1
|
Xu Z, Geng J, Liu X, Zhao Z, Suo D, Zhang S, Zhong J, Suo G. The extracellular matrix with a continuous gradient of SDF1 αguides the oriented migration of human umbilical cord mesenchymal stem cells. Biomed Mater 2024; 19:065019. [PMID: 39312941 DOI: 10.1088/1748-605x/ad7e91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 09/23/2024] [Indexed: 09/25/2024]
Abstract
The extracellular matrix (ECM) plays a crucial role in maintaining cell morphology and facilitating intercellular signal transmission within the human body. ECM has been extensively utilized for tissue injury repair. However, the consideration of factor gradients during ECM preparation has been limited. In this study, we developed a novel approach to generate sheet-like ECM with a continuous gradient of stromal cell-derived factor-1 (SDF1α). Briefly, we constructed fibroblasts to overexpress SDF1αfused with the collagen-binding domain (CBD-SDF1α), and cultured these cells on a slanted plate to establish a gradual density cell layer at the bottom surface. Subsequently, excess parental fibroblasts were evenly distributed on the plate laid flat to fill the room between cells. Following two weeks of culture, the monolayer cells were lyophilized to form a uniform ECM sheet possessing a continuous gradient of SDF1α. This engineered ECM material demonstrated its ability to guide oriented migration of human umbilical cord mesenchymal stem cells on the ECM sheet. Our simple yet effective method holds great potential for advancing research in regenerative medicine.
Collapse
Affiliation(s)
- Zhongjuan Xu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
- Guangdong Institute of Semiconductor Micro-Nano Manufacturing Technology, Foshan 528000, People's Republic of China
| | - Junsa Geng
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Xingzhi Liu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Zhe Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Dylan Suo
- Westlake High School, Austin, TX 78746, United States of America
| | - Sheng Zhang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Junjie Zhong
- Department of Neurosurgery, National Center for Neurological Disorders, National KeyLaboratory for Medical Neurobiology, Shanghai Key Laboratory of Brain Function and Regeneration, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University Huashan Hospital, Shanghai Medical College-Fudan University, Shanghai 200040, People's Republic of China
| | - Guangli Suo
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
- Division of Nanomaterials, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Nanchang 330200, People's Republic of China
| |
Collapse
|
2
|
Cramer TML, Tyagarajan SK. Protocol for the culturing of primary hippocampal mouse neurons for functional in vitro studies. STAR Protoc 2024; 5:102991. [PMID: 38607922 PMCID: PMC11017351 DOI: 10.1016/j.xpro.2024.102991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/22/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
Primary hippocampal cultures grown from genetically modified mice provide a simplified context to study molecular mechanisms underlying neuronal development, synaptogenesis, and synapse plasticity in vitro. Here, we describe a simple protocol for culturing hippocampal neurons from P0 to P2 mice and a strategy for inducing alterations in synaptic strength at inhibitory and excitatory synapses in vitro. We also describe approaches for immunofluorescent labeling, image acquisition, and quantification of synaptic proteins. For complete details on the use and execution of this protocol, please refer to Cramer et al.1.
Collapse
Affiliation(s)
- Teresa M L Cramer
- University of Zurich, Institute of Pharmacology and Toxicology, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Shiva K Tyagarajan
- University of Zurich, Institute of Pharmacology and Toxicology, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
3
|
Tariq U, Sarkar S, Malladi N, Kumar R, Bugga P, Chakraborty P, Banerjee SK. Knockdown of SCN5A alters metabolic-associated genes and aggravates hypertrophy in the cardiomyoblast. Mol Biol Rep 2024; 51:661. [PMID: 38758505 DOI: 10.1007/s11033-024-09594-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024]
Abstract
SCN5A mutations have been reported to cause various cardiomyopathies in humans. Most of the SCN5A mutations causes loss of function and thereby, alters the overall cellular function. Therefore, to understand the loss of SCN5A function in cardiomyocytes, we have knocked down the SCN5A gene (SCN5A-KD) in H9c2 cells and explored the cell phenotype and molecular behaviors in the presence and absence of isoproterenol (ISO), an adrenergic receptor agonist that induces cardiac hypertrophy. Expression of several genes related to hypertrophy, inflammation, fibrosis, and energy metabolism pathways were evaluated. It was found that the mRNA expression of hypertrophy-related gene, brain (B-type) natriuretic peptide (BNP) was significantly increased in SCN5A-KD cells as compared to 'control' H9c2 cells. There was a further increase in the mRNA expressions of BNP and βMHC in SCN5A-KD cells after ISO treatment compared to their respective controls. Pro-inflammatory cytokine, tumor necrosis factor-alpha expression was significantly increased in 'SCN5A-KD' H9c2 cells. Further, metabolism-related genes like glucose transporter type 4, cluster of differentiation 36, peroxisome proliferator-activated receptor alpha, and peroxisome proliferator-activated receptor-gamma were significantly elevated in the SCN5A-KD cells as compared to the control cells. Upregulation of these metabolic genes is associated with increased ATP production. The study revealed that SCN5A knock-down causes alteration of gene expression related to cardiac hypertrophy, inflammation, and energy metabolism pathways, which may promote cardiac remodelling and cardiomyopathy.
