Genetic enhancement: an avenue to combat aging-related diseases

Yusheng Cai, Zhejun Ji, Si Wang, Weiqi Zhang, Jing Qu, Juan Carlos Izpisúa-Belmonte, Guang-Hui Liu

PDF(535 KB)
PDF(535 KB)
Life Medicine ›› 2022, Vol. 1 ›› Issue (3) : 307-318. DOI: 10.1093/lifemedi/lnac054
Review
Review

Genetic enhancement: an avenue to combat aging-related diseases

Author information +
History +

Abstract

Aging is a major risk factor for multiple diseases, including cardiovascular diseases, neurodegenerative disorders, osteoarthritis, and cancer. It is accompanied by the dysregulation of stem cells and other differentiated cells, and the impairment of their microenvironment. Cell therapies to replenish the abovementioned cells provide a promising approach to restore tissue homeostasis and alleviate aging and aging-related chronic diseases. Importantly, by leveraging gene editing technologies, genetic enhancement, an enhanced strategy for cell therapy, can be developed to improve the safety and efficacy of transplanted therapeutic cells. In this review, we provide an overview and discussion of the current progress in the genetic enhancement field, including genetic modifications of mesenchymal stem cells, neural stem cells, hematopoietic stem cells, vascular cells, and T cells to target aging and aging-associated diseases. We also outline questions regarding safety and current limitations that need to be addressed for the continued development of genetic enhancement strategies for cell therapy to enable its further applications in clinical trials to combat aging-related diseases.

Keywords

genetic enhancement / gene editing / stem cell / cell therapy / aging

Cite this article

Download citation ▾
Yusheng Cai, Zhejun Ji, Si Wang, Weiqi Zhang, Jing Qu, Juan Carlos Izpisúa-Belmonte, Guang-Hui Liu. Genetic enhancement: an avenue to combat aging-related diseases. Life Medicine, 2022, 1(3): 307‒318 https://doi.org/10.1093/lifemedi/lnac054

