1
|
Nabi B, Rehman S, Pottoo FH, Baboota S, Ali J. Dissecting the Therapeutic Relevance of Gene Therapy in NeuroAIDS: An Evolving Epidemic. Curr Gene Ther 2021; 20:174-183. [PMID: 32538727 DOI: 10.2174/1566523220666200615151805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/09/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
Abstract
NeuroAIDS, a disease incorporating both infectious and neurodegenerative pathways, is still a formidable challenge for the researchers to deal with. The primary concern for the treatment of neuroAIDS still remains the inaccessibility of the viral reservoir, making it indispensable for novel techniques to be continuously innovated. Since the brain serves as a reservoir for viral replication, it is pragmatic and a prerequisite to overcome the related barriers in order to improve the drug delivery to the brain. The current treatment ideology is based on the combinatorial approach of a mocktail of antiretroviral drugs. However, complete eradication of the disease could not be achieved. Thereby the arena of gene-based cellular delivery is trending and has created a niche for itself in the present scenario. To establish the supremacy of gene delivery, it is advisable to have a better understanding of the molecular mechanism involved in the due process. The mechanism associated with the activity of the anti-HIV gene lies in their intrinsic property to impart resistance to the HIV infection by targeting the viral entry channels. This review principally emphasizes on different types of gene therapies explored so far for the management of AIDS and its associated neurological conditions. Therefore it could rightly be said that we are at the crossroad where the need of the hour is to develop novel strategies for curbing AIDS and its associated neurological conditions.
Collapse
Affiliation(s)
- Bushra Nabi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Saleha Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam, 31441, Saudi Arabia
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
2
|
Mançanares ACF, Cabezas J, Manríquez J, de Oliveira VC, Wong Alvaro YS, Rojas D, Navarrete Aguirre F, Rodriguez-Alvarez L, Castro FO. Edition of Prostaglandin E2 Receptors EP2 and EP4 by CRISPR/Cas9 Technology in Equine Adipose Mesenchymal Stem Cells. Animals (Basel) 2020; 10:E1078. [PMID: 32585798 PMCID: PMC7341266 DOI: 10.3390/ani10061078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 01/14/2023] Open
Abstract
In mesenchymal stem cells (MSCs), it has been reported that prostaglandin E2 (PGE2) stimulation of EP2 and EP4 receptors triggers processes such as migration, self-renewal, survival, and proliferation, and their activation is involved in homing. The aim of this work was to establish a genetically modified adipose (aMSC) model in which receptor genes EP2 and EP4 were edited separately using the CRISPR/Cas9 system. After edition, the genes were evaluated as to if the expression of MSC surface markers was affected, as well as the migration capacity in vitro of the generated cells. Adipose MSCs were obtained from Chilean breed horses and cultured in DMEM High Glucose with 10% fetal bovine serum (FBS). sgRNA were cloned into a linearized LentiCRISPRv2GFP vector and transfected into HEK293FT cells for producing viral particles that were used to transduce aMSCs. GFP-expressing cells were separated by sorting to obtain individual clones. Genomic DNA was amplified, and the site-directed mutation frequency was assessed by T7E1, followed by Sanger sequencing. We selected 11 clones of EP2 and 10 clones of EP4, and by Sanger sequencing we confirmed 1 clone knock-out to aMSC/EP2 and one heterozygous mutant clone of aMSC/EP4. Both edited cells had decreased expression of EP2 and EP4 receptors when compared to the wild type, and the edition of EP2 and EP4 did not affect the expression of MSC surface markers, showing the same pattern in filling the scratch. We can conclude that the edition of these receptors in aMSCs does not affect their surface marker phenotype and migration ability when compared to wild-type cells.
Collapse
Affiliation(s)
- Ana Carolina Furlanetto Mançanares
- Department of Animal Science, Faculty of Veterinary Science, Universidad de Concepción, Campus Chillan, Chillán 3780000, Chile; (J.C.); (J.M.); (Y.S.W.A.); (F.N.A.); (L.R.-A.)
| | - Joel Cabezas
- Department of Animal Science, Faculty of Veterinary Science, Universidad de Concepción, Campus Chillan, Chillán 3780000, Chile; (J.C.); (J.M.); (Y.S.W.A.); (F.N.A.); (L.R.-A.)
| | - José Manríquez
- Department of Animal Science, Faculty of Veterinary Science, Universidad de Concepción, Campus Chillan, Chillán 3780000, Chile; (J.C.); (J.M.); (Y.S.W.A.); (F.N.A.); (L.R.-A.)
| | - Vanessa Cristina de Oliveira
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo 13630-000, Brazil;
| | - Yat Sen Wong Alvaro
- Department of Animal Science, Faculty of Veterinary Science, Universidad de Concepción, Campus Chillan, Chillán 3780000, Chile; (J.C.); (J.M.); (Y.S.W.A.); (F.N.A.); (L.R.-A.)
| | - Daniela Rojas
- Department of Animal Pathology, Faculty of Veterinary Sciences, Universidad de Concepción, Campus Chillan, Chillán 3780000, Chile;
| | - Felipe Navarrete Aguirre
- Department of Animal Science, Faculty of Veterinary Science, Universidad de Concepción, Campus Chillan, Chillán 3780000, Chile; (J.C.); (J.M.); (Y.S.W.A.); (F.N.A.); (L.R.-A.)
| | - Lleretny Rodriguez-Alvarez
- Department of Animal Science, Faculty of Veterinary Science, Universidad de Concepción, Campus Chillan, Chillán 3780000, Chile; (J.C.); (J.M.); (Y.S.W.A.); (F.N.A.); (L.R.-A.)
| | - Fidel Ovidio Castro
- Department of Animal Science, Faculty of Veterinary Science, Universidad de Concepción, Campus Chillan, Chillán 3780000, Chile; (J.C.); (J.M.); (Y.S.W.A.); (F.N.A.); (L.R.-A.)
| |
Collapse
|
3
|
Liu D, Ndongwe TP, Puray-Chavez M, Casey MC, Izumi T, Pathak VK, Tedbury PR, Sarafianos SG. Effect of P-body component Mov10 on HCV virus production and infectivity. FASEB J 2020; 34:9433-9449. [PMID: 32496609 DOI: 10.1096/fj.201800641r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 03/28/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022]
Abstract
Mov10 is a processing body (P-body) protein and an interferon-stimulated gene that can affect replication of retroviruses, hepatitis B virus, and hepatitis C virus (HCV). The mechanism of HCV inhibition by Mov10 is unknown. Here, we investigate the effect of Mov10 on HCV infection and determine the virus life cycle steps affected by changes in Mov10 overexpression. Mov10 overexpression suppresses HCV RNA in both infectious virus and subgenomic replicon systems. Additionally, Mov10 overexpression decreases the infectivity of released virus, unlike control P-body protein DCP1a that has no effect on HCV RNA production or infectivity of progeny virus. Confocal imaging of uninfected cells shows endogenous Mov10 localized at P-bodies. However, in HCV-infected cells, Mov10 localizes in circular structures surrounding cytoplasmic lipid droplets with NS5A and core protein. Mutagenesis experiments show that the RNA binding activity of Mov10 is required for HCV inhibition, while its P-body localization, helicase, and ATP-binding functions are not required. Unexpectedly, endogenous Mov10 promotes HCV replication, as CRISPR-Cas9-based Mov10 depletion decreases HCV replication and infection levels. Our data reveal an important and complex role for Mov10 in HCV replication, which can be perturbed by excess or insufficient Mov10.