Collapse
Affiliation(s)
- Ubaid Tariq
- Non-communicable Disease Group, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Soumalya Sarkar
- Non-communicable Disease Group, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Navya Malladi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, 781101, India
| | - Roshan Kumar
- Non-communicable Disease Group, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Paramesha Bugga
- Non-communicable Disease Group, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Praloy Chakraborty
- Department of Cardiac Electrophysiology, Adult Cardiology, Toronto General Hospital, Toronto, ON, Canada
| | - Sanjay K Banerjee
- Non-communicable Disease Group, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India.
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, 781101, India.
| |
Collapse
|
4
|
Hao J, Li Z, Xie L, Yu B, Ma B, Yang Y, Ma X, Wang B, Zhou X. Syringaresinol promotes the recovery of spinal cord injury by inhibiting neuron apoptosis via activating the ubiquitination factor E4B/AKT Serine/Threonine kinase signal pathway. Brain Res 2024; 1824:148684. [PMID: 37992795 DOI: 10.1016/j.brainres.2023.148684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 11/24/2023]
Abstract
Spinal cord injury (SCI) is a serious traumatic disease with no effective treatment. This study aimed to explore the therapeutic effect of syringaresinol on SCI. First, the potential targets and associated signaling pathways of syringaresinol were predicted by bioinformatics analysis and molecular docking. Second, MTT was employed to evaluate cell proliferation rate, Western blot was performed to detect protein expression, RT-qPCR was conducted to detect mRNA expression levels, flow cytometry and 5-ethynyl-2'-deoxyuridine (EDU) staining were used to determine cell apoptosis, and immunofluorescence and immunohistochemistry were used to estimate the expression of RNA binding fox-1 homolog 3 and clipped caspase 3. Basso-Beattie-Bresnahan scores and inclined plate tests were conducted to analyze hindlimb locomotor function. Results showed that syringaresinol could inhibit the apoptosis of glutamate-treated SHSY5Y cells by upregulating the expression of ubiquitination factor E4B (UBE4B) and activating the AKT serine/threonine kinase (AKT) signaling pathway. This effect can be rescued by UBE4B knockdown or AKT pathway inhibition. Syringaresinol remarkably improved locomotor function and increased neuronal survival in SCI rats. Our results suggested that syringaresinol could promote locomotor functional recovery by reducing neuronal apoptosis by activating the UBE4B/AKT signaling pathway.
