Potential CRISPR/Cas9 Associated Lenti-sgR5-Cas9 for CCR5 and CXCR4 Disruption Protects CD4+T

Nurul Hidayah, Totalenesya Reforrent Sutikno, Yuna Annisa Salsabila

Abstract

HIV reached 37.6 million people by the end of 2020. Antiretroviral has many side effects and is potentially resistant to HIV treatment. The development of advanced medical technology has led to the development of CRISPR. CRISPR is proven better than ZFN and TALEN. CRISPR-Cas9 can be specific and efficient on target. This scientific literature review uses the PRISMA method. HIV requires specific CC-chemokine receptor 4 (CCR5) and CX-chemokine receptor 4 (CXCR4) to invade host cells. Previous studies have shown that disrupting one of the co-receptors still allows HIV to invade. The vector used for CRISPR/Cas9 is Lenti-sgR5-Cas9. Both CCR5 and CXCR4 co-receptor target gRNAs containing U6-gX4-1/-2-crRNA-loop-tracrRNA were amplified then inserted into lenti-sgR5-Cas9. Primary isolation of CD4+T cells was taken from human blood. Lentivirus will be inserted into CD4+T. As a result, this disruption was shown to be able to protect CD4+ cells from HIV-1. The purpose of this literature review is to explain that there is the effectiveness of CD4+T resistance in disrupting CXCR4 and CCR5 receptors so that in the future, it can be used for HIV therapy development based on CRISPR/Cas9.

Keywords: CD4+T, CRISPR-Cas9, CXCR4, CCR5, lentivirus vector.


Full Text:

PDF


References


WHO. Why the HIV epidemic is not over. World Health Organization. 2021. Available from: https://www.who.int/news-room/spotlight/why-the-hiv-epidemic-is-not-over

MCQUILLAN, G.M., KRUSZON-MORAN, D., MASCIOTRA, S., GU, Q., and STORANDT, R. Prevalence and trends in HIV infection and testing among adults in the United States: The National Health and Nutrition Examination Surveys, 1999–2018. Journal of Acquired Immune Deficit Syndrome, 2021, 86(5): 523.

WAYMACK, J.R., and SUNDARESHAN, V. Acquired Immune Deficiency Syndrome. StatPearls. 2020.

IACOB, S.A., IACOB, D.G., and JUGULETE, G. Improving the Adherence to Antiretroviral Therapy, a Difficult but Essential Task for a Successful HIV Treatment—Clinical Points of View and Practical Considerations. Frontier of Pharmacology, 2017, 8(NOV): 831.

NDASHIMYE, E., and ARTS, E.J. The urgent need for more potent antiretroviral therapy in low-income countries to achieve UNAIDS 90-90-90 and complete eradication of AIDS by 2030. Infectious Diseases of Poverty, 2019, 8(1): 1–8.

MARBAN, C., FOROUZANFAR, F., AIT-AMMAR, A., FAHMI, F., EL MEKDAD, H., and DAOUAD, F. Targeting the Brain Reservoirs: Toward an HIV Cure. Frontier of Immunology, 2016, 0(SEP): 397.

GAJ, T., SIRK, S.J., SHUI, S., and LIU, J. Genome-Editing Technologies: Principles and Applications. Cold Spring Harbor perspectives in biology, 2016, 8(12).

LINO, C.A., HARPER, J.C., CARNEY, J.P., and TIMLIN, J.A. Delivering CRISPR: a review of the challenges and approaches. Drug Delivery, 2018, 25(1): 1234.

GUHA, T.K., WAI, A., and HAUSNER, G. Programmable Genome Editing Tools and their Regulation for Efficient Genome Engineering. Computational and Structural Biotechnology Journal, 2017, 15: 146–60.

LI, H., YANG, Y., HONG, W., HUANG, M., WU, M., and ZHAO, X. Applications of genome editing technology in the targeted therapy of human diseases: mechanisms, advances, and prospects. Signal Transduction and Targeted Therapy, 2020, 5(1): 1–23.