Collapse
Affiliation(s)
- Dandan Liu
- Christopher Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Tanyaradzwa P Ndongwe
- Christopher Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Maritza Puray-Chavez
- Christopher Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Mary C Casey
- Christopher Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Taisuke Izumi
- Viral Mutation Section, HIV Dynamics and Replication Program, National Cancer Institute-Frederick, Frederick, MD, USA
| | - Vinay K Pathak
- Viral Mutation Section, HIV Dynamics and Replication Program, National Cancer Institute-Frederick, Frederick, MD, USA
| | - Philip R Tedbury
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Stefan G Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
Hayashi Y, Jono H. Recent Advances in Oligonucleotide-Based Therapy for Transthyretin Amyloidosis: Clinical Impact and Future Prospects. Biol Pharm Bull 2019; 41:1737-1744. [PMID: 30504675 DOI: 10.1248/bpb.b18-00625] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transthyretin (TTR) amyloidosis, also known as transthyretin-related familial amyloidotic polyneuropathy (ATTR-FAP), is a fatal hereditary systemic amyloidosis caused by mutant forms of TTR. Although conventional treatments for ATTR-FAP, such as liver transplantation (LT) and TTR tetramer stabilizer, reportedly halt the progression of clinical manifestation, these therapies have several limitations. Oligonucleotide-based therapy, e.g. small interfering RNA (siRNA)- and antisense oligonucleotides (ASOs)-based therapy, hold enormous potential for the treatment of intractable diseases such as ATTR-FAP, by specifically regulating the gene responsible for the disease. Clinical evidence strongly suggests that LT inhibits mutant TTR production, thus improving the manifestation of ATTR-FAP. Therefore, an oligonucleotide-based therapy for ATTR-FAP, which reduces the production of TTR by the liver, has recently been developed in preclinical and clinical studies. This review focuses on recent advances in oligonucleotide-based therapy and future prospects of next-generation oligonucleotide-based drugs for therapeutic use against ATTR-FAP.
Collapse
Affiliation(s)
- Yuya Hayashi
- Department of Pharmacy, Kumamoto University Hospital
| | - Hirofumi Jono
- Department of Pharmacy, Kumamoto University Hospital.,Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
5
|
Wang Y, Xu C, Ma L, Mou Y, Zhang B, Zhou S, Tian Y, Trinh J, Zhang X, Li XJ. Drug screening with human SMN2 reporter identifies SMN protein stabilizers to correct SMA pathology. Life Sci Alliance 2019; 2:2/2/e201800268. [PMID: 30910806 PMCID: PMC6435041 DOI: 10.26508/lsa.201800268] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 01/14/2023] Open
Abstract
Spinal muscular atrophy (SMA), the leading genetic cause of infant mortality, is caused by reduced levels of functional survival motor neuron (SMN) protein. To identify therapeutic agents for SMA, we established a versatile SMN2-GFP reporter line by targeting the human SMN2 gene. We then screened a compound library and identified Z-FA-FMK as a potent candidate. Z-FA-FMK, a cysteine protease inhibitor, increased functional SMN through inhibiting the protease-mediated degradation of both full-length and exon 7-deleted forms of SMN. Further studies reveal that CAPN1, CAPN7, CTSB, and CTSL mediate the degradation of SMN proteins, providing novel targets for SMA. Notably, Z-FA-FMK mitigated mitochondriopathy and neuropathy in SMA patient-derived motor neurons and showed protective effects in SMA animal model after intracerebroventricular injection. E64d, another cysteine protease inhibitor which can pass through the blood-brain barrier, showed even more potent therapeutic effects after subcutaneous delivery to SMA mice. Taken together, we have successfully established a human SMN2 reporter for future drug discovery and identified the potential therapeutic value of cysteine protease inhibitors in treating SMA via stabilizing SMN proteins.
Collapse
Affiliation(s)
- Yiran Wang
- Brain and Spinal Cord Innovative Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chongchong Xu
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Lin Ma
- Brain and Spinal Cord Innovative Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Key Laboratory of Reconstruction and Regeneration of Spine and Spinal Cord Injury, Ministry of Education, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University, School of Medicine, Shanghai, China
| | - Yongchao Mou
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Bowen Zhang
- Brain and Spinal Cord Innovative Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shanshan Zhou
- Brain and Spinal Cord Innovative Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yue Tian
- Brain and Spinal Cord Innovative Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jessica Trinh
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, USA
| | - Xiaoqing Zhang
- Brain and Spinal Cord Innovative Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China .,Key Laboratory of Reconstruction and Regeneration of Spine and Spinal Cord Injury, Ministry of Education, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, Tongji University, School of Medicine, Shanghai, China.,Tsingtao Advanced Research Institute, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xue-Jun Li
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, USA .,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
6
|
Tian X, Gu T, Patel S, Bode AM, Lee MH, Dong Z. CRISPR/Cas9 - An evolving biological tool kit for cancer biology and oncology. NPJ Precis Oncol 2019; 3:8. [PMID: 30911676 PMCID: PMC6423228 DOI: 10.1038/s41698-019-0080-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
The development of genetic engineering in the 1970s marked a new frontier in genome-editing technology. Gene-editing technologies have provided a plethora of benefits to the life sciences. The clustered regularly interspaced short palindromic repeats/CRISPR associated protein 9 (CRISPR/ Cas9) system is a versatile technology that provides the ability to add or remove DNA in the genome in a sequence-specific manner. Serious efforts are underway to improve the efficiency of CRISPR/Cas9 targeting and thus reduce off-target effects. Currently, various applications of CRISPR/Cas9 are used in cancer biology and oncology to perform robust site-specific gene editing, thereby becoming more useful for biological and clinical applications. Many variants and applications of CRISPR/Cas9 are being rapidly developed. Experimental approaches that are based on CRISPR technology have created a very promising tool that is inexpensive and simple for developing effective cancer therapeutics. This review discusses diverse applications of CRISPR-based gene-editing tools in oncology and potential future cancer therapies.