Collapse
Affiliation(s)
- Jian Hao
- Orthopedic Department, The 2(nd) Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Zhenhan Li
- School of Clinical, Wannan Medical College, Wuhu, China
| | - Li Xie
- Department of Anesthesiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Bingbing Yu
- Department of Orthopedics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Boyuan Ma
- Orthopedic Department, The 2(nd) Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yubiao Yang
- Orthopedic Department, The 2(nd) Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuchen Ma
- Orthopedic Department, The 2(nd) Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bitao Wang
- Orthopedic Department, The 2(nd) Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xianhu Zhou
- Orthopedic Department, The 2(nd) Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Mondal T, Gaur H, Wamba BEN, Michalak AG, Stout C, Watson MR, Aleixo SL, Singh A, Condello S, Faller R, Leiserowitz GS, Bhatnagar S, Tushir-Singh J. Characterizing the regulatory Fas (CD95) epitope critical for agonist antibody targeting and CAR-T bystander function in ovarian cancer. Cell Death Differ 2023; 30:2408-2431. [PMID: 37838774 PMCID: PMC10657439 DOI: 10.1038/s41418-023-01229-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/14/2023] [Accepted: 09/28/2023] [Indexed: 10/16/2023] Open
Abstract
Receptor clustering is the most critical step to activate extrinsic apoptosis by death receptors belonging to the TNF superfamily. Although clinically unsuccessful, using agonist antibodies, the death receptors-5 remains extensively studied from a cancer therapeutics perspective. However, despite its regulatory role and elevated function in ovarian and other solid tumors, another tumor-enriched death receptor called Fas (CD95) remained undervalued in cancer immunotherapy until recently, when its role in off-target tumor killing by CAR-T therapies was imperative. By comprehensively analyzing structure studies in the context of the binding epitope of FasL and various preclinical Fas agonist antibodies, we characterize a highly significant patch of positively charged residue epitope (PPCR) in its cysteine-rich domain 2 of Fas. PPCR engagement is indispensable for superior Fas agonist signaling and CAR-T bystander function in ovarian tumor models. A single-point mutation in FasL or Fas that interferes with the PPCR engagement inhibited apoptotic signaling in tumor cells and T cells. Furthermore, considering that clinical and immunological features of the autoimmune lymphoproliferative syndrome (ALPS) are directly attributed to homozygous mutations in FasL, we reveal differential mechanistic details of FasL/Fas clustering at the PPCR interface compared to described ALPS mutations. As Fas-mediated bystander killing remains vital to the success of CAR-T therapies in tumors, our findings highlight the therapeutic analytical design for potentially effective Fas-targeting strategies using death agonism to improve cancer immunotherapy in ovarian and other solid tumors.
Collapse
Affiliation(s)
- Tanmoy Mondal
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Himanshu Gaur
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Brice E N Wamba
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Abby Grace Michalak
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- Undergraduate Research Program Volunteers, University of California Davis, Davis, CA, USA
| | - Camryn Stout
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- Undergraduate Research Program Volunteers, University of California Davis, Davis, CA, USA
| | - Matthew R Watson
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- Undergraduate Research Program Volunteers, University of California Davis, Davis, CA, USA
| | - Sophia L Aleixo
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- Undergraduate Research Program Volunteers, University of California Davis, Davis, CA, USA
| | - Arjun Singh
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Salvatore Condello
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roland Faller
- Department of Chemical Engineering, University of California Davis, Davis, CA, USA
| | - Gary Scott Leiserowitz
- Department of Obstetrics and Gynecology, UC Davis School of Medicine, Sacramento, CA, USA
- UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Sanchita Bhatnagar
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Jogender Tushir-Singh
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA.
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA.
- UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA.
- Ovarian Cancer Academy Early Career Investigator at UC Davis, Davis, CA, USA.
| |
Collapse
|
6
|
Chiu CH. CRISPR/Cas9 genetic screens in hepatocellular carcinoma gene discovery. CURRENT RESEARCH IN BIOTECHNOLOGY 2023; 5:100127. [DOI: 10.1016/j.crbiot.2023.100127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
7
|
Shivange G, Mondal T, Lyerly E, Bhatnagar S, Landen CN, Reddy S, Kim J, Doan B, Riddle P, Tushir-Singh J. A patch of positively charged residues regulates the efficacy of clinical DR5 antibodies in solid tumors. Cell Rep 2021; 37:109953. [PMID: 34731630 PMCID: PMC8720280 DOI: 10.1016/j.celrep.2021.109953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/19/2021] [Accepted: 10/15/2021] [Indexed: 11/16/2022] Open
Abstract
Receptor clustering is the first and critical step to activate apoptosis by death receptor-5 (DR5). The recent discovery of the autoinhibitory DR5 ectodomain has challenged the long-standing view of its mechanistic activation by the natural ligand Apo2L. Because the autoinhibitory residues have remained unknown, here we characterize a crucial patch of positively charged residues (PPCR) in the highly variable domain of DR5. The PPCR electrostatically separates DR5 receptors to autoinhibit their clustering in the absence of ligand and antibody binding. Mutational substitution and antibody-mediated PPCR interference resulted in increased apoptotic cytotoxic function. A dually specific antibody that enables sustained tampering with PPCR function exceptionally enhanced DR5 clustering and apoptotic activation and distinctively improved the survival of animals bearing aggressive metastatic and recurrent tumors, whereas clinically tested DR5 antibodies without PPCR blockade function were largely ineffective. Our study provides mechanistic insights into DR5 activation and a therapeutic analytical design for potential clinical success.