References

[1]
Cai Y, Song W, Li J, et al. The landscape of aging. Sci China Life Sci 2022;65:2354–454
CrossRef Google scholar
[2]
Lopez-Otin C, Blasco MA, Partridge L, et al. The hallmarks of aging. Cell 2013;153:1194–217
CrossRef Google scholar
[3]
Franceschi C, Garagnani P, Parini P, et al. Inflammaging: a new immune–metabolic viewpoint for age-related diseases. Nat Rev Endocrinol 2018;14:576–90
CrossRef Google scholar
[4]
Liang C, Liu Z, Song M, et al. Stabilization of heterochromatin by CLOCK promotes stem cell rejuvenation and cartilage regeneration. Cell Res 2020;31:187–205
CrossRef Google scholar
[5]
Zhang H, Li J, Ren J, et al. Single-nucleus transcriptomic landscape of primate hippocampal aging. Protein Cell 2021;12:695–716
CrossRef Google scholar
[6]
Calimport SRG, Bentley BL, Stewart CE, et al. To help aging populations, classify organismal senescence. Science 2019;366:576–8
CrossRef Google scholar
[7]
Campisi J, Kapahi P, Lithgow GJ, et al. From discoveries in ageing research to therapeutics for healthy ageing. Nature 2019;571:183–92
CrossRef Google scholar
[8]
Leng SX, Pawelec G. Single-cell immune atlas for human aging and frailty. Life Med 2022:lnac013
CrossRef Google scholar
[9]
Aging Atlas C. Aging Atlas: a multi-omics database for aging biology. Nucleic Acids Res 2021;49:D825–30
CrossRef Google scholar
[10]
Cai Y, Wang S, Qu J, et al. Rejuvenation of tissue stem cells by intrinsic and extrinsic factors. Stem Cells Transl Med 2022b;11:231–8
CrossRef Google scholar
[11]
Zhang W, Qu J, Liu GH, et al. The ageing epigenome and its rejuvenation. Nat Rev Mol Cell Biol 2020a;21:137–50
CrossRef Google scholar
[12]
Boch J, Scholze H, Schornack S, et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 2009;326:1509–12
CrossRef Google scholar
[13]
Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013;339:819–23
CrossRef Google scholar
[14]
Jinek M, Chylinski K, Fonfara I, et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012;337:816–21
CrossRef Google scholar
[15]
Moscou MJ, Bogdanove AJ. A simple cipher governs DNA recognition by TAL effectors. Science 2009;326:1501
CrossRef Google scholar
[16]
Cui T, Li B, Li W. NTLA-2001: opening a new era for gene therapy. Life Med 2022:lnac036
CrossRef Google scholar
[17]
Maeder ML, Gersbach CA. Genome-editing technologies for gene and cell therapy. Mol Ther 2016;24:430–46
CrossRef Google scholar
[18]
Song G, Ma Y, Gao X, et al. CRISPR/Cas9-mediated genetic correction reverses spinocerebellar ataxia 3 disease-associated phenotypes in differentiated cerebellar neurons. Life Med 2022:lnac020
CrossRef Google scholar
[19]
Wilson JM. Lessons learned from the gene therapy trial for ornithine transcarbamylase deficiency. Mol Genet Metab 2009;96:151–7.
CrossRef Google scholar
[20]
Turinetto V, Vitale E, Giachino C, et al. Senescence in human mesenchymal stem cells: functional changes and implications in stem cell-based therapy. Int J Mol Sci 2016; 17:1164
CrossRef Google scholar
[21]
Schultz MB, Sinclair DA. When stem cells grow old: phenotypes and mechanisms of stem cell aging. Development 2016;143:3–14
CrossRef Google scholar
[22]
Diao Z, Ji Q, Wu Z, et al. SIRT3 consolidates heterochromatin and counteracts senescence. Nucleic Acids Res 2021;49:4203–19
CrossRef Google scholar
[23]
Salazar-Noratto GE, Luo G, Denoeud C, et al. Understanding and leveraging cell metabolism to enhance mesenchymal stem cell transplantation survival in tissue engineering and regenerative medicine applications. Stem Cells 2020;38:22–33
CrossRef Google scholar
[24]
Zhao H, Ji Q, Wu Z, et al. Destabilizing heterochromatin by APOE mediates senescence. Nature Aging 2022;2:303–16
CrossRef Google scholar
[25]
Chu Q, Liu F, He Y, et al. mTORC2/RICTOR exerts differential levels of metabolic control in human embryonic, mesenchymal and neural stem cells. Protein Cell 2022;13:676–82