Collapse
Affiliation(s)
- Xueli Tian
- Basic Medical College, Zhengzhou University, 450001 Zhengzhou, Henan China
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, 450008 Zhengzhou, Henan China
| | - Tingxuan Gu
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, 450008 Zhengzhou, Henan China
| | - Satyananda Patel
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, 450008 Zhengzhou, Henan China
| | - Ann M. Bode
- The Hormel Institute, University of Minnesota, Austin, 55912 USA
| | - Mee-Hyun Lee
- Basic Medical College, Zhengzhou University, 450001 Zhengzhou, Henan China
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, 450008 Zhengzhou, Henan China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China
| | - Zigang Dong
- Basic Medical College, Zhengzhou University, 450001 Zhengzhou, Henan China
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, 450008 Zhengzhou, Henan China
- The Hormel Institute, University of Minnesota, Austin, 55912 USA
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China
| |
Collapse
|
7
|
Cavaliere G. Genome editing and assisted reproduction: curing embryos, society or prospective parents? MEDICINE, HEALTH CARE, AND PHILOSOPHY 2018; 21:215-225. [PMID: 28725950 PMCID: PMC5956052 DOI: 10.1007/s11019-017-9793-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
This paper explores the ethics of introducing genome-editing technologies as a new reproductive option. In particular, it focuses on whether genome editing can be considered a morally valuable alternative to preimplantation genetic diagnosis (PGD). Two arguments against the use of genome editing in reproduction are analysed, namely safety concerns and germline modification. These arguments are then contrasted with arguments in favour of genome editing, in particular with the argument of the child's welfare and the argument of parental reproductive autonomy. In addition to these two arguments, genome editing could be considered as a worthy alternative to PGD as it may not be subjected to some of the moral critiques moved against this technology. Even if these arguments offer sound reasons in favour of introducing genome editing as a new reproductive option, I conclude that these benefits should be balanced against other considerations. More specifically, I maintain that concerns regarding the equality of access to assisted reproduction and the allocation of scarce resources should be addressed prior to the adoption of genome editing as a new reproductive option.
Collapse
Affiliation(s)
- Giulia Cavaliere
- Wellcome Trust PhD Student in Bioethics & Society, Department of Global Health & Social Medicine, King's College London, London, UK.
| |
Collapse
|
8
|
García-Tuñón I, Hernández-Sánchez M, Ordoñez JL, Alonso-Pérez V, Álamo-Quijada M, Benito R, Guerrero C, Hernández-Rivas JM, Sánchez-Martín M. The CRISPR/Cas9 system efficiently reverts the tumorigenic ability of BCR/ABL in vitro and in a xenograft model of chronic myeloid leukemia. Oncotarget 2018; 8:26027-26040. [PMID: 28212528 PMCID: PMC5432235 DOI: 10.18632/oncotarget.15215] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 01/27/2017] [Indexed: 11/25/2022] Open
Abstract
CRISPR/Cas9 technology was used to abrogate p210 oncoprotein expression in the Boff-p210 cell line, a pro-B line derived from interlukin-3-dependent Baf/3, that shows IL-3-independence arising from the constitutive expression of BCR-ABL p210. Using this approach, pools of Boff-p210-edited cells and single edited cell-derived clones were obtained and functionally studied in vitro. The loss of p210 expression in Boff-p210 cells resulted in the loss of ability to grow in the absence of IL-3, as the Baf/3 parental line, showing significantly increased apoptosis levels. Notably, in a single edited cell-derived clone carrying a frame-shift mutation that prevents p210 oncoprotein expression, the effects were even more drastic, resulting in cell death. These edited cells were injected subcutaneously in immunosuppressed mice and tumor growth was followed for three weeks. BCR/ABL-edited cells developed smaller tumors than those originating from unedited Boff-p210 parental cells. Interestingly, the single edited cell-derived clone was unable to develop tumors, similar to what is observed with the parental Baf/3 cell line. CRISPR/Cas9 genomic editing technology allows the ablation of the BCR/ABL fusion gene, causing an absence of oncoprotein expression, and blocking its tumorigenic effects in vitro and in the in vivo xenograft model of CML. The future application of this approach in in vivo models of CML will allow us to more accurately assess the value of CRISPR/Cas9 technology as a new therapeutic tool that overcomes resistance to the usual treatments for CML patients.
Collapse
Affiliation(s)
- Ignacio García-Tuñón
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
| | - María Hernández-Sánchez
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
| | - José Luis Ordoñez
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Veronica Alonso-Pérez
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Miguel Álamo-Quijada
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Rocio Benito
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Carmen Guerrero
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain.,Instituto Biología Molecular y Celular del Cáncer (USAL/CSIC), Salamanca, Spain.,Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Jesús María Hernández-Rivas
- Unidad de Diagnóstico Molecular y Celular del Cáncer, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain.,IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain.,Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Manuel Sánchez-Martín
- IBSAL, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain.,Servicio de Transgénesis, Nucleus, Universidad de Salamanca, Salamanca, Spain.,Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
9
|
Shen Y, Zhang J, Yu T, Qi C. Generation of PTEN knockout bone marrow mesenchymal stem cell lines by CRISPR/Cas9-mediated genome editing. Cytotechnology 2018; 70:783-791. [PMID: 29387984 DOI: 10.1007/s10616-017-0183-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/15/2017] [Indexed: 12/15/2022] Open
Abstract
The tumor suppressor PTEN is involved in the regulation of cell proliferation, lineage determination, motility, adhesion and apoptosis. Loss of PTEN in the bone mesenchymal stem cells (BMSCs) was shown to change their function in the repair tissue. So far, the CRISPR/Cas9 system has been proven extremely simple and flexible. Using this system to manipulate PTEN gene editing could produce the PTEN-Knocking-out (PTEN-KO) strain. We knocked out PTEN in MSCs and validated the expression by PCR and Western blot. To clarify the changes in proliferation, CCK-8 assay was applied. In support, living cell proportion was assessed by Trypan blue staining. For osteogenic and adipogenic induction, cells were cultured in different media for 2 weeks. Oil red staining and alizarin red staining were performed for assessment of osteogenic or adipogenic differentiation. The expression of Id4, Runx2, ALP and PPARγ was examined by qPCR and immunocytochemistry staining. The PTEN-KO strain was identified by sequencing. The PTEN-KO cells had an increased cell viability and higher survival compared with the wild type. However, decreased expression of Runx2 and PPARγ was found in the PTEN loss strain after induction, and consistently decreased osteogenic or adipogenic differentiation was observed by alizarin and oil red staining. Together, PTEN-KO strain showed an increased proliferation capability but decreased multi-directional differentiation potential. When BMSCs serve as seed cells for tissue engineering, the PTEN gene may be used as an indicator.