Collapse
MESH Headings
- A549 Cells
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/metabolism
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody Specificity
- Antineoplastic Agents, Immunological/immunology
- Antineoplastic Agents, Immunological/metabolism
- Antineoplastic Agents, Immunological/pharmacology
- Apoptosis/drug effects
- Epitopes
- Humans
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Neoplasms/drug therapy
- Neoplasms/immunology
- Neoplasms/metabolism
- Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors
- Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- Signal Transduction
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Gururaj Shivange
- Laboratory of Novel Biologics, Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville VA 22908, USA
| | - Tanmoy Mondal
- Laboratory of Novel Biologics, Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA; Department of Medical Microbiology and Immunology, University of California School of Medicine, University of California, Davis, Davis, CA 95616, USA; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville VA 22908, USA
| | - Evan Lyerly
- Laboratory of Novel Biologics, Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA; Undergraduate Research Program Volunteers, University of Virginia, Charlottesville VA; Blavatnik Institute, Harvard Medical School, Boston MA
| | - Sanchita Bhatnagar
- Department of Medical Microbiology and Immunology, University of California School of Medicine, University of California, Davis, Davis, CA 95616, USA; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville VA 22908, USA
| | | | - Shivani Reddy
- Laboratory of Novel Biologics, Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA; Undergraduate Research Program Volunteers, University of Virginia, Charlottesville VA
| | - Jonathan Kim
- Laboratory of Novel Biologics, Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA; Undergraduate Research Program Volunteers, University of Virginia, Charlottesville VA
| | - Britney Doan
- Laboratory of Novel Biologics, Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA; Undergraduate Research Program Volunteers, University of Virginia, Charlottesville VA
| | - Paula Riddle
- Laboratory of Novel Biologics, Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA; Undergraduate Research Program Volunteers, University of Virginia, Charlottesville VA
| | - Jogender Tushir-Singh
- Laboratory of Novel Biologics, Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA; Department of Medical Microbiology and Immunology, University of California School of Medicine, University of California, Davis, Davis, CA 95616, USA; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville VA 22908, USA; University of Virginia Comprehensive Cancer Center, Charlottesville VA; UC Davis Comprehensive Cancer Center, University of California School of Medicine, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
8
|
Mondal T, Shivange GN, Tihagam RGT, Lyerly E, Battista M, Talwar D, Mosavian R, Urbanek K, Rashid NS, Harrell JC, Bos PD, Stelow EB, Stack MS, Bhatnagar S, Tushir‐Singh J. Unexpected PD-L1 immune evasion mechanism in TNBC, ovarian, and other solid tumors by DR5 agonist antibodies. EMBO Mol Med 2021; 13:e12716. [PMID: 33587338 PMCID: PMC7933954 DOI: 10.15252/emmm.202012716] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Lack of effective immune infiltration represents a significant barrier to immunotherapy in solid tumors. Thus, solid tumor-enriched death receptor-5 (DR5) activating antibodies, which generates tumor debulking by extrinsic apoptotic cytotoxicity, remains a crucial alternate therapeutic strategy. Over past few decades, many DR5 antibodies moved to clinical trials after successfully controlling tumors in immunodeficient tumor xenografts. However, DR5 antibodies failed to significantly improve survival in phase-II trials, leading in efforts to generate second generation of DR5 agonists to supersize apoptotic cytotoxicity in tumors. Here we have discovered that clinical DR5 antibodies activate an unexpected immunosuppressive PD-L1 stabilization pathway, which potentially had contributed to their limited success in clinics. The DR5 agonist stimulated caspase-8 signaling not only activates ROCK1 but also undermines proteasome function, both of which contributes to increased PD-L1 stability on tumor cell surface. Targeting DR5-ROCK1-PD-L1 axis markedly increases immune effector T-cell function, promotes tumor regression, and improves overall survival in animal models. These insights have identified a potential clinically viable combinatorial strategy to revive solid cancer immunotherapy using death receptor agonism.
