG glioblastoma cells from humans and 31.8% were co-cultivated with HUVEC, hCMEC/D3, and astrocytes. Chemotherapy was the majority of therapeutic approaches, assessing mainly the cellular viability and proliferation. Furthermore, some alternative therapies were reported in a few studies (22.6%). This study identified a diversity of glioblastoma on-a-chip to assess therapeutic approaches, often using intermediate levels of complexity. The most advanced level implemented the intersection between different biological systems (liver–brain or intestine–liver–brain), BBB model, allowing in vitro studies with greater human genetic similarity, reproducibility, and low cost, in a highly customizable platform.
Collapse
|
11
|
Li SZ, Ren KX, Zhao J, Wu S, Li J, Zang J, Fei Z, Zhao JL. miR-139/PDE2A-Notch1 feedback circuit represses stemness of gliomas by inhibiting Wnt/β-catenin signaling. Int J Biol Sci 2021; 17:3508-3521. [PMID: 34512162 PMCID: PMC8416740 DOI: 10.7150/ijbs.62858] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/01/2021] [Indexed: 01/13/2023] Open
Abstract
Rationale: The malignant phenotypes of glioblastomas (GBMs) are primarily attributed to glioma stem cells (GSCs). Our previous study and other reports have suggested that both miR-139 and its host gene PDE2A are putative antitumor genes in various cancers. The aim of this study was to investigate the roles and mechanisms of miR-139/PDE2A in GSC modulation. Methods: Clinical samples were used to determine miR-139/PDE2A expression. Patient-derived glioma stem-like cells (PD-GSCs) were stimulated for immunofluorescent staining, sphere formation assays and orthotopic GBM xenograft models. Bioinformatic analysis and further in vitro experiments demonstrated the downstream molecular mechanisms of miR-139 and PDE2A. OX26/CTX-conjugated PEGylated liposome (OCP) was constructed to deliver miR-139 or PDE2A into glioma tissue specifically. Results: We demonstrated that miR-139 was concomitantly transcribed with its host gene PDE2A. Both PDE2A and miR-139 indicated better prognosis of gliomas and were inversely correlated with GSC stemness. PDE2A or miR-139 overexpression suppressed the stemness of PD-GSCs. FZD3 and β-catenin, which induced Wnt/β-catenin signaling activation, were identified as targets of miR-139 and mediated the effects of miR-139 on GSCs. Meanwhile, PDE2A suppressed Wnt/β-catenin signaling by inhibiting cAMP accumulation and GSK-3β phosphorylation, thereby modulating the self-renewal of PD-GSCs. Notably, Notch1, which is also a target of miR-139, suppressed PDE2A/miR-139 expression directly via downstream Hes1, indicating that miR-139 promoted its own expression by the miR-139-Notch1/Hes1 feedback circuit. Expectedly, targeted overexpression miR-139 or PDE2A in glioma with OCP system significantly repressed the stemness and decelerated glioma progression. Conclusions: Our findings elaborate on the inhibitory functions of PDE2A and miR-139 on GSC stemness and tumorigenesis, which may provide new prognostic markers and therapeutic targets for GBMs.
Collapse
Affiliation(s)
- San-Zhong Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Kai-Xi Ren
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Zhao
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Shuang Wu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Juan Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jian Zang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jun-Long Zhao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
12
|
Chelliah SS, Paul EAL, Kamarudin MNA, Parhar I. Challenges and Perspectives of Standard Therapy and Drug Development in High-Grade Gliomas. Molecules 2021; 26:1169. [PMID: 33671796 PMCID: PMC7927069 DOI: 10.3390/molecules26041169] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Despite their low incidence rate globally, high-grade gliomas (HGG) remain a fatal primary brain tumor. The recommended therapy often is incapable of resecting the tumor entirely and exclusively targeting the tumor leads to tumor recurrence and dismal prognosis. Additionally, many HGG patients are not well suited for standard therapy and instead, subjected to a palliative approach. HGG tumors are highly infiltrative and the complex tumor microenvironment as well as high tumor heterogeneity often poses the main challenges towards the standard treatment. Therefore, a one-fit-approach may not be suitable for HGG management. Thus, a multimodal approach of standard therapy with immunotherapy, nanomedicine, repurposing of older drugs, use of phytochemicals, and precision medicine may be more advantageous than a single treatment model. This multimodal approach considers the environmental and genetic factors which could affect the patient's response to therapy, thus improving their outcome. This review discusses the current views and advances in potential HGG therapeutic approaches and, aims to bridge the existing knowledge gap that will assist in overcoming challenges in HGG.
Collapse
Affiliation(s)
- Shalini Sundramurthi Chelliah
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Ervin Ashley Lourdes Paul
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| | - Muhamad Noor Alfarizal Kamarudin
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| | - Ishwar Parhar
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| |
Collapse
|
13
|
Ghaemi A, Bagheri E, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. CRISPR-cas9 genome editing delivery systems for targeted cancer therapy. Life Sci 2020; 267:118969. [PMID: 33385410 DOI: 10.1016/j.lfs.2020.118969] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
The prokaryotic CRISPR-Cas systems could be applied as revolutionized genome editing tool in live cells of various species to modify, visualize and identify definite sequences of DNA and RNA. CRISPR-Cas could edit the genome by homology-directed repair and non-homologous end joining mechanisms. Furthermore, DNA-targeting modification by CRISPR-Cas methodology provides opportunity for diagnosis, therapy and the genetic disorders investigation. Here, we summarized delivery systems employed for CRISPR-Cas9 for genome editing. Then preclinical studies of the CRISPR-Cas9-based therapeutics will be discussed considering the associated challenges and developments in its translation to clinic for cancer therapy.