CrossRef Google scholar
[26]
Pittenger MF, Discher DE, Péault BM, et al. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 2019;4:22
CrossRef Google scholar
[27]
Wang H, Jiang C, Cai J, et al. Nestin prevents mesenchymal stromal cells from apoptosis in LPS-induced lung injury via inhibition of unfolded protein response sensor IRE1α. Life Med 2022a:lnac049
CrossRef Google scholar
[28]
Zhang S, Wu Z, Shi Y, et al. FTO stabilizes MIS12 and counteracts senescence. Protein Cell 2022;13:954–60
CrossRef Google scholar
[29]
Liu GH, Suzuki K, Qu J, et al. Targeted gene correction of laminopathy-associated LMNA mutations in patient-specific iPSCs. Cell Stem Cell 2011;8:688–94
CrossRef Google scholar
[30]
Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663–76
CrossRef Google scholar
[31]
Wang F, Zhang W, Yang Q, et al. Generation of a Hutchinson–Gilford progeria syndrome monkey model by base editing. Protein Cell 2020;11:809–24
CrossRef Google scholar
[32]
Shi Y, Inoue H, Wu JC, et al. Induced pluripotent stem cell technology: a decade of progress. Nat Rev Drug Discov 2017;16:115–30
CrossRef Google scholar
[33]
Yamanaka S. Pluripotent stem cell-based cell therapy-promise and challenges. Cell Stem Cell 2020;27:523–31
CrossRef Google scholar
[34]
Liu GH, Ding Z, Izpisua Belmonte JC. iPSC technology to study human aging and aging-related disorders. Curr Opin Cell Biol 2012;24:765–74
CrossRef Google scholar
[35]
Minagawa A, Yoshikawa T, Yasukawa M, et al. Enhancing T cell receptor stability in rejuvenated iPSC-derived T cells improves their use in cancer immunotherapy. Cell Stem Cell 2018;23:850–8.e4 e854
CrossRef Google scholar
[36]
Saetersmoen ML, Hammer Q, Valamehr B, et al. Off-the-shelf cell therapy with induced pluripotent stem cell-derived natural killer cells. Semin Immunopathol 2019;41:59–68
CrossRef Google scholar
[37]
Zhang W, Qu J, Suzuki K, et al. Concealing cellular defects in pluripotent stem cells. Trends Cell Biol 2013b;23:587–92
CrossRef Google scholar
[38]
Liang Y, Lin Q, Zhu J, et al. The caspase-8 shRNA-modified mesenchymal stem cells improve the function of infarcted heart. Mol Cell Biochem 2014;397:7–16
CrossRef Google scholar
[39]
Depil S, Duchateau P, Grupp SA, et al. ‘Off-the-shelf’ allogeneic CAR T cells: development and challenges. Nat Rev Drug Discov 2020;19:185–99
CrossRef Google scholar
[40]
Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol 2020;17:147–67
CrossRef Google scholar
[41]
Amariglio N, Hirshberg A, Scheithauer BW, et al. Donor-derived brain tumor following neural stem cell transplantation in an ataxia telangiectasia patient. PLoS Med 2009;6:e1000029
CrossRef Google scholar
[42]
Maxwell KG, Millman JR. Applications of iPSC-derived beta cells from patients with diabetes. Cell Rep Med 2021;2:100238
CrossRef Google scholar
[43]
Ramzy A, Thompson DM, Ward-Hartstonge KA, et al. Implanted pluripotent stem-cell-derived pancreatic endoderm cells secrete glucose-responsive C-peptide in patients with type 1 diabetes. Cell Stem Cell 2021;28:2047–61
CrossRef Google scholar
[44]
Yan P, Li Q, Wang L, et al. FOXO3-engineered human ESC-derived vascular cells promote vascular protection and regeneration. Cell Stem Cell 2019;24:447–61.e8 e448
CrossRef Google scholar
[45]
Yang J, Li J, Suzuki K, et al. Genetic enhancement in cultured human adult stem cells conferred by a single nucleotide recoding. Cell Res 2017;27:1178–81
CrossRef Google scholar
[46]
Bianco P, Robey G, Simmons PJ. Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell 2008;2:313–9.
CrossRef Google scholar
[47]
Caplan AI. Mesenchymal stem cells. J Orthop Res 1991;9:641–50.
CrossRef Google scholar
[48]
Bianco P. “Mesenchymal” stem cells. Annu Rev Cell Dev Biol 2014;30:677–704
CrossRef Google scholar
[49]