Collapse
Affiliation(s)
- Tanmoy Mondal
- Laboratory of Novel BiologicsUniversity of VirginiaCharlottesvilleVAUSA
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
| | - Gururaj N Shivange
- Laboratory of Novel BiologicsUniversity of VirginiaCharlottesvilleVAUSA
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
| | - Rachisan GT Tihagam
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
| | - Evan Lyerly
- Laboratory of Novel BiologicsUniversity of VirginiaCharlottesvilleVAUSA
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
- Undergraduate Research ProgramUniversity of VirginiaCharlottesvilleVAUSA
| | - Michael Battista
- Laboratory of Novel BiologicsUniversity of VirginiaCharlottesvilleVAUSA
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
- Undergraduate Research ProgramUniversity of VirginiaCharlottesvilleVAUSA
| | - Divpriya Talwar
- Laboratory of Novel BiologicsUniversity of VirginiaCharlottesvilleVAUSA
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
- Undergraduate Research ProgramUniversity of VirginiaCharlottesvilleVAUSA
| | - Roxanna Mosavian
- Laboratory of Novel BiologicsUniversity of VirginiaCharlottesvilleVAUSA
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
- Undergraduate Research ProgramUniversity of VirginiaCharlottesvilleVAUSA
| | - Karol Urbanek
- Laboratory of Novel BiologicsUniversity of VirginiaCharlottesvilleVAUSA
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
| | | | - J Chuck Harrell
- Department of PathologyMassey Cancer Center, VCURichmondVAUSA
| | - Paula D Bos
- Department of PathologyMassey Cancer Center, VCURichmondVAUSA
| | - Edward B Stelow
- Department of PathologyUniversity of VirginiaCharlottesvilleVAUSA
| | - M Sharon Stack
- Harper Cancer Research InstituteUniversity of Notre DameNotre DameINUSA
| | - Sanchita Bhatnagar
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
- University of Virginia Cancer Center and Medical SchoolCharlottesvilleVAUSA
| | - Jogender Tushir‐Singh
- Laboratory of Novel BiologicsUniversity of VirginiaCharlottesvilleVAUSA
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVAUSA
- University of Virginia Cancer Center and Medical SchoolCharlottesvilleVAUSA
- DoD Ovarian Cancer Academy Early Career InvestigatorCharlottesvilleVAUSA
| |
Collapse
|
9
|
Concise review on optimized methods in production and transduction of lentiviral vectors in order to facilitate immunotherapy and gene therapy. Biomed Pharmacother 2020; 128:110276. [PMID: 32502836 DOI: 10.1016/j.biopha.2020.110276] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/10/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Lentiviral vectors (LVs) have provided an efficient way to integrate our gene of interest into eukaryote cells. Human immunodeficiency virus (HIV)-derived LVs have been vastly studied to become an invaluable asset in gene delivery. This abled LVs to be used in both research laboratories and gene therapy. Pseudotyping HIV-1 based LVs, abled it to transduce different types of cells, especially hematopoietic stem cells. A wide range of tropism, plus to the ability to integrate genes into target cells, made LVs an armamentarium in gene therapy. The third and fourth generations of self-inactivating LVs are being used to achieve safe gene therapy. Not only advanced methods enabled the clinical-grade LV production on a large scale, but also considerably heightened transduction efficiency. One of which is microfluidic systems that revolutionized gene delivery approaches. Since gene therapy using LVs attracted lots of attention to itself, we provided a brief review of LV structure and life-cycle along with methods for improving both LV production and transduction. Also, we mentioned some of their utilization in immunotherapy and gene therapy.