Collapse
Affiliation(s)
- Youliang Shen
- Department of Orthopaedics, Jiao Zhou Central Hospital of Qingdao City, Qingdao, 266300, China
| | - Jingjing Zhang
- Department of Orthopaedics, Jiao Zhou Central Hospital of Qingdao City, Qingdao, 266300, China
| | - Tengbo Yu
- Orthopaedic Center, The Affiliated Hospital of Qingdao University, Qingdao, 266300, China
| | - Chao Qi
- Orthopaedic Center, The Affiliated Hospital of Qingdao University, Qingdao, 266300, China.
| |
Collapse
|
10
|
White MK, Khalili K. CRISPR/Cas9 and cancer targets: future possibilities and present challenges. Oncotarget 2017; 7:12305-17. [PMID: 26840090 PMCID: PMC4914286 DOI: 10.18632/oncotarget.7104] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/23/2016] [Indexed: 02/07/2023] Open
Abstract
All cancers have multiple mutations that can largely be grouped into certain classes depending on the function of the gene in which they lie and these include oncogenic changes that enhance cellular proliferation, loss of function of tumor suppressors that regulate cell growth potential and induction of metabolic enzymes that confer resistance to chemotherapeutic agents. Thus the ability to correct such mutations is an important goal in cancer treatment. Recent research has led to the developments of reagents which specifically target nucleotide sequences within the cellular genome and these have a huge potential for expanding our anticancer armamentarium. One such a reagent is the clustered regulatory interspaced short palindromic repeat (CRISPR)-associated 9 (Cas9) system, a powerful, highly specific and adaptable tool that provides unparalleled control for editing the cellular genome. In this short review, we discuss the potential of CRISPR/Cas9 against human cancers and the current difficulties in translating this for novel therapeutic approaches.
Collapse
Affiliation(s)
- Martyn K White
- Department of Neuroscience, Center for Neurovirology and Comprehensive Neuroaids Center, Temple University School of Medicine, Philadelphia, PA, USA
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology and Comprehensive Neuroaids Center, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
11
|
Gautron AS, Juillerat A, Guyot V, Filhol JM, Dessez E, Duclert A, Duchateau P, Poirot L. Fine and Predictable Tuning of TALEN Gene Editing Targeting for Improved T Cell Adoptive Immunotherapy. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 9:312-321. [PMID: 29246309 PMCID: PMC5684446 DOI: 10.1016/j.omtn.2017.10.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 12/25/2022]
Abstract
Using a TALEN-mediated gene-editing approach, we have previously described a process for the large-scale manufacturing of “off-the-shelf” CAR T cells from third-party donor T cells by disrupting the gene encoding TCRα constant chain (TRAC). Taking advantage of a previously described strategy to control TALEN targeting based on the exclusion capacities of non-conventional RVDs, we have developed highly efficient and specific nucleases targeting a key T cell immune checkpoint, PD-1, to improve engineered CAR T cells’ functionalities. Here, we demonstrate that this approach allows combined TRAC and PDCD1 TALEN processing at the desired locus while eliminating low-frequency off-site processing. Thus, by replacing few RVDs, we provide here an easy and rapid redesign of optimal TALEN combinations. We anticipate that this method can greatly benefit multiplex editing, which is of key importance especially for therapeutic applications where high editing efficiencies need to be associated with maximal specificity and safety.
Collapse
Affiliation(s)
| | | | - Valérie Guyot
- Cellectis SA, 8 Rue de la Croix Jarry, 75013 Paris, France
| | | | - Emilie Dessez
- Cellectis SA, 8 Rue de la Croix Jarry, 75013 Paris, France
| | | | | | - Laurent Poirot
- Cellectis SA, 8 Rue de la Croix Jarry, 75013 Paris, France
| |
Collapse
|
12
|
Cui J, Chew SJL, Shi Y, Gong Z, Shen HM. CRISPR system for genome engineering: the application for autophagy study. BMB Rep 2017; 50:247-256. [PMID: 28288698 PMCID: PMC5458674 DOI: 10.5483/bmbrep.2017.50.5.044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Indexed: 11/20/2022] Open
Abstract
CRISPR/Cas9 is the latest tool introduced in the field of genome engineering and is so far the best genome-editing tool as compared to its precedents such as, meganucleases, zinc finger nucleases (ZFNs) and transcription activator-like effectors (TALENs). The simple design and assembly of the CRISPR/Cas9 system makes genome editing easy to perform as it uses small guide RNAs that correspond to their DNA targets for high efficiency editing. This has helped open the doors for multiplexible genome targeting in many species that were intractable using old genetic perturbation techniques. Currently, The CRISPR system is revolutionizing the way biological researches are conducted and paves a bright future not only in research but also in medicine and biotechnology. In this review, we evaluated the history, types and structure, the mechanism of action of CRISPR/Cas System. In particular, we focused on the application of this powerful tool in autophagy research. [BMB Reports 2017; 50(5): 247-256].