Shahror RA, Wu CC, Chiang YH, et al. Genetically modified mesenchymal stem cells: the next generation of stem cell-based therapy for TBI. Int J Mol Sci 2020;21:4051
CrossRef Google scholar
[50]
Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 1999;96:857–68
CrossRef Google scholar
[51]
Gnecchi M, He H, Noiseux N, et al. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J 2006;20:661–9
CrossRef Google scholar
[52]
Mangi AA, Noiseux N, Kong D, et al. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med 2003;9:1195–201
CrossRef Google scholar
[53]
Shibata T, Ismailoglu UB, Kittan NA, et al. Role of growth arrest-specific gene 6 in the development of fungal allergic airway disease in mice. Am J Respir Cell Mol Biol 2014;51:615–25
CrossRef Google scholar
[54]
Shan S, Liu Z, Guo T, et al. Growth arrest-specific gene 6 transfer promotes mesenchymal stem cell survival and cardiac repair under hypoxia and ischemia via enhanced autocrine signaling and paracrine action. Arch Biochem Biophys 2018;660:108–20
CrossRef Google scholar
[55]
Osyczka AM, Diefenderfer DL, Bhargave G, et al. Different effects of BMP-2 on marrow stromal cells from human and rat bone. Cells Tissues Organs 2004;176:109–19
CrossRef Google scholar
[56]
Salazar VS, Gamer LW, Rosen V. BMP signalling in skeletal development, disease and repair. Nat Rev Endocrinol 2016;12:203–21
CrossRef Google scholar
[57]
Ding HF, Liu R, Li BG, et al. Biologic effect and immunoisolating behavior of BMP-2 gene-transfected bone marrow-derived mesenchymal stem cells in APA microcapsules. Biochem Biophys Res Commun 2007;362:923–7
CrossRef Google scholar
[58]
Shibuya M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies. Genes Cancer 2011;2:1097–105
CrossRef Google scholar
[59]
Tello-Montoliu A, Patel JV, Lip GYH. Angiogenin: a review of the pathophysiology and potential clinical applications. J Thromb Haemost 2006;4:1864–74
CrossRef Google scholar
[60]
Garcia KO, Ornellas FL, Martin PK, et al. Therapeutic effects of the transplantation of VEGF overexpressing bone marrow mesenchymal stem cells in the hippocampus of murine model of Alzheimer’s disease. Front Aging Neurosci 2014;6:30
CrossRef Google scholar
[61]
Kim SH, Moon H-H, Kim HA, et al. Hypoxia-inducible vascular endothelial growth factor-engineered mesenchymal stem cells prevent myocardial ischemic injury. Mol Ther 2011;19:741–50
CrossRef Google scholar
[62]
Shi D, Chen G, Lv L, et al. The effect of lentivirus-mediated TH and GDNF genetic engineering mesenchymal stem cells on Parkinson’s disease rat model. Neurol Sci 2011;32:41–51
CrossRef Google scholar
[63]
Ma Q. Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol 2013;53:401–26
CrossRef Google scholar
[64]
Zhang W, Zhang S, Yan P, et al. A single-cell transcriptomic landscape of primate arterial aging. Nat Commun 2020b;11:2202
CrossRef Google scholar
[65]
Lei J, Wang S, Kang W, et al. FOXO3-engineered human mesenchymal progenitor cells efficiently promote cardiac repair after myocardial infarction. Protein Cell 2021;12:145–51
CrossRef Google scholar
[66]
Mattson MP, Magnus T. Ageing and neuronal vulnerability. Nat Rev Neurosci 2006;7:278–94
CrossRef Google scholar
[67]
Hou Y, Dan X, Babbar M, et al. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol 2019;15:565–81
CrossRef Google scholar
[68]
Lee I-S, Jung K, Kim I-Set al. Human neural stem cells alleviate Alzheimer-like pathology in a mouse model. Mol Neurodegener 2015;10:1–16
CrossRef Google scholar
[69]
Mendes-Pinheiro B, Teixeira FG, Anjo SI, et al. Secretome of undifferentiated neural progenitor cells induces histological and motor improvements in a rat model of Parkinson’s disease. Stem Cells Transl Med 2018;7:829–38
CrossRef Google scholar
[70]
Wu C-C, Lien C-C, Hou W-H, et al. Gain of BDNF function in engrafted neural stem cells promotes the therapeutic potential for Alzheimer’s disease. Sci Rep 2016;6:1–16
