MOLECULAR THERAPY

In situ blockade of TNF-TNFR2 axis via oncolytic adenovirus improves antitumor efficacy in solid tumors
Kang X, Han Y, Wu M, Li Y, Qian P, Xu C, Zou Z, Dong J and Wei J
RNA and epigenetic editing: The new frontier in gene and cell therapy
Avila-Moreno F and Giangrande PH
A promising advance using oncolytic adenovirus to locally block tumorigenic TNF signaling
Zhang Y and Wang P
Transforming TGF-β suppression into IL-15 stimulation: Advancing engineered CAR-T therapy for solid tumors
Seo H
Vitronectin regulates lung tissue remodeling and emphysema in chronic obstructive pulmonary disease
Liu G, Hsu AC, Geirnaert S, Cong C, Nair PM, Shen SS, Marshall J, Haw TJ, Fricker M, Philp AM, Hansbro NG, Pavlidis S, Guo Y, Burgess JK, Castellano L, Ieni A, Caramori G, Oliver BG, Chung KF, Adcock IM, Knight DA, Polverino F, Bracke K, Wark PA and Hansbro PM
Vitronectin (VTN) is an important extracellular matrix protein in tissue remodeling, but its role in COPD is unknown. We show that VTN regulates tissue remodeling through urokinase plasminogen activator (uPA) signaling pathway in COPD. In human COPD airways and bronchoepithelial cells and the airways of mice with cigarette smoke (CS)-induced experimental COPD, VTN protein was not changed, but downstream uPA signaling was altered (increased plasminogen activator inhibitor-1, uPAR) that induced collagen and airway remodeling. In the parenchyma, VTN levels were decreased, uPA signalling pathway differentially altered and collagen reduced in lung fibroblasts from human and lung parenchyma in experimental COPD. Vtn inhibition with siRNA in mouse fibroblasts altered uPA signalling increased matrix metalloproteinase-12, and reduced collagen, whereas over-expression restored collagen production after CS extract challenge. Vtn and Vtn siRNA-treated mice had exaggerated inflammation, emphysema and impaired lung function compared to controls with CS-induced COPD. Restoration of VTN in the parenchyma may be a therapeutic option for emphysema and COPD.
PBAE-PEG based lipid nanoparticles for lung cell-specific gene delivery
Liu B, Sajiki Y, Littlefield N, Hu Y, Stuart WD, Sridharan A, Cui X, Siefert ME, Araki K, Ziady AG, Shi D, Whitsett JA and Yutaka M
Exemplified by successful use in COVID-19 vaccination, delivery of modified mRNA encapsulated in lipid nanoparticles provides a framework for treating various genetic and acquired disorders. However, lipid nanoparticles that can deliver mRNA into specific lung cell types have not yet been established. Here, we sought whether poly(®-amino ester)s (PBAE) or PEGylated PBAE (PBAE-PEG) in combination with 4A3-SC8/DOPE/cholesterol/DOTAP lipid nanoparticles (LNP) could deliver mRNA into different types of lung cells in vivo. PBAE-PEG/LNP was similar to Lipofectamine MessengerMAX followed by PBAE/LNP for mRNA transfection efficiency in HEK293T cells in vitro. PBAE-PEG/LNP administered by intravenous (IV) injection achieved 55% mRNA transfection efficiency into lung endothelial cells while PBAE-PEG/LNP administered by intratracheal (IT) injection achieved 73% efficiency into lung alveolar type II (ATII) epithelial cells in mice in vivo. PBAE/LNP administered by IT injection were superior for specific delivery into lung airway club epithelial cells compared to PBAE-PEG/LNP. Lipofectamine MessengerMAX was inactive in vivo. 5-methoxyuridine modified (5moU) mRNA was more efficient than unmodified mRNA in vivo but not in vitro. Our findings indicate that PBAE-PEG/LNP and PBAE/LNP can transfect multiple lung cell types in vivo, which can be applied for gene therapy targeting genetic lung diseases.