Collapse
Affiliation(s)
- Jianzhou Cui
- Department of Physiology, Yong Loo Lin School of Medicine, National University of
Singapore
| | - Shirley Jia Li Chew
- Department of Physiology, Yong Loo Lin School of Medicine, National University of
Singapore
| | - Yin Shi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of
Singapore
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of
Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of
Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore,
Singapore
| |
Collapse
|
13
|
Cho HM, Kim PH, Chang HK, Shen YM, Bonsra K, Kang BJ, Yum SY, Kim JH, Lee SY, Choi MC, Kim HH, Jang G, Cho JY. Targeted Genome Engineering to Control VEGF Expression in Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells: Potential Implications for the Treatment of Myocardial Infarction. Stem Cells Transl Med 2017; 6:1040-1051. [PMID: 28186692 PMCID: PMC5442764 DOI: 10.1002/sctm.16-0114] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 08/11/2016] [Accepted: 09/01/2016] [Indexed: 12/19/2022] Open
Abstract
Human umbilical cord blood‐derived mesenchymal stem cells (hUCB‐MSCs) exhibit potency for the regeneration of infarcted hearts. Vascular endothelial growth factor (VEGF) is capable of inducing angiogenesis and can boost stem cell‐based therapeutic effects. However, high levels of VEGF can cause abnormal blood vessel growth and hemangiomas. Thus, a controllable system to induce therapeutic levels of VEGF is required for cell therapy. We generated an inducible VEGF‐secreting stem cell (VEGF/hUCB‐MSC) that controls the expression of VEGF and tested the therapeutic efficacy in rat myocardial infarction (MI) model to apply functional stem cells to MI. To introduce the inducible VEGF gene cassette into a safe harbor site of the hUCB‐MSC chromosome, the transcription activator‐like effector nucleases system was used. After confirming the integration of the cassette into the locus, VEGF secretion in physiological concentration from VEGF/hUCB‐MSCs after doxycycline (Dox) induction was proved in conditioned media. VEGF secretion was detected in mice implanted with VEGF/hUCB‐MSCs grown via a cell sheet system. Vessel formation was induced in mice transplanted with Matrigel containing VEGF/hUCB‐MSCs treated with Dox. Moreover, seeding of the VEGF/hUCB‐MSCs onto the cardiac patch significantly improved the left ventricle ejection fraction and fractional shortening in a rat MI model upon VEGF induction. Induced VEGF/hUCB‐MSC patches significantly decreased the MI size and fibrosis and increased muscle thickness, suggesting improved survival of cardiomyocytes and protection from MI damage. These results suggest that our inducible VEGF‐secreting stem cell system is an effective therapeutic approach for the treatment of MI. Stem Cells Translational Medicine2017;6:1040–1051
Collapse
Affiliation(s)
- Hyun-Min Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Pyung-Hwan Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyun-Kyung Chang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Yi-Ming Shen
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Kwaku Bonsra
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Byung-Jae Kang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Soo-Young Yum
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Joo-Hyun Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - So-Yeong Lee
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Min-Cheol Choi
- Department of Veterinary Radiology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyongbum Henry Kim
- Department of Pharmacology, College of Medicine, Yonsei University, Seoul, South Korea
| | - Goo Jang
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
14
|
Chen KY, Knoepfler PS. To CRISPR and beyond: the evolution of genome editing in stem cells. Regen Med 2016; 11:801-816. [PMID: 27905217 PMCID: PMC5221123 DOI: 10.2217/rme-2016-0107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/11/2016] [Indexed: 12/17/2022] Open
Abstract
The goal of editing the genomes of stem cells to generate model organisms and cell lines for genetic and biological studies has been pursued for decades. There is also exciting potential for future clinical impact in humans. While recent, rapid advances in targeted nuclease technologies have led to unprecedented accessibility and ease of gene editing, biology has benefited from past directed gene modification via homologous recombination, gene traps and other transgenic methodologies. Here we review the history of genome editing in stem cells (including via zinc finger nucleases, transcription activator-like effector nucleases and CRISPR-Cas9), discuss recent developments leading to the implementation of stem cell gene therapies in clinical trials and consider the prospects for future advances in this rapidly evolving field.
Collapse
Affiliation(s)
- Kuang-Yui Chen
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Davis, CA, USA
- Institute of Pediatric Regenerative Medicine, Shriners Hospital For Children Northern California, Sacramento, CA, USA
- Genome Center, University of California Davis, Davis, CA, USA
| | - Paul S Knoepfler
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Davis, CA, USA
- Institute of Pediatric Regenerative Medicine, Shriners Hospital For Children Northern California, Sacramento, CA, USA
- Genome Center, University of California Davis, Davis, CA, USA
| |
Collapse
|
15
|
Modeling Axonal Defects in Hereditary Spastic Paraplegia with Human Pluripotent Stem Cells. ACTA ACUST UNITED AC 2016; 11:339-354. [PMID: 27956894 DOI: 10.1007/s11515-016-1416-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cortical motor neurons, also known as upper motor neurons, are large projection neurons whose axons convey signals to lower motor neurons to control the muscle movements. Degeneration of cortical motor neuron axons is implicated in several debilitating disorders, including hereditary spastic paraplegia (HSP) and amyotrophic lateral sclerosis (ALS). Since the discovery of the first HSP gene, SPAST that encodes spastin, over 70 distinct genetic loci associated with HSP have been identified. How the mutations of these functionally diverse genes result in axonal degeneration and why certain axons are affected in HSP remains largely unknown. The development of induced pluripotent stem cell (iPSC) technology has provided researchers an excellent resource to generate patient-specific human neurons to model human neuropathologic processes including axonal defects. METHODS In this article, we will frst review the pathology and pathways affected in the common forms of HSP subtypes by searching the PubMed database. We will then summurize the findings and insights gained from studies using iPSC-based models, and discuss the challenges and future directions. RESULTS HSPs, a heterogeneous group of genetic neurodegenerative disorders, are characterized by lower extremity weakness and spasticity that result from retrograde axonal degeneration of cortical motor neurons. Recently, iPSCs have been generated from several common forms of HSP including SPG4, SPG3A, and SPG11 patients. Neurons derived from HSP iPSCs exhibit disease-relevant axonal defects, such as impaired neurite outgrowth, increased axonal swellings, and reduced axonal transport. CONCLUSION These patient-derived neurons offer unique tools to study the pathogenic mechanisms and explore the treatments for rescuing axonal defects in HSP, as well as other diseases involving axonopathy.
Collapse
|
16
|
Swamy MN, Wu H, Shankar P. Recent advances in RNAi-based strategies for therapy and prevention of HIV-1/AIDS. Adv Drug Deliv Rev 2016; 103:174-186. [PMID: 27013255 PMCID: PMC4935623 DOI: 10.1016/j.addr.2016.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/15/2022]
Abstract
RNA interference (RNAi) provides a powerful tool to silence specific gene expression and has been widely used to suppress host factors such as CCR5 and/or viral genes involved in HIV-1 replication. Newer nuclease-based gene-editing technologies, such as zinc finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN) and the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system, also provide powerful tools to ablate specific genes. Because of differences in co-receptor usage and the high mutability of the HIV-1 genome, a combination of host factors and viral genes needs to be suppressed for effective prevention and treatment of HIV-1 infection. Whereas the continued presence of small interfering/short hairpin RNA (si/shRNA) mediators is needed for RNAi to be effective, the continued expression of nucleases in the gene-editing systems is undesirable. Thus, RNAi provides the only practical way for expression of multiple silencers in infected and uninfected cells, which is needed for effective prevention/treatment of infection. There have been several advances in the RNAi field in terms of si/shRNA design, targeted delivery to HIV-1 susceptible cells, and testing for efficacy in preclinical humanized mouse models. Here, we comprehensively review the latest advances in RNAi technology towards prevention and treatment of HIV-1.