LIU, Z., CHEN, S., JIN, X., WANG, Q., YANG, K., and LI, C. Genome editing of the HIV co-receptors CCR5 and CXCR4 by CRISPR-Cas9 protects CD4+ T cells from HIV-1 infection. Cell & Bioscience, 2017, 7(1): 47.

LIU, S., WANG, Q., YU, X., LI, Y., GUO, Y., and LIU, Z. HIV-1 inhibition in cells with CXCR4 mutant genome created by CRISPR-Cas9 and piggyBac recombinant technologies. Scientific Reports, 2018, 8(1).

YU, S., YAO, Y., XIAO, H., LI, J., LIU, Q., YANG, Y., ADAH, D., LU, J., ZHAO, S., QIN, L., and CHEN, X. Simultaneous Knockout of CXCR4 and CCR5 Genes in CD4+ T Cells via CRISPR/Cas9 Confers Resistance to Both X4- and R5-Tropic Human Immunodeficiency Virus Type 1 Infection. Human gene therapy, 2018, 29(1): 51-67.

ALLEN, A.G., CHUNG, C., ATKINS, A.J., DAMPIER, W., KHALILI, K., NONNEMACHER, M.R., and WIGDAHL, B. Gene Editing of HIV-1 Co-receptors to Prevent and/or Cure Virus Infection. Frontiers in Microbiology, 2018, 9.

ZHANG, Z., QIU, S., ZHANG, X., and CHEN, W. Optimized DNA electroporation for primary human T cell engineering. BMC Biotechnology, 2018, 18(1): 1–9.

WANG, Q., CHEN, S., XIAO, Q., LIU, Z., LIU, S., HOU, P., ZHOU, L., HOU, W., HO, W., LI, C., WU, L., and GUO, D. Genome modification of CXCR4 by Staphylococcus aureus Cas9 renders cells resistance to HIV-1 infection. Retrovirology, 2017, 14: 51.

LI, Y., LIU, D., WANG, Y., SU, W., LIU, G., and DONG, W. The Importance of Glycans of Viral and Host Proteins in Enveloped Virus Infection. Frontiers in Immunology, 2021, 12(0):1544.

Poletti, V., and Mavilio, F. Interactions between Retroviruses and the Host Cell Genome. Molecular Therapy. Methods & Clinical Development, 2018, 8: 31-41.

SEITS, R. Human Immunodeficiency Virus (HIV). Transfusion Medicine and Hemotherapy, 2016, 43(3): 203.

CHEN, B. Molecular Mechanism of HIV-1 Entry. Trends in microbiology, 2019, 27(10): 878.

KELLER, P.W., MORRISON, O., VASSELL, R.A., and WEISS, C.D. HIV-1 gp41 Residues Modulate CD4-Induced Conformational Changes in the Envelope Glycoprotein and Evolution of a Relaxed Conformation of gp120. Journal of Virology, 2018, 92(16).

NAQVI, A.A., FATIMA, K., MOHAMMAD, T., FATIMA, U., SINGH, I.K., SINGH, A., ATIF, S.M., HARIPRASAD, G., HASAN, G.M., and HASSAN, M.I. Insights into SARS-CoV-2 genome, structure, evolution, pathogenesis and therapies: Structural genomics approach. Biochimica et Biophysica Acta. Molecular Basis of Disease, 2020, 1866(10): 165878 - 165878.

WOODHAM, A.W., SKEATE, J.G., SANNA, A.M., TAYLOR, J.R., SILVA, D.M.D., CANNON, P.M., and MARTIN, K. Human Immunodeficiency Virus Immune Cell Receptors, Coreceptors, and Cofactors: Implications for Prevention and Treatment. Aids Patient Care and Stds, 2016, 30(7): 291-306.