Collapse
Affiliation(s)
- Asma Ghaemi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Bagheri
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Dong Z, Xue X, Liang H, Guan J, Chang L. DNA Nanomachines for Identifying Cancer Biomarkers in Body Fluids and Cells. Anal Chem 2020; 93:1855-1865. [PMID: 33325676 DOI: 10.1021/acs.analchem.0c03518] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Identifying molecular biomarkers promises to significantly improve the accuracy in cancer diagnosis at its early stage. DNA nanomachines, which are designable and switchable nanostructures made of DNA, show broad potential to detect tumor biomarkers with noninvasive, inexpensive, highly sensitive, and highly specific advantages. This Feature summarizes the recent DNA nanomachine-based platforms for the early detection of cancer biomarkers, both from body fluids and in cells.
Collapse
Affiliation(s)
- Zaizai Dong
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.,Department of Respiratory and Critical Care, Chinese PLA General Hospital, Beijing 100853, China
| | - Hailun Liang
- School of Public Administration and Policy, Renmin University of China, Beijing 100872, China
| | - Jingjiao Guan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida 32310, United States
| | - Lingqian Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.,School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
15
|
Brooks J, Minnick G, Mukherjee P, Jaberi A, Chang L, Espinosa HD, Yang R. High Throughput and Highly Controllable Methods for In Vitro Intracellular Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004917. [PMID: 33241661 PMCID: PMC8729875 DOI: 10.1002/smll.202004917] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/06/2020] [Indexed: 05/03/2023]
Abstract
In vitro and ex vivo intracellular delivery methods hold the key for releasing the full potential of tissue engineering, drug development, and many other applications. In recent years, there has been significant progress in the design and implementation of intracellular delivery systems capable of delivery at the same scale as viral transfection and bulk electroporation but offering fewer adverse outcomes. This review strives to examine a variety of methods for in vitro and ex vivo intracellular delivery such as flow-through microfluidics, engineered substrates, and automated probe-based systems from the perspective of throughput and control. Special attention is paid to a particularly promising method of electroporation using micro/nanochannel based porous substrates, which expose small patches of cell membrane to permeabilizing electric field. Porous substrate electroporation parameters discussed include system design, cells and cargos used, transfection efficiency and cell viability, and the electric field and its effects on molecular transport. The review concludes with discussion of potential new innovations which can arise from specific aspects of porous substrate-based electroporation platforms and high throughput, high control methods in general.
Collapse
Affiliation(s)
- Justin Brooks
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Grayson Minnick
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Arian Jaberi
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Horacio D. Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
16
|
Shukla VC, Duarte-Sanmiguel S, Panic A, Senthilvelan A, Moore J, Bobba C, Benner B, Carson WE, Ghadiali SN, Gallego-Perez D. Reciprocal Signaling between Myeloid Derived Suppressor and Tumor Cells Enhances Cellular Motility and is Mediated by Structural Cues in the Microenvironment. ADVANCED BIOSYSTEMS 2020; 4:e2000049. [PMID: 32419350 PMCID: PMC7489303 DOI: 10.1002/adbi.202000049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/10/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022]
Abstract
Myeloid derived suppressor cells (MDSCs) have gained significant attention for their immunosuppressive role in cancer and their ability to contribute to tumor progression and metastasis. Understanding the role of MDSCs in driving cancer cell migration, a process fundamental to metastasis, is essential to fully comprehend and target MDSC-tumor cell interactions. This study employs microfabricated platforms, which simulate the structural cues present in the tumor microenvironment (TME) to elucidate the effects of MDSCs on the migratory phenotype of cancer cells at the single cell level. The results indicate that the presence of MDSCs enhances the motility of cancer-epithelial cells when directional cues (either topographical or spatial) are present. This behavior appears to be independent of cell-cell contact and driven by soluble byproducts from heterotypic interactions between MDSCs and cancer cells. Moreover, MDSC cell-motility is also impacted by the presence of cancer cells and the cancer cell secretome in the presence of directional cues. Epithelial dedifferentiation is the likely mechanism for changes in cancer cell motility in response to MDSCs. These results highlight the biochemical and biostructural conditions under which MDSCs can support cancer cell migration, and could therefore provide new avenues of research and therapy aimed at stemming cancer progression.
Collapse
Affiliation(s)
- Vasudha C. Shukla
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 USA
| | - Silvia Duarte-Sanmiguel
- Department of Biomedical Engineering, OSU Nutrition, The Ohio State University, Columbus, OH, 43210, USA
| | - Ana Panic
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Abirami Senthilvelan
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Jordan Moore
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Christopher Bobba
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Brooke Benner
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, 43210, USA
| | - William E. Carson
- Department of Surgery, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Samir N. Ghadiali
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Dorothy M. Davis Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart and lung Research Institute, Department of Surgery, The Ohio State Wexner Medical Center, Columbus, OH, 43210, USA
| |
Collapse
|
17
|
Guided migration analyses at the single-clone level uncover cellular targets of interest in tumor-associated myeloid-derived suppressor cell populations. Sci Rep 2020; 10:1189. [PMID: 31988310 PMCID: PMC6985212 DOI: 10.1038/s41598-020-57941-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/08/2020] [Indexed: 12/15/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immune cells that exert immunosuppression within the tumor, protecting cancer cells from the host’s immune system and/or exogenous immunotherapies. While current research has been mostly focused in countering MDSC-driven immunosuppression, little is known about the mechanisms by which MDSCs disseminate/infiltrate cancerous tissue. This study looks into the use of microtextured surfaces, coupled with in vitro and in vivo cellular and molecular analysis tools, to videoscopically evaluate the dissemination patterns of MDSCs under structurally guided migration, at the single-cell level. MDSCs exhibited topographically driven migration, showing significant intra- and inter-population differences in motility, with velocities reaching ~40 μm h−1. Downstream analyses coupled with single-cell migration uncovered the presence of specific MDSC subpopulations with different degrees of tumor-infiltrating and anti-inflammatory capabilities. Granulocytic MDSCs showed a ~≥3-fold increase in maximum dissemination velocities and traveled distances, and a ~10-fold difference in the expression of pro- and anti-inflammatory markers. Prolonged culture also revealed that purified subpopulations of MDSCs exhibit remarkable plasticity, with homogeneous/sorted subpopulations giving rise to heterogenous cultures that represented the entire hierarchy of MDSC phenotypes within 7 days. These studies point towards the granulocytic subtype as a potential cellular target of interest given their superior dissemination ability and enhanced anti-inflammatory activity.