CrossRef Google scholar
[71]
Ohnishi K, Semi K, Yamamoto T, et al. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell 2014;156:663–77
CrossRef Google scholar
[72]
Wagoner ZW, Zhao W. Therapeutic implications of transplanted-cell death. Nat Biomed Eng 2021;5:379–84
CrossRef Google scholar
[73]
Korshunova I, Rhein S, García-González D, et al. Genetic modification increases the survival and the neuroregenerative properties of transplanted neural stem cells. JCI Insight 2020;5:e126268
CrossRef Google scholar
[74]
Li X, Peng Z, Long L, et al. Transplantation of Wnt5a-modified NSCs promotes tissue repair and locomotor functional recovery after spinal cord injury. Exp Mol Med 2020;52:2020–33
CrossRef Google scholar
[75]
Dacks PA, Moreno CL, Kim ES, et al. Role of the hypothalamus in mediating protective effects of dietary restriction during aging. Front Neuroendocrinol 2013;34:95–106
CrossRef Google scholar
[76]
Satoh A, Brace CS, Rensing N, et al. Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab 2013a;18:416–30
CrossRef Google scholar
[77]
Zhang G, Li J, Purkayastha S, et al. Hypothalamic programming of systemic ageing involving IKK-β, NF-κB and GnRH. Nature 2013a;497:211–6
CrossRef Google scholar
[78]
Zhang Y, Kim MS, Jia B, et al. Hypothalamic stem cells control ageing speed partly through exosomal miRNAs. Nature 2017;548:52–7
CrossRef Google scholar
[79]
Sadagurski M, Landeryou T, Cady G, et al. Transient early food restriction leads to hypothalamic changes in the long-lived crowded litter female mice. Physiol Rep 2015;3:e12379
CrossRef Google scholar
[80]
Dampney R. Functional organization of central pathways regulating the cardiovascular system. Physiol Rev 1994;74:323–64
CrossRef Google scholar
[81]
Ferrari AU, Radaelli A, Centola M. Invited review: aging and the cardiovascular system. J Appl Physiol 2003;95:2591–7
CrossRef Google scholar
[82]
Baumgartner I, Pieczek A, Manor O, et al. Constitutive expression of phVEGF165 after intramuscular gene transfer promotes collateral vessel development in patients with critical limb ischemia. Circulation 1998;97:1114–23
CrossRef Google scholar
[83]
Bersini S, Schulte R, Huang L, et al. Direct reprogramming of human smooth muscle and vascular endothelial cells reveals defects associated with aging and Hutchinson-Gilford progeria syndrome. Elife 2020;9:e54383
CrossRef Google scholar
[84]
Gill M, Dias S, Hattori K, et al. Vascular trauma induces rapid but transient mobilization of VEGFR2+ AC133+ endothelial precursor cells. Circ Res 2001;88:167–74
CrossRef Google scholar
[85]
Kalka C, Masuda H, Takahashi T, et al. Vascular endothelial growth factor165 gene transfer augments circulating endothelial progenitor cells in human subjects. Circ Res 2000;86:1198–202
CrossRef Google scholar
[86]
Lacolley P, Regnault V, Avolio AP. Smooth muscle cell and arterial aging: basic and clinical aspects. Cardiovasc Res 2018;114:513–28
CrossRef Google scholar
[87]
van Bussel BC, Schouten F, Henry RM, et al. Endothelial dysfunction and low-grade inflammation are associated with greater arterial stiffness over a 6-year period. Hypertension 2011;58:588–95
CrossRef Google scholar
[88]
Morris BJ, Willcox DC, Donlon TA, et al. FOXO3: a major gene for human longevity-a mini-review. Gerontology 2015;61:515–25
CrossRef Google scholar
[89]
Kang W, Jin T, Zhang T, et al. Regeneration Roadmap: database resources for regenerative biology. Nucleic Acids Res 2022;50:D1085–90
[90]
Denkinger MD, Leins H, Schirmbeck R, et al. HSC aging and senescent immune remodeling. Trends Immunol 2015;36:815–24
CrossRef Google scholar
[91]
Liu B, Qu J, Zhang W, et al. A stem cell aging framework, from mechanisms to interventions. Cell Rep 2022a;41:111451
CrossRef Google scholar
[92]
Rossi DJ, Bryder D, Weissman IL. Hematopoietic stem cell aging: mechanism and consequence. Exp Gerontol 2007;42:385–90