Collapse
|
10
|
He WY, Zhang B, Zhao WC, He J, Zhang L, Xiong QM, Wang J, Wang HB. Contributions of mTOR Activation-Mediated Upregulation of Synapsin II and Neurite Outgrowth to Hyperalgesia in STZ-Induced Diabetic Rats. ACS Chem Neurosci 2019; 10:2385-2396. [PMID: 30785256 DOI: 10.1021/acschemneuro.8b00680] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Painful diabetic neuropathy (PDN) is among the common complications in diabetes mellitus (DM), with its underlying mechanisms largely unknown. Synapsin II is primarily expressed in the spinal dorsal horn, and its upregulation mediates a superfluous release of glutamate and a deficiency of GABAergic interneuron synaptic transmission, which is directly implicated in the facilitation of pain signals in the hyperalgesic nociceptive response. Recently, synapsin II has been revealed to be associated with the modulation of neurite outgrowth, whereas the process of this neuronal structural neuroplasticity following neuronal hyperexcitability still remains unclear. In this study, we found that under conditions of elevated glucose, TNF-α induced the activation of mTOR, mediating the upregulation of synapsin II and neurite outgrowth in dorsal horn neurons. In vivo, we demonstrated that mTOR and synapsin II were upregulated and coexpressed in the spinal dorsal horn neurons in rats with streptozotocin (STZ)-induced diabetes. Furthermore, the intrathecal administration of the mTOR inhibitor rapamycin or synapsin II shRNA significantly diminished the expression of synapsin II, effectively mitigating hyperalgesia in PDN rats. We are the first to discover that in STZ-induced diabetic rats the activation of mTOR mediates the upregulation of synapsin II and neurite outgrowth, both contributing to hyperalgesia. These findings may benefit the clinical therapy of PDN by provision of a novel target.
Collapse
Affiliation(s)
- Wan-you He
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Bin Zhang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Wei-cheng Zhao
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Jian He
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Lei Zhang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Qing-ming Xiong
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Jing Wang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Han-bing Wang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| |
Collapse
|
11
|
Riemann D, Petkova A, Dresbach T, Wallrafen R. An Optical Assay for Synaptic Vesicle Recycling in Cultured Neurons Overexpressing Presynaptic Proteins. J Vis Exp 2018:58043. [PMID: 30010661 PMCID: PMC6101998 DOI: 10.3791/58043] [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: 10/31/2022] Open
Abstract
At active presynaptic nerve terminals, synaptic vesicles undergo cycles of exo- and endocytosis. During recycling, the luminal domains of SV transmembrane proteins become exposed at the cell surface. One of these proteins is Synaptotagmin-1 (Syt1). An antibody directed against the luminal domain of Syt1, once added to the culture medium, is taken up during the exo-endocytotic cycle. This uptake is proportional to the amount of SV recycling and can be quantified through immunofluorescence. Here, we combine Syt1 antibody uptake with double transfection of cultured hippocampal neurons. This allows us to (1) localize presynaptic sites based on expression of recombinant presynaptic marker Synaptophysin, (2) determine their functionality using Syt1 uptake, and (3) characterize the targeting and effects of a protein of interest, GFP-Rogdi.
Collapse
Affiliation(s)
- Donatus Riemann
- Institute for Anatomy and Embryology, University Medical Centre Göttingen
| | - Andoniya Petkova
- Institute for Anatomy and Embryology, University Medical Centre Göttingen
| | - Thomas Dresbach
- Institute for Anatomy and Embryology, University Medical Centre Göttingen;
| | - Rebecca Wallrafen
- Institute for Anatomy and Embryology, University Medical Centre Göttingen
| |
Collapse
|
12
|
Shi T, Li L, Zhou G, Wang C, Chen X, Zhang R, Xu J, Lu X, Jiang H, Chen J. Toll-like receptor 5 agonist CBLB502 induces radioprotective effects in vitro. Acta Biochim Biophys Sin (Shanghai) 2018; 49:487-495. [PMID: 28407032 DOI: 10.1093/abbs/gmx034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Indexed: 12/31/2022] Open
Abstract
CBLB502 derived from Salmonella flagellin is a novel agonist of Toll-like receptor 5 (TLR5). It has been shown that CBLB502 can exert high radioprotective efficacy on mice and primates from both GI and hematopoietic syndromes during whole-body irradiation with low toxicity and immunogenicity. However, no effective system has been used to investigate the protective effect of CBLB502 against irradiation and the related mechanism in vitro. In this study, we investigated the radioprotective properties of CBLB502 in HEK293-N-T cells constitutively expressing human TLR5 and NF-κB-dependent luciferase. HEK293-N-T cells were treated with different doses of CBLB502 prior to 60Co-γ ray irradiation. After irradiation, cell viability was real-time measured for 4 days by using the real-time cell analysis system. We found that CBLB502 was capable of efficiently maintaining the survival rate of irradiated HEK293-N-T cells. Then apoptotic cell death and cell cycle were detected by flow cytometry. The results showed that CBLB502 pre-treatment could reduce the apoptosis and promote the recovery of irradiated HEK293-N-T cells from G2-phase arrest in a dose-dependent manner. Our data indicated that CBLB502 has a direct radioprotective effect in vitro via anti-apoptosis and promotes cell cycle recovery. The method developed here could be an effective in vitro system to screen other TLR5-target radioprotectants like CBLB502.