A Comprehensive Atlas of AAV Tropism in the Mouse
Walkey CJ, Snow KJ, Bulcha J, Cox AR, Martinez AE, Ljungberg MC, Lanza DG, De Giorgi M, Chuecos MA, Alves-Bezerra M, Suarez CF, Hartig SM, Hilsenbeck SG, Hsu CW, Saville E, Gaitan Y, Duryea J, Hannigan S, Dickinson ME, Mirochnitchenko O, Wang D, Lutz CM, Heaney JD, Gao G, Murray SA and Lagor WR
Gene therapy with Adeno-Associated Virus (AAV) vectors requires knowledge of their tropism within the body. Here we analyze the tropism of ten naturally occurring AAV serotypes (AAV3B, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAVrh8, AAVrh10 and AAVrh74) following systemic delivery into male and female mice. A transgene expressing ZsGreen and Cre recombinase was used to identify transduction in a cell-dependent manner based on fluorescence. Cre-driven activation of tdTomato fluorescence offered superior sensitivity for transduced cells. All serotypes except AAV3B and AAV4 had high liver tropism. Fluorescence activation revealed transduction of unexpected tissues, including adrenals, testes and ovaries. Rare transduced cells within tissues were also readily visualized. Biodistribution of AAV genomes correlated with fluorescence, except in immune tissues. AAV4 was found to have a pan-endothelial tropism while also targeting pancreatic beta cells. This public resource enables selection of the best AAV serotypes for basic science and preclinical applications in mice.
Low-inflammatory lipid nanoparticles facilitate safe mRNA vaccination against influenza virus infection
Xu Y, Zhai G, Yang DP and Chen G
Tissue nanotransfection-based endothelial PLCγ2-targeted epigenetic gene editing in vivo rescues perfusion and diabetic ischemic wound healing
Verma SS, Sen CK, Srivastava R, Gnyawali SC, Katiyar P, Sahi AK, Kumar M, Rustagi Y, Liu S, Pandey D, Abouhashem AS, Fehme LN, Kacar S, Mohanty SK, Faden-McCormack J, Murphy MP, Roy S, Wan J, Yoder MC and Singh K
Diabetic wounds are complicated by underlying peripheral vasculopathy. Reliance on vascular endothelial growth factor (VEGF) therapy to improve perfusion makes logical sense, yet clinical study outcomes on rescuing diabetic wound vascularization have yielded disappointing results. Our previous work has identified that low endothelial phospholipase Cγ2 (PLCγ2) expression hinders the therapeutic effect of VEGF on the diabetic ischemic limb. In this work, guided by single-cell RNA sequencing of human wound-edge, we test the efficacy of gene-targeted therapeutic demethylation intending to improve VEGF-mediated neovascularization. PLCγ2 expression was diminished in all five identified diabetic wound-edge endothelial subclusters encompassing arterial, venous, and capillary cells. Such low expression was associated with hypermethylated PLCγ2 promoter. PLCγ2 promoter was also hypermethylated at murine diabetic ischemic wound-edge. To specifically demethylate endothelial PLCγ2 promoter during VEGF therapy, a CRISPR/dCas9-based demethylation cocktail was delivered to the ischemic wound-edge using tissue nanotransfection (TNT) technology. Demethylation-based upregulation of PLCγ2 during VEGF therapy improved wound tissue blood flow with an increased abundance of vWF/PLCγ2 vascular tissue elements by activating p44/p42-MAPK→HIF1α pathway. Taken together, TNT-based delivery of plasmids to demethylate the PLCγ2 gene promoter activity led to significant improvements in VEGF therapy for cutaneous diabetic wounds, resulting in better perfusion and accelerated wound closure.
Pharmacological blocking of microfibrillar-associated protein 4 reduces retinal neoangiogenesis and vascular leakage
Schlosser A, Pilecki B, Allen C, Benest AV, Lynch AP, Hua J, Ved N, Blackley Z, Andersen TL, Hennig D, Graversen JH, Möller S, Skallerup S, Ormhøj M, Lange C, Agostini HT, Grauslund J, Heegaard S, Dacheva I, Koss M, Hu W, Iglesias B, Lawrence MS, Beck HC, Steffensen LB, Laursen NS, Andersen GR, Holmskov U, Bates DO and Sorensen GL
Neovascular age-related macular degeneration and diabetic macular edema are leading causes of vision-loss evoked by retinal neovascularization and vascular leakage. The glycoprotein microfibrillar-associated protein 4 (MFAP4) is an integrin αβ ligand present in the extracellular matrix. Single-cell transcriptomics reveal MFAP4 expression in cell-types in close proximity to vascular endothelial cells including choroidal vascular mural cells and retinal astrocytes and Müller cells. Binding of the anti-MFAP4 antibody, hAS0326, makes MFAP4 inaccessible for integrin receptor interaction and thereby hAS0326 blocked endothelial cell motility in vitro. Intravitreal hAS0326 inhibited retinal vascular lesion area and neovessel volume in a laser-induced choroidal neovascularization mouse model, vascular permeability in streptozotocin-induced retinopathy and vascular leakage area in a chronic non-human primate model of DL-2-aminoadipic acid-induced retinopathy. One dose of hAS0326 showed duration of efficacy of at least 12 weeks in the latter model. Moreover, hAS0326-treatment significantly enriched gene ontology terms involving reduction of integrin binding. Our data suggest that hAS0326 constitutes a promising treatment of neovascularization and vascular leakage in retinal diseases.