CrossRef Google scholar
[93]
Bianchi-Frias D, Damodarasamy M, Hernandez SA, et al. The aged microenvironment influences the tumorigenic potential of malignant prostate epithelial cells. Mol Cancer Res 2019;17:321–31
CrossRef Google scholar
[94]
Maloberti A, Vallerio P, Triglione N, et al. Vascular aging and disease of the large vessels: role of inflammation. High Blood Press Cardiovasc Prev 2019;26:175–82
CrossRef Google scholar
[95]
Akunuru S, Geiger H. Aging, clonality, and rejuvenation of hematopoietic stem cells. Trends Mol Med 2016;22:701–12
CrossRef Google scholar
[96]
Wahlestedt M, Pronk CJ, Bryder D. Concise review: hematopoietic stem cell aging and the prospects for rejuvenation. Stem Cells Transl Med 2015;4:186–94
CrossRef Google scholar
[97]
Moss P, Rickinson A. Cellular immunotherapy for viral infection after HSC transplantation. Nat Rev Immunol 2005;5:9–20
CrossRef Google scholar
[98]
Rožman P. The potential of non-myeloablative heterochronous autologous hematopoietic stem cell transplantation for extending a healthy life span. Geroscience 2018;40:221–42
CrossRef Google scholar
[99]
Malmegrim KCR, Lima-Júnior JR, Arruda LCM, et al. Autologous hematopoietic stem cell transplantation for autoimmune diseases: from mechanistic insights to biomarkers. Front Immunol 2018;9:2602
CrossRef Google scholar
[100]
Steinberg MH. Sickle cell anemia, the first molecular disease: overview of molecular etiology, pathophysiology, and therapeutic approaches. Sci World J 2008;8:1295–324
CrossRef Google scholar
[101]
Demirci S, Zeng J, Wu Y, et al. BCL11A enhancer–edited hematopoietic stem cells persist in rhesus monkeys without toxicity. J Clin Invest 2020;130:6677–87
CrossRef Google scholar
[102]
Brown K, Xie S, Qiu X, et al. SIRT3 reverses aging-associated degeneration. Cell Rep 2013;3:319–27
CrossRef Google scholar
[103]
Mohrin M, Shin J, Liu Y, et al. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science 2015;347:1374–7
CrossRef Google scholar
[104]
Satoh Y, Yokota T, Sudo T, et al. The Satb1 protein directs hematopoietic stem cell differentiation toward lymphoid lineages. Immunity 2013b;38:1105–15
CrossRef Google scholar
[105]
Agrelo R, Souabni A, Novatchkova M, et al. SATB1 defines the developmental context for gene silencing by Xist in lymphoma and embryonic cells. Dev Cell 2009;16:507–16
CrossRef Google scholar
[106]
Ma S, Wang S, Ye Y, et al. Heterochronic parabiosis induces stem cell revitalization and systemic rejuvenation across aged tissues. Cell Stem Cell 2022;29:990–1005.e10 e1010
CrossRef Google scholar
[107]
Wang L, Liu J, Liu H, et al. The secret of youth—how is systemic rejuvenation achieved at the single cell level? Life Med 2022b:lnac018
CrossRef Google scholar
[108]
Nicholson LB. The immune system. Essays Biochem 2016;60: 275–301
CrossRef Google scholar
[109]
Brentjens RJ, Santos E, Nikhamin Y, et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin Cancer Res 2007;13:5426–35
CrossRef Google scholar
[110]
Kershaw MH, Westwood JA, Parker LL, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 2006;12:6106–15
CrossRef Google scholar
[111]
Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci USA 1989;86:10024–8
CrossRef Google scholar
[112]
Di Stasi A, De Angelis B, Rooney CM, et al. T lymphocytes coexpressing CCR4 and a chimeric antigen receptor targeting CD30 have improved homing and antitumor activity in a Hodgkin tumor model. Blood 2009;113:6392–402
CrossRef Google scholar
[113]
Hong M, Clubb JD, Chen YY. Engineering CAR-T cells for next-generation cancer therapy. Cancer Cell 2020;38:473–88
CrossRef Google scholar
[114]
Milone MC, Fish JD, Carpenito C, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther 2009;17:1453–64
CrossRef Google scholar
[115]
Maude SL, Teachey DT, Porter DL, et al. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 2015;125:4017–23