Collapse
Affiliation(s)
- Tong Shi
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | | | - Guochao Zhou
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Chen Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Xuejun Chen
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Ruihua Zhang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Jianfu Xu
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Xiaojing Lu
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Hui Jiang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Jisheng Chen
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| |
Collapse
|
13
|
Wang J, Zhao Y, Xin M, Pan L, Wang L, Yang K. Effectiveness of lentivirus-mediated RNA interference targeting mouse tumor necrosis factor α in vitro and in vivo. Exp Ther Med 2018; 15:2134-2139. [PMID: 29434816 PMCID: PMC5776641 DOI: 10.3892/etm.2017.5609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 09/07/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to identify the effectiveness of lentivirus-mediated RNA interference (RNAi) targeting mouse tumor necrosis factor-α (TNF-α). RNAi lentivirus was used in vitro to transfect RAW264.7 cells, and the expression of TNF-α, interleukin (IL)-1β and IL-6 mRNAs and TNF-α protein in RAW264.7 cells was measured by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay, respectively. In vivo, mice with collagen-induced arthritis (CIA) were injected intravenously with RNAi lentivirus, and CIA arthritis scores and the serum levels of TNF-α were detected. Additionally, joint tissues were subjected to pathological examination. In the cells, the expression level of TNF-α mRNA in the RNAi lentivirus group was 0.29±0.02, which was significantly lower than that of the lentivirus negative control (0.93±0.01; t=25.4, P<0.001). In the mice, the serum TNF-α level in the RNAi lentivirus group was 249.25±11.22 ng/ml, which was significantly lower than that of the negative control group (381.86±6.28 ng/ml; P<0.05). However, no difference in IL-1α and IL-6 mRNA levels was identified among the groups (t=1.00, P=0.37; t=1.22, P=0.29). The CIA arthritis score in the RNAi lentivirus group was significantly reduced compared with those in the control and negative control groups (P<0.05). Furthermore, the arthritis scores in the RNAi lentivirus and positive control groups continued to decrease for ≥2 weeks, and the serum TNF-α levels in the RNAi lentivirus and positive control groups were 31.58±2.18 and 35.21±2.25 pg/ml, which were significantly lower than those in the negative control group (46.62±3.02 pg/ml; P<0.05). Thus, targeting of the TNF-α gene in mice via lentivirus-mediated RNAi in vitro and in vivo achieved TNF-α gene downregulation, which indicates that lentivirus-mediated RNA interference may be an effective form of gene therapy against rheumatoid arthritis.