Collapse
Affiliation(s)
- Manjunath N Swamy
- Center of Emphasis in Infectious Disease, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA.
| | - Haoquan Wu
- Center of Emphasis in Infectious Disease, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Premlata Shankar
- Center of Emphasis in Infectious Disease, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA.
| |
Collapse
|
17
|
Yu B, Lu R, Yuan Y, Zhang T, Song S, Qi Z, Shao B, Zhu M, Mi F, Cheng Y. Efficient TALEN-mediated myostatin gene editing in goats. BMC DEVELOPMENTAL BIOLOGY 2016; 16:26. [PMID: 27461387 PMCID: PMC4962387 DOI: 10.1186/s12861-016-0126-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 07/15/2016] [Indexed: 12/27/2022]
Abstract
Background Myostatin (MSTN) encodes a negative regulator of skeletal muscle mass that might have applications for promoting muscle growth in livestock. In this study, we aimed to test whether targeted MSTN editing, mediated by transcription activator-like effector nucleases (TALENs), is a viable approach to create myostatin-modified goats (Capra hircus). Results We obtained a pair of TALENs (MTAL-2) that could recognize and cut the targeted MSTN site in the goat genome. Fibroblasts from pedigreed goats were co-transfected with MTAL-2, and 272 monoclonal cell strains were confirmed to have mono- or bi-allelic mutations in MSTN. Ten cell strains with different genotypes were used as donor cells for somatic cell nuclear transfer, which produced three cloned kids (K179/MSTN−/−, K52-2/MSTN+/−, and K52-1/MSTN+/+). Conclusions The results suggested that the MTAL-2 could disrupt MSTN efficiently in the goat genome. The mutated somatic cells could be used to produce MSTN-site mutated goats without developmental disruption. Thus, TALENs is an effective method for accurate genome editing to produce site-modified goats. Electronic supplementary material The online version of this article (doi:10.1186/s12861-016-0126-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Baoli Yu
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Rui Lu
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Yuguo Yuan
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Ting Zhang
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Shaozheng Song
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Zhengqiang Qi
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Bin Shao
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Mengmin Zhu
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Fei Mi
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China
| | - Yong Cheng
- College of Veterinary Medicine, Yangzhou University, No. 12 Wenhui Road, Yangzhou, 225009, Jiangsu Province, People's Republic of China.
| |
Collapse
|
18
|
Choi JG, Dang Y, Abraham S, Ma H, Zhang J, Guo H, Cai Y, Mikkelsen JG, Wu H, Shankar P, Manjunath N. Lentivirus pre-packed with Cas9 protein for safer gene editing. Gene Ther 2016; 23:627-33. [PMID: 27052803 DOI: 10.1038/gt.2016.27] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 02/02/2016] [Accepted: 02/29/2016] [Indexed: 12/22/2022]
Abstract
The CRISPR/Cas9 system provides an easy way to edit specific site/s in the genome and thus offers tremendous opportunity for human gene therapy for a wide range of diseases. However, one major concern is off-target effects, particularly with long-term expression of Cas9 nuclease when traditional expression methods such as via plasmid/viral vectors are used. To overcome this limitation, we pre-packaged Cas9 protein (Cas9P LV) in lentiviral particles for transient exposure and showed its effectiveness for gene disruption in cells, including primary T cells expressing specific single guide RNAs (sgRNAs). We then constructed an 'all in one virus' to express sgRNAs in association with pre-packaged Cas9 protein (sgRNA/Cas9P LV). We successfully edited CCR5 in TZM-bl cells by this approach. Using an sgRNA-targeting HIV long terminal repeat, we also were able to disrupt HIV provirus in the J-LAT model of viral latency. Moreover, we also found that pre-packaging Cas9 protein in LV particle reduced off-target editing of chromosome 4:-29134166 locus by CCR5 sgRNA, compared with continued expression from the vector. These results show that sgRNA/Cas9P LV can be used as a safer approach for human gene therapy applications.
Collapse
Affiliation(s)
- J G Choi
- Department of Biomedical Sciences, Center of Emphasis in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Y Dang
- Department of Biomedical Sciences, Center of Emphasis in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - S Abraham
- Department of Biomedical Sciences, Center of Emphasis in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - H Ma
- Department of Biomedical Sciences, Center of Emphasis in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - J Zhang
- Department of Biomedical Sciences, Center of Emphasis in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - H Guo
- Department of Biomedical Sciences, Center of Emphasis in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Y Cai
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - J G Mikkelsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - H Wu
- Department of Biomedical Sciences, Center of Emphasis in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - P Shankar
- Department of Biomedical Sciences, Center of Emphasis in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - N Manjunath
- Department of Biomedical Sciences, Center of Emphasis in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| |
Collapse
|
19
|
Vassena R, Heindryckx B, Peco R, Pennings G, Raya A, Sermon K, Veiga A. Genome engineering through CRISPR/Cas9 technology in the human germline and pluripotent stem cells. Hum Reprod Update 2016; 22:411-9. [PMID: 26932460 DOI: 10.1093/humupd/dmw005] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND With the recent development of CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 genome editing technology, the possibility to genetically manipulate the human germline (gametes and embryos) has become a distinct technical possibility. Although many technical challenges still need to be overcome in order to achieve adequate efficiency and precision of the technology in human embryos, the path leading to genome editing has never been simpler, more affordable, and widespread. OBJECTIVE AND RATIONALE In this narrative review we seek to understand the possible impact of CRISR/Cas9 technology on human reproduction from the technical and ethical point of view, and suggest a course of action for the scientific community. SEARCH METHODS This non-systematic review was carried out using Medline articles in English, as well as technical documents from the Human Fertilisation and Embryology Authority and reports in the media. The technical possibilities of the CRISPR/Cas9 technology with regard to human reproduction are analysed based on results obtained in model systems such as large animals and laboratory rodents. Further, the possibility of CRISPR/Cas9 use in the context of human reproduction, to modify embryos, germline cells, and pluripotent stem cells is reviewed based on the authors' expert opinion. Finally, the possible uses and consequences of CRISPR/cas9 gene editing in reproduction are analysed from the ethical point of view. OUTCOMES We identify critical technical and ethical issues that should deter from employing CRISPR/Cas9 based technologies in human reproduction until they are clarified. WIDER IMPLICATIONS Overcoming the numerous technical limitations currently associated with CRISPR/Cas9 mediated editing of the human germline will depend on intensive research that needs to be transparent and widely disseminated. Rather than a call to a generalized moratorium, or banning, of this type of research, efforts should be placed on establishing an open, international, collaborative and regulated research framework. Equally important, a societal discussion on the risks, benefits, and preferred applications of the new technology, including all relevant stakeholders, is urgently needed and should be promoted, and ultimately guide research priorities in this area.