Collapse
|
18
|
Dong Z, Jiao Y, Xie B, Hao Y, Wang P, Liu Y, Shi J, Chitrakar C, Black S, Wang YC, Lee LJ, Li M, Fan Y, Chang L. On-chip multiplexed single-cell patterning and controllable intracellular delivery. MICROSYSTEMS & NANOENGINEERING 2020; 6:2. [PMID: 34567617 PMCID: PMC8433345 DOI: 10.1038/s41378-019-0112-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/23/2019] [Accepted: 10/10/2019] [Indexed: 05/03/2023]
Abstract
Conventional electroporation approaches show limitations in the delivery of macromolecules in vitro and in vivo. These limitations include low efficiency, noticeable cell damage and nonuniform delivery of cells. Here, we present a simple 3D electroporation platform that enables massively parallel single-cell manipulation and the intracellular delivery of macromolecules and small molecules. A pyramid pit micropore array chip was fabricated based on a silicon wet-etching method. A controllable vacuum system was adopted to trap a single cell on each micropore. Using this chip, safe single-cell electroporation was performed at low voltage. Cargoes of various sizes ranging from oligonucleotides (molecular beacons, 22 bp) to plasmid DNA (CRISPR-Cas9 expression vectors, >9 kb) were delivered into targeted cells with a significantly higher transfection efficiency than that of multiple benchmark methods (e.g., commercial electroporation devices and Lipofectamine). The delivered dose of the chemotherapeutic drug could be controlled by adjusting the applied voltage. By using CRISPR-Cas9 transfection with this system, the p62 gene and CXCR7 gene were knocked out in tumor cells, which effectively inhibited their cellular activity. Overall, this vacuum-assisted micropore array platform provides a simple, efficient, high-throughput intracellular delivery method that may facilitate on-chip cell manipulation, intracellular investigation and cancer therapy.
Collapse
Affiliation(s)
- Zaizai Dong
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Yanli Jiao
- College of Agricultural and Life Sciences, University of Florida, Gainesville, FL 32611 USA
| | - Bingteng Xie
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yongcun Hao
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, Northwestern Polytechnical University, 710072 Xi’an, China
| | - Pan Wang
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yuanyuan Liu
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Junfeng Shi
- Chemical and Biomolecular Engineering Department, Ohio State University, Columbus, OH 43209 USA
| | - Chandani Chitrakar
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| | - Stephen Black
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| | - Yu-Chieh Wang
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - L. James Lee
- Chemical and Biomolecular Engineering Department, Ohio State University, Columbus, OH 43209 USA
| | - Mo Li
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yubo Fan
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| |
Collapse
|
19
|
Chang L, Wang YC, Ershad F, Yang R, Yu C, Fan Y. Wearable Devices for Single-Cell Sensing and Transfection. Trends Biotechnol 2019; 37:1175-1188. [DOI: 10.1016/j.tibtech.2019.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/31/2019] [Accepted: 04/02/2019] [Indexed: 02/01/2023]
|
20
|
Park D, Lee J, Chung JJ, Jung Y, Kim SH. Integrating Organs-on-Chips: Multiplexing, Scaling, Vascularization, and Innervation. Trends Biotechnol 2019; 38:99-112. [PMID: 31345572 DOI: 10.1016/j.tibtech.2019.06.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022]
Abstract
Organs-on-chips (OoCs) have attracted significant attention because they can be designed to mimic in vivo environments. Beyond constructing a single OoC, recent efforts have tried to integrate multiple OoCs to broaden potential applications such as disease modeling and drug discoveries. However, various challenges remain for integrating OoCs towards in vivo-like operation, such as incorporating various connections for integrating multiple OoCs. We review multiplexed OoCs and challenges they face: scaling, vascularization, and innervation. In our opinion, future OoCs will be constructed to have increased predictive power for in vivo phenomena and will ultimately become a mainstream tool for high quality biomedical and pharmaceutical research.
Collapse
Affiliation(s)
- DoYeun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jaeseo Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Justin J Chung
- Biomaterials Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Soo Hyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Biomaterials Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
21
|
Wang J, Zuo J, Wahafu A, Wang MD, Li RC, Xie WF. Combined elevation of TRIB2 and MAP3K1 indicates poor prognosis and chemoresistance to temozolomide in glioblastoma. CNS Neurosci Ther 2019; 26:297-308. [PMID: 31318172 PMCID: PMC7053231 DOI: 10.1111/cns.13197] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/27/2019] [Accepted: 07/03/2019] [Indexed: 01/09/2023] Open
Abstract
Introduction Glioblastoma (GBM) is the most lethal primary malignant brain tumor in adults with poor survival due to acquired therapeutic resistance and rapid recurrence. Currently, the standard clinical strategy for glioma includes maximum surgical resection, radiotherapy, and temozolomide (TMZ) chemotherapy; however, the median survival of patients with GBM remains poor despite these comprehensive therapies. Therefore, the identification of new prognostic biomarkers is urgently needed to evaluate the malignancy and long‐term outcome of glioma. Aims To further investigate prognostic biomarkers and potential therapeutic targets for GBM. Results In this study, we identified tribbles pseudokinase 2 (TRIB2) as one of the genes that is most correlated with pathological classification, radioresistance, and TMZ resistance in glioma. Additionally, the expression of mitogen‐activated protein kinase kinase kinase 1 (MAP3K1) showed a strong correlation with TRIB2. Moreover, a combined increase in TRIB2 and MAP3K1 was observed in GBM and indicated a poor prognosis of patients with glioma. Finally, enriched TRIB2 expression and MAP3K1 expression were shown to be associated with resistance to TMZ and radiotherapy. Conclusion Combined elevation of TRIB2 and MAP3K1 could be novel prognostic biomarkers and potential therapeutic targets to evaluate the malignancy and long‐term outcomes of GBM.