Engineering Resilient CAR T Cells for Immunosuppressive Environment
Khalifeh M and Salman H
Chimeric Antigen Receptor (CAR) T cell therapy has revolutionized cancer treatment and is now being explored for other diseases, such as autoimmune disorders. While the tumor microenvironment (TME) in cancer is often immunosuppressive, in autoimmune diseases, the environment is typically inflammatory. Both environments can negatively impact CAR T cell survival: the former through direct suppression, hypoxia, and nutrient deprivation, and the latter through chronic T cell receptor (TCR) engagement, risking exhaustion. Mechanisms of resistance include T cell exhaustion, dysfunction, and the impact of the TME. Chronic antigenic stimulation leads to CAR T cell exhaustion. CAR construct design, including co-stimulatory domains, hinge, transmembrane regions, promoters, the affinity of the binder site and on /off rate plays a crucial role in modulating CAR T cell function and resistance. This review discusses the impact of the in vitro development of CAR T cells, albeit in relation to the TME, on therapeutic outcomes. The use of alternative cell sources, multi-antigen targeting, and re-engineering the TME, are discussed. The review emphasizes the need for continued innovation in CAR T cell design and manufacturing to optimize therapeutic efficacy and durability, especially in the face of varying environmental challenges.
TRAF1 promotes osteoclastogenesis by enhancing metabolic adaptation to oxidative phosphorylation in an AKT-dependent manner
Kang H, Peng R, Dong Y, Liao F, Zhu M, Wang P, Hu SA, Hu P, Wang J, Liu Z, Song K and Li F
Tumor necrosis factor receptor-associated factor 1 (TRAF1) is a crucial signaling adaptor involved in multiple cellular events. However, its role in regulating osteoclastogenesis and energy metabolism remains unclear. Here, we report that TRAF1 promotes osteoclastogenesis and oxidative phosphorylation (OXPHOS). Employing RNA-sequencing, we found that TRAF1 is markedly upregulated during osteoclastogenesis and is positively associated with osteoporosis. TRAF1 knockout inhibits osteoclastogenesis and increases bone mass in both normal and ovariectomized adult mice, without affecting bone mass in childhood. Furthermore, TRAF1 promotes osteoclast OXPHOS by increasing the phosphorylation level of AKT. Mechanistically, TRAF1 functions to inhibit TRAF2-induced ubiquitination of Gβl, a known activator of AKT, and further upregulates AKT phosphorylation. Rescue experiments revealed that the inhibitory effects of TRAF1 knockout on osteoclastogenesis, OXPHOS, and bone mass are dependent on AKT. Collectively, our findings uncover a previously unrecognized function of TRAF1 in regulating osteoclastogenesis and energy metabolism, and establish a novel TRAF1-AKT-OXPHOS axis in osteoclasts.