Collapse
Affiliation(s)
- R Vassena
- Clínica EUGIN, Barcelona 08029, Spain
| | - B Heindryckx
- Ghent Fertility and Stem cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - R Peco
- Center for Regenerative Medicine in Barcelona (CMRB), 08003 Barcelona, Spain
| | - G Pennings
- Bioethics Institute Ghent (BIG), Faculty of Arts and Philosophy, Ghent University, Ghent, Belgium
| | - A Raya
- Center for Regenerative Medicine in Barcelona (CMRB), 08003 Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - K Sermon
- Research Group Reproduction and Genetics, Vrije Universiteit Brussel, Brussels, Belgium
| | - A Veiga
- Center for Regenerative Medicine in Barcelona (CMRB), 08003 Barcelona, Spain Reproductive Medicine Service, Hospital Universitari Quiron Dexeus, Barcelona, Spain
| |
Collapse
|
20
|
Darbandi S, Darbandi M, Khorshid HRK, Sadeghi MR, Al-Hasani S, Agarwal A, Shirazi A, Heidari M, Akhondi MM. Experimental strategies towards increasing intracellular mitochondrial activity in oocytes: A systematic review. Mitochondrion 2016; 30:8-17. [PMID: 27234976 DOI: 10.1016/j.mito.2016.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/04/2016] [Accepted: 05/20/2016] [Indexed: 12/19/2022]
Abstract
PURPOSE The mitochondrial complement is critical in sustaining the earliest stages of life. To improve the Assisted Reproductive Technology (ART), current methods of interest were evaluated for increasing the activity and copy number of mitochondria in the oocyte cell. METHODS This covered the researches from 1966 to September 2015. RESULTS The results provided ten methods that can be studied individually or simultaneously. CONCLUSION Though the use of these techniques generated great concern about heteroplasmy observation in humans, it seems that with study on these suggested methods there is real hope for effective treatments of old oocyte or oocytes containing mitochondrial problems in the near future.
Collapse
Affiliation(s)
- Sara Darbandi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| | - Mahsa Darbandi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| | | | - Mohammad Reza Sadeghi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| | - Safaa Al-Hasani
- Reproductive Medicine Unit, University of Schleswig-Holstein, Luebeck, Germany.
| | - Ashok Agarwal
- Center for Reproductive Medicine, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Abolfazl Shirazi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| | - Mahnaz Heidari
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran. M.@avicenna.ar.ir
| | - Mohammad Mehdi Akhondi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| |
Collapse
|
21
|
Nii T, Kohara H, Marumoto T, Sakuma T, Yamamoto T, Tani K. Single-Cell-State Culture of Human Pluripotent Stem Cells Increases Transfection Efficiency. Biores Open Access 2016; 5:127-36. [PMID: 27257519 PMCID: PMC4876534 DOI: 10.1089/biores.2016.0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Efficient gene transfer into human pluripotent stem cells (hPSCs) holds great promise for regenerative medicine and pharmaceutical development. In the past decade, various methods were developed for gene transfer into hPSCs; however, hPSCs form tightly packed colonies, making gene transfer difficult. In this study, we established a stable culture method of hPSCs at a single-cell state to reduce cell density and investigated gene transfection efficiency followed by gene editing efficiency. hPSCs cultured in a single-cell state were transfected using nonliposomal transfection reagents with plasmid DNA or mRNA encoding enhanced green fluorescent protein. We found that most cells (DNA > 90%; mRNA > 99%) were transfected without the loss of undifferentiated PSC marker expression or pluripotency. Moreover, we demonstrated an efficient gene editing method using transcription activator-like effector nucleases (TALENs) targeting the adenomatous polyposis coli (APC) gene. Our new method may improve hPSC gene transfer techniques, thus facilitating their use for human regenerative medicine.
Collapse
Affiliation(s)
- Takenobu Nii
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University , Fukuoka, Japan
| | - Hiroshi Kohara
- Project Division of ALA Advanced Medical Research, The Institute of Medical Science, The University of Tokyo , Tokyo, Japan
| | - Tomotoshi Marumoto
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.; Department of Advanced Molecular and Cell Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Tetsushi Sakuma
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University , Hiroshima, Japan
| | - Takashi Yamamoto
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University , Hiroshima, Japan
| | - Kenzaburo Tani
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.; Project Division of ALA Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.; Department of Advanced Molecular and Cell Therapy, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
22
|
White MK, Khalili K. CRISPR/Cas9 and cancer targets: future possibilities and present challenges. Oncotarget 2016. [PMID: 26840090 DOI: 10.18632/oncotarget.7104.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
All cancers have multiple mutations that can largely be grouped into certain classes depending on the function of the gene in which they lie and these include oncogenic changes that enhance cellular proliferation, loss of function of tumor suppressors that regulate cell growth potential and induction of metabolic enzymes that confer resistance to chemotherapeutic agents. Thus the ability to correct such mutations is an important goal in cancer treatment. Recent research has led to the developments of reagents which specifically target nucleotide sequences within the cellular genome and these have a huge potential for expanding our anticancer armamentarium. One such a reagent is the clustered regulatory interspaced short palindromic repeat (CRISPR)-associated 9 (Cas9) system, a powerful, highly specific and adaptable tool that provides unparalleled control for editing the cellular genome. In this short review, we discuss the potential of CRISPR/Cas9 against human cancers and the current difficulties in translating this for novel therapeutic approaches.
Collapse
Affiliation(s)
- Martyn K White
- Department of Neuroscience, Center for Neurovirology and Comprehensive Neuroaids Center, Temple University School of Medicine, Philadelphia, PA, USA
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology and Comprehensive Neuroaids Center, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
23
|
Frame FM, Pellacani D, Collins AT, Maitland NJ. Harvesting Human Prostate Tissue Material and Culturing Primary Prostate Epithelial Cells. Methods Mol Biol 2016; 1443:181-201. [PMID: 27246341 DOI: 10.1007/978-1-4939-3724-0_12] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In order to fully explore the biology of a complex solid tumor such as prostate cancer, it is desirable to work with patient tissue. Only by working with cells from a tissue can we take into account patient variability and tumor heterogeneity. Cell lines have long been regarded as the workhorse of cancer research and it could be argued that they are of most use when considered within a panel of cell lines, thus taking into account specified mutations and variations in phenotype between different cell lines. However, often very different results are obtained when comparing cell lines to primary cells cultured from tissue. It stands to reason that cells cultured from patient tissue represents a close-to-patient model that should and does produce clinically relevant data. This chapter aims to illustrate the methods of processing, storing and culturing cells from prostate tissue, with a description of potential uses.