Collapse
Affiliation(s)
- Jia Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jie Zuo
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Alafate Wahafu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mao-de Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui-Chun Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wan-Fu Xie
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Vlachou M, Kikionis S, Siamidi A, Tragou K, Kapoti S, Ioannou E, Roussis V, Tsotinis A. Fabrication and Characterization of Electrospun Nanofibers for the Modified Release of the Chronobiotic Hormone Melatonin. Curr Drug Deliv 2019; 16:79-85. [PMID: 30215335 PMCID: PMC6340153 DOI: 10.2174/1567201815666180914095701] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/20/2018] [Accepted: 09/11/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Aiming at the modified release of melatonin (MLT), electrospun-MLT loaded nanofibers, filled into hard gelatin and DRcapsTM capsules, were used as formulants. METHODS Cellulose acetate, polyvinylpyrrolidinone and hydroxypropylmethylcellusose (HPMC 2910) were used for the preparation of the fiber matrices through electrospinning. The in vitro modified release profile of MLT from the fabricated matrices in gastrointestinal-like fluids was studied. At pH 1.2, the formulations CA1, CA2, PV1, HP1, HP2 and the composite formulations CAPV1-CAPV5 in hard gelatin capsules exhibited fast MLT release. RESULTS In general, the same trend was observed at pH 6.8, with the exception of CAPV1 and CAPV2. These two composite formulations delivered 52.08% and 75.25% MLT, respectively at a slower pace (6 h) when encapsulated in DRcapsTM capsules. In all other cases, the release of MLT from DRcapsTM capsules filled with the MLT-loaded nanofibers reached 100% at 6h. CONCLUSION These findings suggest that the MLT-loaded nanofibrous mats developed in this study exhibit a promising profile for treating sleep dysfunctions.
Collapse
Affiliation(s)
- Marilena Vlachou
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefanos Kikionis
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Angeliki Siamidi
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantina Tragou
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefania Kapoti
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathia Ioannou
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilios Roussis
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Andrew Tsotinis
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
23
|
Huang D, Zhao D, Li J, Wu Y, Du L, Xia XH, Li X, Deng Y, Li Z, Huang Y. Continuous Vector-free Gene Transfer with a Novel Microfluidic Chip and Nanoneedle Array. Curr Drug Deliv 2019; 16:164-170. [PMID: 30332957 DOI: 10.2174/1567201815666181017095044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/24/2018] [Accepted: 10/09/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Delivery of foreign cargoes into cells is of great value for bioengineering research and therapeutic applications. OBJECTIVE In this study, we proposed and established a carrier-free gene delivery platform utilizing staggered herringbone channel and silicon nanoneedle array, to achieve high-throughput in vitro gene transfection. METHODS With this microchip, fluidic micro vortices could be induced by the staggered-herringboneshaped grooves within the channel, which increased the contact frequency of the cells with the channel substrate. Transient disruptions on the cell membrane were well established by the nanoneedle array on the substrate. RESULT Compared to the conventional nanoneedle-based delivery system, proposed microfluidic chip achieved flow-through treatment with high gene transfection efficiency (higher than 20%) and ideal cell viability (higher than 95%). CONCLUSION It provides a continuous processing environment that can satisfy the transfection requirement of large amounts of biological molecules, showing high potential and promising prospect for both basic research and clinical application.
Collapse
Affiliation(s)
- Dong Huang
- Institute of Molecular Medicine; Institute of Microelectronics, National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Peking University, Beijing 100871, China
| | - Deyao Zhao
- Institute of Molecular Medicine; Institute of Microelectronics, National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Peking University, Beijing 100871, China
| | - Jinhui Li
- Institute of Molecular Medicine; Institute of Microelectronics, National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Peking University, Beijing 100871, China
| | - Yuting Wu
- Institute of Molecular Medicine; Institute of Microelectronics, National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Peking University, Beijing 100871, China
| | - Lili Du
- Institute of Molecular Medicine; Institute of Microelectronics, National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Peking University, Beijing 100871, China
| | - Xin-Hua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xiaoqiong Li
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Key Laboratory of Molecular Medicine and Biotheranotics, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yulin Deng
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Key Laboratory of Molecular Medicine and Biotheranotics, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhihong Li
- Institute of Molecular Medicine; Institute of Microelectronics, National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Peking University, Beijing 100871, China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Key Laboratory of Molecular Medicine and Biotheranotics, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
24
|
He X, Maimaiti M, Jiao Y, Meng X, Li H. Sinomenine Induces G1-Phase Cell Cycle Arrest and Apoptosis in Malignant Glioma Cells Via Downregulation of Sirtuin 1 and Induction of p53 Acetylation. Technol Cancer Res Treat 2019; 17:1533034618770305. [PMID: 29756546 PMCID: PMC5952277 DOI: 10.1177/1533034618770305] [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: 12/25/2022] Open
Abstract
Sinomenine, a bioactive alkaloid isolated from the traditional Chinese herb Sinomenium acutum, possesses antiinflammatory, antinociceptive, antifibrotic, and antitumorigenic properties. In this work, we sought to explore the biological effects of sinomenine on glioma cells. It was found that sinomenine caused a concentration-dependent inhibition of viability in both U87 and U251 glioma cells. Sinomenine at 16 μmol/L caused 55% to 60% reduction in the proliferation of U87 and U251 cells. Moreover, sinomenine treatment induced a G0/G1 cell cycle arrest and apoptosis. Mechanistically, sinomenine promoted p53 expression and acetylation and reduced the expression of sirtuin 1. Ectopic expression of sirtuin 1 significantly prevented sinomenine-induced p53 acetylation and growth suppression in glioma cells. Moreover, sinomenine inhibited the growth of U87 xenograft tumors in vivo and raised the p53 protein expression. Collectively, sinomenine shows antiproliferative effects against glioma cells which is mediated through downregulation of sirtuin 1 and induction of p53 activity.