CDK1-loaded extracellular vesicles promote cell cycle to reverse impaired wound healing in diabetic obese mice
Choi W, Park DJ, Dorschner RA, Nakatsutsumi K, Yi M and Eliceiri BP
Small extracellular vesicles (sEVs) mediate intercellular signaling to coordinate proliferation of cell types that promote re-epithelialization of skin following injury. Cyclin-dependent kinase 1 (CDK1) drives cell division and is a key regulator of entry to cell cycle. To understand the potential of sEV-mediated delivery of CDK1 to reverse impaired wound healing, we generated CDK1-loaded sEVs (CDK1-sEVs) and evaluated their ability to mediate cell proliferation, re-epithelialization and downstream signaling responses in the wound bed. We found that treatment of human keratinocytes with CDK1-sEVs increased phosphorylation of the CDK1 target, eukaryotic translation inhibition factor 4E-binding protein 1 (4E-BP1), and histone H3 within 24 hours via AKT and ERK phosphorylation, driving increased proliferation and cell migration. Treatment of the wound bed of diabetic obese mice, a model of delayed wound healing, with a single dose of CDK1-sEVs accelerated wound closure, increased re-epithelialization, and promoted proliferation of keratinocytes. These studies show that delivery of CDK1 by sEVs can stimulate selective and transient proliferation of cell types that increase re-epithelialization and promote proliferation of keratinocytes to accelerate wound healing.
Promotion or inhibition? This is a question in gene editing
Tsai LK, Han R, Yang D, Chen YE, Zhang J and Xu J
GITRL enhances cytotoxicity and persistence of CAR-T cells in cancer therapy
Tan B, Tu C, Xiong H, Xu Y, Shi X, Zhang X, Yang R, Zhang N, Lin B, Liu M, Qin J and Du B
CAR T-cell therapy has achieved remarkable clinical success in treating hematological malignancies. However, its clinical efficacy in solid tumors is less satisfactory, partially due to poor in vivo expansion and limited persistence of CAR-T cells. Here, we demonstrated that the overexpression of glucocorticoid-induced tumor necrosis factor receptor-related protein ligand (GITRL) enhances the anti-tumor activity of CAR-T cells. Compared to PSMA-BB-Z CAR-T, PSMA-BB-Z-GITRL CAR-T cells have much more IFN-γ, TNF-α, and IL-9 secretion, a higher proportion of central memory T (T) cells and Th9 cells, less expression of exhaustion markers, and robust proliferation capacity. Consequently, PSMA-BB-Z-GITRL CAR-T cells exhibited more potent anti-tumor activity against established solid tumors in vivo than PSMA-BB-Z CAR-T cells. The results of in vivo persistence experiment also indicated that PSMA-BB-Z-GITRL CAR-T cells exhibited much more retention in mouse blood, spleen, and tumor tissue than PSMA-BB-Z CAR-T cells 15 days after CAR-T cell therapy. In addition, PSMA-BB-Z-GITRL CAR-T cells produce higher levels of IFN-γ, TNF-α and IL-9 in mouse blood, exhibiting a higher proportion of T cells and a lower proportion of Treg cells compared to PSMA-BB-Z CAR-T cells. Our results demonstrate that the overexpression of GITRL has important implications for improving CAR-T cell-based human solid tumor immunotherapy.
SHIV fights back to evade destruction by eCD4-Ig but not without suffering debilitating wounds
Arts EJ
A path toward developing a universal mucosal influenza vaccine: An upside-down influenza HA vaccine based on extracellular vesicles
Nejabat S, Adloo Z and Nezafat N
Orchestrating cytosolic access: The partnership of cationic lytic peptide L17E and potassium channel KCa3.1
Parang K
Gene therapy in China: From a clinician's perspective
Wang X and Peng J
Treating genetic blood disorders in the era of CRISPR-mediated genome editing
Almotiri A, Abogosh A, Abdelfattah A, Alowaisy D and Rodrigues NP
In the setting of monogenic disease, advances made in genome editing technologies can, in principle, be deployed as a therapeutic strategy to precisely correct a specific gene mutation in an affected cell type and restore functionality. Using the β-hemoglobinopathies and hemophilia as exemplars, we review recent experimental breakthroughs utilizing CRISPR-derived genome editing technology that have translated to significant improvements in the management of inherited hematologic disorders. Yet there are also challenges facing the use of CRISPR mediated genome editing in these patients and we discuss possible ways to obviate those issues for furtherance of clinical benefit.
Rescue from Stx2-Producing E. coli-Associated Encephalopathy by Intravenous Injection of Muse Cells in NOD-SCID Mice
Ozuru R, Wakao S, Tsuji T, Ohara N, Matsuba T, Amran MY, Isobe J, Iino M, Nishida N, Matsumoto S, Iwadate K, Konishi N, Yasuda K, Tashiro K, Hida M, Yadoiwa A, Kato S, Yamashita E, Matsumoto S, Kurozawa Y, Dezawa M and Fujii J