Collapse
Affiliation(s)
- Fiona M Frame
- YCR Cancer Research Unit, Department of Biology, University of York, Heslington, North Yorkshire, YO10 5DD, UK.
| | - Davide Pellacani
- YCR Cancer Research Unit, Department of Biology, University of York, Heslington, North Yorkshire, YO10 5DD, UK
| | - Anne T Collins
- YCR Cancer Research Unit, Department of Biology, University of York, Heslington, North Yorkshire, YO10 5DD, UK
| | - Norman J Maitland
- YCR Cancer Research Unit, Department of Biology, University of York, Heslington, North Yorkshire, YO10 5DD, UK.
| |
Collapse
|
24
|
Hemphill J, Borchardt EK, Brown K, Asokan A, Deiters A. Optical Control of CRISPR/Cas9 Gene Editing. J Am Chem Soc 2015; 137:5642-5. [PMID: 25905628 DOI: 10.1021/ja512664v] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The CRISPR/Cas9 system has emerged as an important tool in biomedical research for a wide range of applications, with significant potential for genome engineering and gene therapy. In order to achieve conditional control of the CRISPR/Cas9 system, a genetically encoded light-activated Cas9 was engineered through the site-specific installation of a caged lysine amino acid. Several potential lysine residues were identified as viable caging sites that can be modified to optically control Cas9 function, as demonstrated through optical activation and deactivation of both exogenous and endogenous gene function.
Collapse
Affiliation(s)
- James Hemphill
- †Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | | | - Kalyn Brown
- †Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | | | - Alexander Deiters
- †Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
25
|
Abstract
For over a century, researchers have observed similar neurodegenerative hallmarks in brains of people affected by rare early-onset lysosomal storage diseases and late-onset neurodegenerative diseases such as Alzheimer's and Parkinson's disease. Increasing evidence suggests these apparently disparate diseases share a common underlying feature, namely, a dysfunctional clearance of cellular cargo through the secretory-endosomal-autophagic-lysosomal-exocytic (SEALE) network. By providing examples of rare and common neurodegenerative diseases known to have pathologically altered cargo flux through the SEALE network, we explore the unifying hypothesis that impaired catabolism or exocytosis of SEALE cargo, places a burden of stress on neurons that initiates pathogenesis. We also describe how a growing understanding of genetic, epigenetic and age-related modifications of the SEALE network, has inspired a number of novel disease-modifying therapeutic approaches aimed at alleviating SEALE storage and providing therapeutic benefit to people affected by these devastating diseases across the age spectrum.
Collapse
Affiliation(s)
- Barry Boland
- Department of Pharmacology and Therapeutics, School of Medicine, University College Cork, Cork, Ireland.
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom.
| |
Collapse
|
26
|
Khalili K, Kaminski R, Gordon J, Cosentino L, Hu W. Genome editing strategies: potential tools for eradicating HIV-1/AIDS. J Neurovirol 2015; 21:310-21. [PMID: 25716921 DOI: 10.1007/s13365-014-0308-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/05/2014] [Accepted: 12/22/2014] [Indexed: 12/26/2022]
Abstract
Current therapy for controlling human immunodeficiency virus (HIV-1) infection and preventing acquired immunodeficiency syndrome (AIDS) progression has profoundly decreased viral replication in cells susceptible to HIV-1 infection, but it does not eliminate the low level of viral replication in latently infected cells, which contain integrated copies of HIV-1 proviral DNA. There is an urgent need for the development of HIV-1 genome eradication strategies that will lead to a permanent or "sterile" cure of HIV-1/AIDS. In the past few years, novel nuclease-initiated genome editing tools have been developing rapidly, including zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the CRISPR/Cas9 system. These surgical knives, which can excise any genome, provide a great opportunity to eradicate the HIV-1 genome by targeting highly conserved regions of the HIV-1 long terminal repeats or essential viral genes. Given the time consuming and costly engineering of target-specific ZFNs and TALENs, the RNA-guided endonuclease Cas9 technology has emerged as a simpler and more versatile technology to allow permanent removal of integrated HIV-1 proviral DNA in eukaryotic cells, and hopefully animal models or human patients. The major unmet challenges of this approach at present include inefficient nuclease gene delivery, potential off-target cleavage, and cell-specific genome targeting. Nanoparticle or lentivirus-mediated delivery of next generation Cas9 technologies including nickase or RNA-guided FokI nuclease (RFN) will further improve the potential for genome editing to become a promising approach for curing HIV-1/AIDS.
Collapse
Affiliation(s)
- Kamel Khalili
- Department of Neuroscience, Center for Neurovirology and the Comprehensive NeuroAIDS Center, Temple University School of Medicine, Philadelphia, PA, 19140, USA,
| | | | | | | | | |
Collapse
|
27
|
Optimized tuning of TALEN specificity using non-conventional RVDs. Sci Rep 2015; 5:8150. [PMID: 25632877 PMCID: PMC4311247 DOI: 10.1038/srep08150] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 12/22/2014] [Indexed: 12/16/2022] Open
Abstract
A key feature when designing DNA targeting tools and especially nucleases is specificity. The ability to control and tune this important parameter represents an invaluable advance to the development of such molecular scissors. Here, we identified and characterized new non-conventional RVDs (ncRVDs) that possess novel intrinsic targeting specificity features. We further report a strategy to control TALEN targeting based on the exclusion capacities of ncRVDs (discrimination between different nucleotides). By implementing such ncRVDs, we demonstrated in living cells the possibility to efficiently promote TALEN-mediated processing of a target in the HBB locus and alleviate undesired off-site cleavage. We anticipate that this method can greatly benefit to designer nucleases, especially for therapeutic applications and synthetic biology.
Collapse
|
28
|
Abstract
The CRISPR (clustered regularly interspaced short palindromic repeat)-Cas9 (CRISPR-associated nuclease 9) system is poised to transform developmental biology by providing a simple, efficient method to precisely manipulate the genome of virtually any developing organism. This RNA-guided nuclease (RGN)-based approach already has been effectively used to induce targeted mutations in multiple genes simultaneously, create conditional alleles, and generate endogenously tagged proteins. Illustrating the adaptability of RGNs, the genomes of >20 different plant and animal species as well as multiple cell lines and primary cells have been successfully modified. Here we review the current and potential uses of RGNs to investigate genome function during development.
Collapse
Affiliation(s)
- Melissa M Harrison
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706, USA
| | - Brian V Jenkins
- Biochemistry Department, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Kate M O'Connor-Giles
- Laboratory of Genetics, Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Jill Wildonger
- Biochemistry Department, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA;
| |
Collapse
|
29
|
Fazio S, Tavori H. Peeking into a cool future: genome editing to delete PCSK9 and control hypercholesterolemia in a single shot. Circ Res 2014; 115:472-4. [PMID: 25124321 PMCID: PMC4137455 DOI: 10.1161/circresaha.114.304575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Sergio Fazio
- From The Knight Cardiovascular Institute, Oregon Health and Science University, Portland.
| | - Hagai Tavori
- From The Knight Cardiovascular Institute, Oregon Health and Science University, Portland
| |
Collapse
|