Collapse
Affiliation(s)
- Xiaoyan He
- 1 Department of Neurology, The Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, China
| | - Mayinur Maimaiti
- 1 Department of Neurology, The Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, China
| | - Yan Jiao
- 1 Department of Neurology, The Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, China
| | - Xuegang Meng
- 1 Department of Neurology, The Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, China
| | - Hongyan Li
- 1 Department of Neurology, The Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, China
| |
Collapse
|
25
|
Hong J, Kim B, Shin H. Mixed-scale poly(methyl methacrylate) channel network-based single-particle manipulation via diffusiophoresis. NANOSCALE 2018; 10:14421-14431. [PMID: 29796559 DOI: 10.1039/c7nr07669j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Despite the unique advantages of nanochannels imparted by their small size, their utility is limited by the lack of affordable and versatile fabrication methods. Moreover, nanochannel-incorporated fluidic devices require micro-sized conduit integration for efficient access of liquid samples. In this study, a simple and cost-effective fabrication method for mixed-scale channel networks via hot-embossing of poly(methyl methacrylate) (PMMA) using a carbon stamp is demonstrated. Due to its high rigidity, PMMA ensures collapse-free channel fabrication. The carbon stamp is fabricated using only batch microfabrication and has a convex architecture that allows the fabrication of a complex channel network via a single imprinting process. In addition, the microchannels are connected to nanochannels via three-dimensional (3D) microfunnels that serve as single-particle-entrapment chambers, ensuring smooth transport of samples into the nanochannels. Owing to the 3D geometry of the microfunnels and the small size of the nanochannels, a solute gradient can be generated locally at the microfunnel. This local solute gradient enables the entrapment of microparticles at the microfunnels via diffusiophoresis, which can manipulate the particle motion in a controllable manner, without any external equipment or additional electrode integration into the channels. To the best of our knowledge, this is the first report of diffusiophoresis-based single-particle entrapment.
Collapse
Affiliation(s)
- Jisoo Hong
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| | | | | |
Collapse
|
26
|
Burtch SR, Sameti M, Olmstead RT, Bashur CA. Rapid generation of three-dimensional microchannels for vascularization using a subtractive printing technique. JOURNAL OF BIOPHOTONICS 2018; 11:e201700226. [PMID: 29356372 DOI: 10.1002/jbio.201700226] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/18/2018] [Indexed: 06/07/2023]
Abstract
The development of tissue-engineered products has been limited by lack of a perfused microvasculature that delivers nutrients and maintains cell viability. Current strategies to promote vascularization such as additive three-dimensional printing techniques have limitations. This study validates the use of an ultra-fast laser subtractive printing technique to generate capillary-sized channels in hydrogels prepopulated with cells by demonstrating cell viability relative to the photodisrupted channels in the gel. The system can move the focal spot laterally in the gel at a rate of 2500 mm/s by using a galvanometric scanner to raster the in plane focal spot. A Galilean telescope allows z-axis movement. Blended hydrogels of polyethylene glycol and collagen with a range of optical clarities, mechanical properties and swelling behavior were tested to demonstrate that the subtractive printing process for writing vascular channels is compatible with all of the blended hydrogels tested. Channel width and patterns were controlled by adjusting the laser energy and focal spot positioning, respectively. After treatment, high cell viability was observed at distances greater than or equal to 18 μm from the fabricated channels. Overall, this study demonstrates a flexible technique that has the potential to rapidly generate channels in tissue-engineered constructs.
Collapse
Affiliation(s)
- Stephanie R Burtch
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, Florida
| | - Mahyar Sameti
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, Florida
| | - Richard T Olmstead
- Department of Electrical Engineering, Florida Institute of Technology, Melbourne, Florida
- Medical Optics and Photonics Incorporated (MEDOPHO), Oviedo, Florida
| | - Chris A Bashur
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, Florida
| |
Collapse
|
27
|
Wiraja C, Yeo DC, Tham KC, Chew SWT, Lim X, Xu C. Real-Time Imaging of Dynamic Cell Reprogramming with Nanosensors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703440. [PMID: 29611333 DOI: 10.1002/smll.201703440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/22/2018] [Indexed: 06/08/2023]
Abstract
Cellular reprogramming, the process by which somatic cells regain pluripotency, is relevant in many disease modeling, therapeutic, and drug discovery applications. Molecular evaluation of reprogramming (e.g., polymerase chain reaction, immunostaining) is typically disruptive, and only provides snapshots of phenotypic traits. Gene reporter constructs facilitate live-cell evaluation but is labor intensive and may risk insertional mutagenesis during viral transfection. Herein, the utilization of a non-integrative nanosensor is demonstrated to visualize key reprogramming events in situ within live cells. Principally based on sustained intracellular release of encapsulated molecular probes, nanosensors successfully monitored mesenchymal-epithelial transition, pluripotency acquisition, and transdifferentiation events. Tracking the dynamic expression of four pivotal biomarkers (i.e., THY1, E-CADHERIN, OCT4, and GATA4 mRNA), nanosensor signal showed great agreement with polymerase chain reaction and gene reporter imaging (R2 > 0.9). Overall, such facile, versatile nanosensor enables real-time monitoring of low-frequency reprogramming events, thereby useful for high-throughput assessment, optimization, and biomarker-specific cell enrichment.