Collapse
Affiliation(s)
- Jibo Wang
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yingjie Zhao
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Miaomiao Xin
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Lin Pan
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Liqin Wang
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Kun Yang
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
14
|
Wang J, Liang H, Zhao Y, Liu X, Yang K, Sui A. Construction and identification of an RNA interference lentiviral vector targeting the mouse TNF-α gene. Exp Ther Med 2015; 10:2283-2288. [PMID: 26668629 DOI: 10.3892/etm.2015.2813] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 08/05/2015] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to construct RNA interference (RNAi) lentiviral vector particles targeting the mouse tumor necrosis factor-α (TNF-α) gene. Three types of small interfering RNA (siRNA) targeting the mouse TNF-α gene were designed, synthesized and transfected into RAW264.7 cells. Screening was performed to identify the siRNA sequence exhibiting the highest inhibition efficiency; based on this, recombinant lentiviral plasmids were constructed and co-transfected into 293T cells with packaging plasmids for the production of lentiviral particles. The screening results showed that the TNF-α mRNA expression levels of the three siRNA groups were significantly lower than those of the negative control group, with the highest inhibition rate in the siRNA2 group (83.09%). Similarly, the expression levels of TNF-α protein in the three siRNA groups were significantly lower than those of the negative control group, and the highest inhibition rate was found in the siRNA2 group (51.16%). The mRNA expression of interleukin (IL)-1β and IL-6 showed no significant difference among the siRNA groups and the negative control. The recombinant lentiviral shuttle plasmid was constructed, and electrophoresis revealed the polymerase chain reaction product to be 343 bp, while that of the empty vector was 306 bp; DNA sequencing showed partial insertion. The virus titer was calculated to be 2×106 TU/µl. In conclusion, RNAi lentiviral vector particles targeting the mouse TNF-α gene were successfully obtained in the present study. This method may be used to produce lentiviral vector for the in vivo study of RNAi gene therapy targeting TNF-α.
Collapse
Affiliation(s)
- Jibo Wang
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Hongda Liang
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yingjie Zhao
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiangping Liu
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Kun Yang
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Aihua Sui
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
15
|
Wollebo HS, Bellizzi A, Kaminski R, Hu W, White MK, Khalili K. CRISPR/Cas9 System as an Agent for Eliminating Polyomavirus JC Infection. PLoS One 2015; 10:e0136046. [PMID: 26360417 PMCID: PMC4567079 DOI: 10.1371/journal.pone.0136046] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/30/2015] [Indexed: 12/21/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a fatal demyelinating disease of the central nervous system (CNS) caused by reactivation of the human polyomavirus JCV gene expression and its replication in oligodendrocytes, the myelin producing cells in the brain. Once a rare disease seen in patients with lymphotproliferative and myeloproliferative disorders, PML has been seen more frequently in HIV-1 positive/AIDS patients as well as patients undergoing immunomodulatory therapy due for autoimmune disorders including multiple sclerosis, rheumatoid arthritis, and others. As of now there is no cure for PML and in most cases disease progression leads to death within two years. Similar to other polyomaviruses, the JCV genome is small circular double stranded DNA that includes coding sequences for the viral early protein, T-antigen, which is critical for directing viral reactivation and lytic infection. Here, we employ a newly developed gene editing strategy, CRISPR/Cas9, to introduce mutations in the viral genome and, by inactivating the gene encoding T-antigen, inhibit viral replication. We first used bioinformatics screening and identified several potential targets within the JCV T-antigen gene that can serve as sites for the creation of guide RNAs (gRNAs) for positioning the Cas9 nuclease on the designated area of the viral genome for editing. Results from a series of integrated genetic and functional studies showed that transient or conditional expression of Cas9 and gRNAs specifically targets the DNA sequences corresponding to the N-terminal region of T-antigen, and by introducing mutation, interferes with expression and function of of the viral protein, hence suppressing viral replication in permissive cells. Results from SURVEYOR assay revealed no off-target effects of the JCV-specific CRISPR/Cas9 editing apparatus. These observations provide the first evidence for the employment of a gene editing strategy as a promising tool for the elimination of the JCV genome and a potential cure for PML.
Collapse
MESH Headings
- Antigens, Viral, Tumor/genetics
- Base Sequence
- CRISPR-Cas Systems
- Cell Line, Tumor
- Gene Expression
- Gene Knockdown Techniques
- Gene Targeting
- Genetic Therapy/methods
- Genome, Viral
- Humans
- JC Virus/genetics
- Leukoencephalopathy, Progressive Multifocal/therapy
- Leukoencephalopathy, Progressive Multifocal/virology
- Molecular Sequence Data
- Mutation
- Promoter Regions, Genetic
- RNA Editing
- RNA, Guide, CRISPR-Cas Systems/chemistry
- RNA, Guide, CRISPR-Cas Systems/genetics
- Sequence Alignment
- Virus Replication
Collapse
Affiliation(s)
- Hassen S. Wollebo
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Anna Bellizzi
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Rafal Kaminski
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Wenhui Hu
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Martyn K. White
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| | - Kamel Khalili
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA, 19122, United States of America
| |
Collapse
|