TONG, O., DUETTE, G., O’NEIL, T.R., ROYLE, C.M., RANA, H., JOHNSON, B., POPOVIC, N., DERVISH, S., BROUWER, M.A., BAHARLOU, H., PATRICK, E., CTERCTEKO, G., PALMER, S.E., LEE, E., HUNTER, E., HARMAN, A.N., CUNNINGHAM, A.L., and NASR, N. Plasmacytoid dendritic cells have divergent effects on HIV infection of initial target cells and induce a pro-retention phenotype. PLoS Pathogens, 2021, 17(4): e1009522.

MU, W., CARRILLO, M., and KITCHEN, S. Engineering CAR T Cells to Target the HIV Reservoir. Frontiers in Cellular and Infection Microbiology, 2020, 10.

ISAGULIANTS, M.G., BAYUROVA, E., AVDOSHINA, D., KONDRASHOVA, A., CHIODI, F., and PALEFSKY, J.M. Oncogenic Effects of HIV-1 Proteins, Mechanisms Behind. Cancers, 2021, 13(2): 1–24.

PASQUEREAU, S., KUMAR, A., and HERBEIN, G. Targeting TNF and TNF Receptor Pathway in HIV-1 Infection: from Immune Activation to Viral Reservoirs. Viruses, 2017, 9(4).

WOODHAM, A.W., SKEATE, J.G., SANNA, A.M., TAYLOR, J.R., SILVA, D.M.D., CANNON, P.M., and MARTIN, K. Human Immunodeficiency Virus Immune Cell Receptors, Coreceptors, and Cofactors: Implications for Prevention and Treatment. Aids Patient Care and Stds, 2016, 30(7): 291-306.

STONE, M.J., HAYWARD, J.A., HUANG, C., HUMA, Z., and SANCHEZ, J. Mechanisms of Regulation of the Chemokine-Receptor Network. International Journal of Molecular Sciences, 2017, 18(2): 342.

COWIE, C.C., CASAGRANDE, S.S., GEISS, L.S. Prevalence and Incidence of Type 2 Diabetes and Prediabetes. Diabetes Am, 2018.

ZHAO, J., FANG, H., and ZHANG, D. Expanding application of CRISPR-Cas9 system in microorganisms. Synthetic and Systems Biotechnology, 2020, 5(4):269.

MIR, A., EDRAKI, A., LEE, J., and SONTHEIMER, E.J. Type II-C CRISPR-Cas9 Biology, Mechanism, and Application. ACS chemical biology, 2018, 13(2): 357.

GLEDITZSCH, D., PAUSCH, P., MÜLLER-ESPARZA, H., ÖZCAN, A., GUO, X., BANGE, G., and RANDAU, L. PAM identification by CRISPR-Cas effector complexes: diversified mechanisms and structures. RNA Biology, 2019, 16(4): 504-517.

LOUREIRO, A., and DA SILVA, G.J. CRISPR-Cas: Converting A Bacterial Defence Mechanism into A State-of-the-Art Genetic Manipulation Tool. Antibiotics, 2019, 8(1).

VENUTI, A., PASTORI, C., and LOPALCO, L. The Role of Natural Antibodies to CC Chemokine Receptor 5 in HIV Infection. Frontiers in Immunology, 2017, 8(OCT): 1358.

XU, M. CCR5-Δ32 biology, gene editing, and warnings for the future of CRISPR-Cas9 as a human and humane gene-editing tool. Cell & Bioscience, 2020 10(1).

LIU, Z., LIANG, J., CHEN, S., WANG, K., LIU, X., LIU, B., XIA, Y., GUO, M., ZHANG, X., SUN, G., and TIAN, G. Genome editing of CCR5 by AsCpf1 renders CD4+T cells resistance to HIV-1 infection. Cell & Bioscience, 2020, 10(1).

ERNST, M.P., BROEDERS, M., HERRERO-HERNANDEZ, P., OUSSOREN, E., VAN DER PLOEG, A.T., and PIJNAPPEL, W.W. Ready for Repair? Gene Editing Enters the Clinic for the Treatment of Human Disease. Molecular Therapy. Methods & Clinical Development, 2020, 18, 532-557.