Collapse
Affiliation(s)
- Christian Wiraja
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
| | - David C Yeo
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
| | - Khek-Chian Tham
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Immunos, Singapore, 138648, Singapore
| | - Sharon W T Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
- NTU Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Xinhong Lim
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Immunos, Singapore, 138648, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 639798, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore, 169857, Singapore
| | - Chenjie Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
- NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
28
|
Shukla VC, Kuang TR, Senthilvelan A, Higuita-Castro N, Duarte-Sanmiguel S, Ghadiali SN, Gallego-Perez D. Lab-on-a-Chip Platforms for Biophysical Studies of Cancer with Single-Cell Resolution. Trends Biotechnol 2018; 36:549-561. [PMID: 29559164 DOI: 10.1016/j.tibtech.2018.02.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/14/2022]
Abstract
Recent cancer research has more strongly emphasized the biophysical aspects of tumor development, progression, and microenvironment. In addition to genetic modifications and mutations in cancer cells, it is now well accepted that the physical properties of cancer cells such as stiffness, electrical impedance, and refractive index vary with tumor progression and can identify a malignant phenotype. Moreover, cancer heterogeneity renders population-based characterization techniques inadequate, as individual cellular features are lost in the average. Hence, platforms for fast and accurate characterization of biophysical properties of cancer cells at the single-cell level are required. Here, we highlight some of the recent advances in the field of cancer biophysics and the development of lab-on-a-chip platforms for single-cell biophysical analyses of cancer cells.
Collapse
Affiliation(s)
- Vasudha C Shukla
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; These authors contributed equally to this work
| | - Tai-Rong Kuang
- The Key Laboratory of Polymer Processing Engineering of Ministry of Education, South China University of Technology, Guangzhou 510640, P.R. China; These authors contributed equally to this work.
| | - Abirami Senthilvelan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Natalia Higuita-Castro
- Department of Internal Medicine (Division of Pulmonary, Critical Care and Sleep Medicine), Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Silvia Duarte-Sanmiguel
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Human Sciences (Human Nutrition), College of Human Ecology, The Ohio State University, Columbus, OH 43210, USA
| | - Samir N Ghadiali
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Internal Medicine (Division of Pulmonary, Critical Care and Sleep Medicine), Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
29
|
Sadahiro H, Kang KD, Gibson JT, Minata M, Yu H, Shi J, Chhipa R, Chen Z, Lu S, Simoni Y, Furuta T, Sabit H, Zhang S, Bastola S, Yamaguchi S, Alsheikh H, Komarova S, Wang J, Kim SH, Hambardzumyan D, Lu X, Newell EW, DasGupta B, Nakada M, Lee LJ, Nabors B, Norian LA, Nakano I. Activation of the Receptor Tyrosine Kinase AXL Regulates the Immune Microenvironment in Glioblastoma. Cancer Res 2018. [PMID: 29531161 DOI: 10.1158/0008-5472.can-17-2433] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glioblastoma (GBM) is a lethal disease with no effective therapies available. We previously observed upregulation of the TAM (Tyro-3, Axl, and Mer) receptor tyrosine kinase family member AXL in mesenchymal GBM and showed that knockdown of AXL induced apoptosis of mesenchymal, but not proneural, glioma sphere cultures (GSC). In this study, we report that BGB324, a novel small molecule inhibitor of AXL, prolongs the survival of immunocompromised mice bearing GSC-derived mesenchymal GBM-like tumors. We show that protein S (PROS1), a known ligand of other TAM receptors, was secreted by tumor-associated macrophages/microglia and subsequently physically associated with and activated AXL in mesenchymal GSC. PROS1-driven phosphorylation of AXL (pAXL) induced NFκB activation in mesenchymal GSC, which was inhibited by BGB324 treatment. We also found that treatment of GSC-derived mouse GBM tumors with nivolumab, a blocking antibody against the immune checkpoint protein PD-1, increased intratumoral macrophages/microglia and activation of AXL. Combinatorial therapy with nivolumab plus BGB324 effectively prolonged the survival of mice bearing GBM tumors. Clinically, expression of AXL or PROS1 was associated with poor prognosis for patients with GBM. Our results suggest that the PROS1-AXL pathway regulates intrinsic mesenchymal signaling and the extrinsic immune microenvironment, contributing to the growth of aggressive GBM tumors.Significance: These findings suggest that development of combination treatments of AXL and immune checkpoint inhibitors may provide benefit to patients with GBM. Cancer Res; 78(11); 3002-13. ©2018 AACR.