CrossRef Google scholar
[116]
Bailey SR, Maus MV. Gene editing for immune cell therapies. Nat Biotechnol 2019;37:1425–34
CrossRef Google scholar
[117]
Rafiq S, Yeku OO, Jackson HJ, et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat Biotechnol 2018;36:847–56
CrossRef Google scholar
[118]
Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancer therapy. Nat Rev Cancer 2013;13:525–41
CrossRef Google scholar
[119]
Roybal KT, Williams JZ, Morsut L, et al. Engineering T cells with customized therapeutic response programs using synthetic notch receptors. Cell 2016;167:419–32.e16
CrossRef Google scholar
[120]
Suarez ER, Chang DK, Sun J, et al. Chimeric antigen receptor T cells secreting anti-PD-L1 antibodies more effectively regress renal cell carcinoma in a humanized mouse model. Oncotarget 2016;7:34341–55
CrossRef Google scholar
[121]
Chinnasamy D, Yu Z, Kerkar SP, et al. Local delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice. Clin Cancer Res 2012;18:1672–83
CrossRef Google scholar
[122]
Rockey DC, Bell D, Hill JA. Fibrosis—a common pathway to organ injury and failure. N Engl J Med 2015;372:1138–49
CrossRef Google scholar
[123]
Weiskirchen R, Weiskirchen S, Tacke F. Organ and tissue fibrosis: molecular signals, cellular mechanisms and translational implications. Mol Aspects Med 2019;65:2–15
CrossRef Google scholar
[124]
Aghajanian H, Kimura T, Rurik JG, et al. Targeting cardiac fibrosis with engineered T cells. Nature 2019;573:430–3
CrossRef Google scholar
[125]
Tillmanns J, Hoffmann D, Habbaba Y, et al. Fibroblast activation protein alpha expression identifies activated fibroblasts after myocardial infarction. J Mol Cell Cardiol 2015;87:194–203
CrossRef Google scholar
[126]
Sun Y, Li Q, Kirkland JL. Targeting senescent cells for a healthier longevity: the roadmap for an era of global aging. Life Med 2022:lnac030
CrossRef Google scholar
[127]
van Deursen JM. The role of senescent cells in ageing. Nature 2014;509:439–46
CrossRef Google scholar
[128]
Cai Y, Zhou H, Zhu Y, et al. Elimination of senescent cells by beta-galactosidase-targeted prodrug attenuates inflammation and restores physical function in aged mice. Cell Res 2020;30:574–89
CrossRef Google scholar
[129]
Amor C, Feucht J, Leibold J, et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 2020;583:127–32
CrossRef Google scholar
[130]
Wei W, Huang Y, Li D, et al. Improved therapeutic potential of MSCs by genetic modification. Gene Ther 2018;25:538–47
CrossRef Google scholar
[131]
Liu G, Lin Q, Jin S, et al. The CRISPR-Cas toolbox and gene editing technologies. Mol Cell 2022b;82:333–47.
CrossRef Google scholar
[132]
Uddin F, Rudin CM, Sen T. CRISPR gene therapy: applications, limitations, and implications for the future. Front Oncol 2020;10:1387
CrossRef Google scholar
[133]
Asmamaw Mengstie M. Viral vectors for the in vivo delivery of CRISPR components: advances and challenges. Front Bioeng Biotechnol 2022;10:895713
CrossRef Google scholar
[134]
Glass Z, Lee M, Li Y, et al. Engineering the delivery system for CRISPR-based genome editing. Trends Biotechnol 2018;36: 173–85
CrossRef Google scholar
[135]
Patil S, Gao YG, Lin X, et al. The development of functional non-viral vectors for gene delivery. Int J Mol Sci 2019;20:5491
CrossRef Google scholar
[136]
Ramamoorth M, Narvekar A. Non viral vectors in gene therapy- an overview. J Clin Diagn Res 2015;9:Ge01–06
CrossRef Google scholar
[137]
Mansouri M, Fussenegger M. Therapeutic cell engineering: designing programmable synthetic genetic circuits in mammalian cells. Protein Cell 2022;13:476–89
CrossRef Google scholar

RIGHTS & PERMISSIONS

2022 The Author(s) 2022. Published by Oxford University Press on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(535 KB)

Accesses

Citations

Detail

Sections
Recommended

/