XIAO, Q., GUO, D., and CHEN, S. Application of CRISPR/Cas9-Based Gene Editing in HIV-1/AIDS Therapy. Frontiers in Cellular and Infection Microbiology, 2019, 9(MAR): 69.

NERYS-JUNIOR, A., BRAGA-DIAS, L.P., PEZZUTO, P., COTTA-DE-ALMEIDA, V., and TANURI, A. Comparison of the editing patterns and editing efficiencies of TALEN and CRISPR-Cas9 when targeting the human CCR5 gene. Genetics and Molecular Biology, 2018, 41(1): 167-179.

CHEN, H., SCHÜRCH, C.M., NOBLE, K.E., KIM, K., KRUTZIK, P.O., O'DONNELL, E.A., VANDER TUIG, J., NOLAN, G.P., and MCILWAIN, D. Functional comparison of PBMCs isolated by Cell Preparation Tubes (CPT) vs. Lymphoprep Tubes. BMC Immunology, 2020, 21(1): 1–13.

CHICAYBAM, L., BARCELOS, C., PEIXOTO, B.C., CARNEIRO, M., LIMIA, C.G., REDONDO, P., LIRA, C., PARAGUASSÚ-BRAGA, F.H., VASCONCELOS, Z., BARROS, L.R., and BONAMINO, M.H. An Efficient Electroporation Protocol for the Genetic Modification of Mammalian Cells. Frontiers in Bioengineering and Biotechnology, 2017, 4: 99.

YU, S., YAO, Y., XIAO, H., LI, J., LIU, Q., YANG, Y., ADAH, D., LU, J., ZHAO, S., QIN, L., and CHEN, X. Simultaneous Knockout of CXCR4 and CCR5 Genes in CD4+ T Cells via CRISPR/Cas9 Confers Resistance to Both X4- and R5-Tropic Human Immunodeficiency Virus Type 1 Infection. Human gene therapy, 2018, 29(1): 51–67.

HOGAN, L.E., VÁSQUEZ, J.J., HOBBS, K.S., HANHAUSER, E.B., AGUILAR-RODRIGUEZ, B., HUSSIEN, R., THANH, C., GIBSON, E.A., CARVIDI, A.B., SMITH, L.C., KHAN, S., TRAPECAR, M., SANJABI, S., SOMSOUK, M., STODDART, C.A., KURITZKES, D.R., DEEKS, S.G., and HENRICH, T.J. Increased HIV-1 transcriptional activity and infectious burden in peripheral blood and gut-associated CD4+ T cells expressing CD30. PLoS Pathogens, 2018, 14(2): e1006856.

LIU, C., ZHANG, L., LIU, H., and CHENG, K. Delivery strategies of the CRISPR‐Cas9 gene‐editing system for therapeutic applications. Journal of Controlled Release, 2017, 266: 17–26.

MUNIS, A.M. Gene Therapy Applications of Non-Human Lentiviral Vectors. Viruses, 2020, 12(10): 1106

CAVALIERI, V., BAIAMONTE, E., and IACONO, M.L. Non-Primate Lentiviral Vectors and Their Applications in Gene Therapy for Ocular Disorders. Viruses, 2018, 10(6).

GÁNDARA, C., AFFLECK, V.S., and STOLL, E.A. Manufacture of Third-Generation Lentivirus for Preclinical Use, with Process Development Considerations for Translation to Good Manufacturing Practice. Human Gene Therapy Methods, 2018, 29(1): 1-15.

MERTEN, O.-W., HEBBEN, M., and BOVOLENTA, C. Production of lentiviral vectors. Molecular Therapy: Methods & Clinical Development, 2016, 3: 16017.

ZHENG, C., WANG, S., BAI, Y., LUO, T., WANG, J., DAI, C., GUO, B., LUO, S., WANG, D., YANG, Y., and WANG, Y. Lentiviral Vectors and Adeno‐Associated Virus Vectors: Useful Tools for Gene Transfer in Pain Research. Anatomical Record, 2018, 301(5): 825 - 836.


Refbacks

  • There are currently no refbacks.