Collapse
Affiliation(s)
- Hirokazu Sadahiro
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Neurosurgery, Yamaguchi University, Yamaguchi, Japan
| | - Kyung-Don Kang
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Justin T Gibson
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mutsuko Minata
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hai Yu
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Junfeng Shi
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Rishi Chhipa
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Zhihong Chen
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| | - Songjian Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yannick Simoni
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore, and the Nanyang Technological University School of Biological Sciences, Singapore
| | - Takuya Furuta
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Hemragul Sabit
- Department of Neurosurgery, Division of Neuroscience, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Suojun Zhang
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Soniya Bastola
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shinobu Yamaguchi
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hebaallah Alsheikh
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Svetlana Komarova
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jun Wang
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sung-Hak Kim
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Dolores Hambardzumyan
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Evan W Newell
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore, and the Nanyang Technological University School of Biological Sciences, Singapore
| | - Biplab DasGupta
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Mitsutoshi Nakada
- Department of Neurosurgery, Division of Neuroscience, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - L James Lee
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Burt Nabors
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lyse A Norian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama.,UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ichiro Nakano
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama. .,UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
30
|
Wang Y, Luo J, Liu J, Li X, Kong Z, Jin H, Cai X. Electrochemical integrated paper-based immunosensor modified with multi-walled carbon nanotubes nanocomposites for point-of-care testing of 17β-estradiol. Biosens Bioelectron 2018; 107:47-53. [PMID: 29428366 DOI: 10.1016/j.bios.2018.02.012] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 10/18/2022]
Abstract
17β-estradiol (17β-E2) plays a critical role in regulating reproduction in human, there is therefore an urgent need to detect it sensitively and precisely in a cost-effective and easy method. In this paper, a label-free integrated microfluidic paper-based analytical device was developed for highly sensitive electrochemical detection of 17β-E2. The microfluidic channel of the paper-based sensor was fabricated with wax printing and the three electrodes, including working, counter and reference electrode were screen-printed. Multi-walled carbon nanotubes (MWCNTs)/ thionine (THI)/ gold nanoparticles (AuNPs) Nano composites were synthesized and coated on screen-printed working electrode (SPWE) for the immobilization of anti-E2. In this electro-chemical system of paper-based immunoassay, THI molecules serving as an electrochemical mediator while MWCNTs and AuNPs, due to their excellent electrical conductivities, could accelerate electron transfer for the signal amplification. Experimental results revealed that the immunoassay is able to detect 17β-E2 as low as 10 pg mL-1, with a linear range from 0.01 to 100 ng mL-1. This microfluidic paper-based immunosensor would provide a new platform for low cost, sensitive, specific, and point-of-care diagnosis of 17β-E2.
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 10090, China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 10090, China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 10090, China
| | - Xinrong Li
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 10090, China
| | - Zhuang Kong
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 10090, China
| | - Hongyan Jin
- Obstetrics and Gynecology Department, First Hospital Peking University, Beijing 100034, China.
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 10090, China.
| |
Collapse
|
31
|
Lin JMG, Kang CC, Zhou Y, Huang H, Herr AE, Kumar S. Linking invasive motility to protein expression in single tumor cells. LAB ON A CHIP 2018; 18:371-384. [PMID: 29299576 PMCID: PMC5771853 DOI: 10.1039/c7lc01008g] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The invasion of malignant cells into tissue is a critical step in the progression of cancer. While it is increasingly appreciated that cells within a tumor differ in their invasive potential, it remains nearly unknown how these differences relate to cell-to-cell variations in protein expression. Here, we introduce a microfluidic platform that integrates measurements of invasive motility and protein expression for single cells, which we use to scrutinize human glioblastoma tumor-initiating cells (TICs). Our live-cell imaging microdevice is comprised of polyacrylamide microchannels that exhibit tissue-like stiffness and present chemokine gradients along each channel. Due to intrinsic differences in motility, cell subpopulations separate along the channel axis. The separated cells are then lysed in situ and each single-cell lysate is subjected to western blotting in the surrounding polyacrylamide matrix. We observe correlations between motility and Nestin and EphA2 expression. We identify protein-protein correlations within single TICs, which would be obscured with population-based assays. The integration of motility traits with single-cell protein analysis - on the same cell - offers a new means to identify druggable targets of invasive capacity.
Collapse
Affiliation(s)
- Jung-Ming G Lin
- UC-Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, CA 94720, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Zhao D, Wu M, Huang D, Liang Z, Wei Z, Li Z. Parametric optimization of electric field strength for cancer electrochemotherapy on a chip-based model. Theranostics 2018; 8:358-368. [PMID: 29290813 PMCID: PMC5743553 DOI: 10.7150/thno.21099] [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: 05/19/2017] [Accepted: 10/08/2017] [Indexed: 12/21/2022] Open
Abstract
Electrochemotherapy (ECT), as one of the very few available treatments for cutaneous and subcutaneous tumors when surgery and radiotherapy are no longer available, requires applying a proper electric field to the tumor to realize electroporation-mediated cytotoxic drug delivery. It is impossible to exhaust all possible electrical parameters on patients to realize the optimal tradeoff between tumor suppression and adverse effects. To address this issue, this study provides a feasible solution by developing a four-leaf micro-electrode chip (F-MEC) in which the electric field was specially designed by linear distribution to cover all possible electric field strengths for ECT. Methods: We developed a F-MEC that provides a linearly varied electric field and a capacity for in situ observation of cell status. By culturing tumor cells on the F-MEC surface and in situ monitoring the cell responses to ECT drugs, the optimal electric field strength for any given cell type could be rapidly and accurately calculated in a few, or even only one, simple assay. Results: Using this chip, we monitored MCF-7 and A315 cell responses to ECT and determined the optimum ECT voltage. More importantly, we successfully verified that the in vitro determined voltage coincided with the optimal value for in vivo ECT in mice. Conclusion: In this proof-of-concept study, the in vivo tumor suppression assays proved that the optimal parameters acquired from in vitro F-MEC assay could be used for in vivo ECT.
Collapse
|
33
|
Carbaugh DJ, Pandya SG, Wright JT, Kaya S, Rahman F. Combination photo and electron beam lithography with polymethyl methacrylate (PMMA) resist. NANOTECHNOLOGY 2017; 28:455301. [PMID: 28895558 DOI: 10.1088/1361-6528/aa8bd5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We describe techniques for performing photolithography and electron beam lithography in succession on the same resist-covered substrate. Larger openings are defined in the resist film through photolithography whereas smaller openings are defined through conventional electron beam lithography. The two processes are carried out one after the other and without an intermediate wet development step. At the conclusion of the two exposures, the resist film is developed once to reveal both large and small openings. Interestingly, these techniques are applicable to both positive and negative tone lithographies with both optical and electron beam exposure. Polymethyl methacrylate, by itself or mixed with a photocatalytic cross-linking agent, is used for this purpose. We demonstrate that such resists are sensitive to both ultraviolet and electron beam irradiation. All four possible combinations, consisting of optical and electron beam lithographies, carried out in positive and negative tone modes have been described. Demonstration grating structures have been shown and process conditions have been described for all four cases.
Collapse
Affiliation(s)
- Daniel J Carbaugh
- School of Electrical Engineering and Computer Science, Russ College of Engineering and Technology, Ohio University, Athens, OH 45701, United States of America
| | | | | | | | | |
Collapse
|
34
|
Xiao G, Song Y, Zhang S, Yang L, Xu S, Zhang Y, Xu H, Gao F, Li Z, Cai X. A high-sensitive nano-modified biosensor for dynamic monitoring of glutamate and neural spike covariation from rat cortex to hippocampal sub-regions. J Neurosci Methods 2017; 291:122-130. [DOI: 10.1016/j.jneumeth.2017.08.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 11/15/2022]
|
35
|
Khodadadian A, Hosseini K, Manzour-Ol-Ajdad A, Hedayati M, Kalantarinejad R, Heitzinger C. Optimal design of nanowire field-effect troponin sensors. Comput Biol Med 2017; 87:46-56. [PMID: 28550739 DOI: 10.1016/j.compbiomed.2017.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 05/09/2017] [Accepted: 05/09/2017] [Indexed: 10/19/2022]
Abstract
We propose a design strategy for affinity-based biosensors using nanowires for sensing and measuring biomarker concentration in biological samples. Such sensors have been shown to have superior properties compared to conventional biosensors in terms of LOD (limit of detection), response time, cost, and size. However, there are several parameters affecting the performance of such devices that must be determined. In order to solve the design problem, we have developed a comprehensive model based on stochastic transport equations that makes it possible to optimize the sensing behavior.
Collapse
Affiliation(s)
- Amirreza Khodadadian
- Institute for Analysis and Scientific Computing, Vienna University of Technology (TU Wien), Wiedner Hauptstraße 8-10, 1040 Vienna, Austria.
| | - Kiarash Hosseini
- Shezan Research and Innovation Centre, No. 25, Innovation 2 St., Pardis TechPark, Tehran, Iran
| | - Ali Manzour-Ol-Ajdad
- Shezan Research and Innovation Centre, No. 25, Innovation 2 St., Pardis TechPark, Tehran, Iran
| | - Marjan Hedayati
- Shezan Research and Innovation Centre, No. 25, Innovation 2 St., Pardis TechPark, Tehran, Iran
| | - Reza Kalantarinejad
- Shezan Research and Innovation Centre, No. 25, Innovation 2 St., Pardis TechPark, Tehran, Iran
| | - Clemens Heitzinger
- Institute for Analysis and Scientific Computing, Vienna University of Technology (TU Wien), Wiedner Hauptstraße 8-10, 1040 Vienna, Austria; School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
36
|
Zhao X, Liu Y, Zheng J, Liu X, Chen J, Liu L, Wang P, Xue Y. GAS5 suppresses malignancy of human glioma stem cells via a miR-196a-5p/FOXO1 feedback loop. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1605-1617. [PMID: 28666797 DOI: 10.1016/j.bbamcr.2017.06.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 06/19/2017] [Accepted: 06/23/2017] [Indexed: 02/06/2023]
Abstract
Glioma stem cells (GSCs) make up highly tumorigenic subpopulations within gliomas, and aberrant expression of GSC genes is a major underlying cause of glioma pathogenesis and treatment failure. The present study characterized the expression and function of long non-coding RNA growth arrest specific 5 (GAS5) in GSCs in order to elucidate the molecular mechanisms by which GAS5 contributes to glioma pathogenesis. We demonstrate that GAS5 suppresses GSC malignancy by binding to miR-196a-5p. miR-196a-5p, an onco-miRNA, stimulates GSC proliferation, migration, and invasion, in addition to reducing levels of apoptosis. miR-196a-5p specifically downregulates the expression of forkhead box protein O1 (FOXO1) by targeting its 3' untranslated region (3'-UTR). FOXO1 upregulates expression of phosphotyrosine interaction domain containing 1 (PID1), thereby inhibiting GSC tumorigenicity and growth. FOXO1 also upregulates migration and invasion inhibitory protein (MIIP), resulting in attenuation of migration and invasion activities. Interestingly, we also show that FOXO1 promotes GAS5 transcription, thus forminga positive feedback loop. These data provide insights into potential new pathways for GSC molecular therapy and suggest that GAS5 may be an efficacious target for glioma treatments.
Collapse
Affiliation(s)
- Xihe Zhao
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China; Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang 110004, People's Republic of China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China; Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang 110004, People's Republic of China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China; Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang 110004, People's Republic of China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Jiajia Chen
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Ping Wang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110122, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, People's Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110122, People's Republic of China.
| |
Collapse
|
37
|
Kuang T, Chang L, Peng X, Hu X, Gallego-Perez D. Molecular Beacon Nano-Sensors for Probing Living Cancer Cells. Trends Biotechnol 2017; 35:347-359. [DOI: 10.1016/j.tibtech.2016.09.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 01/30/2023]
|