16 results on '"Muhuri M"'
Search Results
2. Divalent siRNAs are bioavailable in the lung and efficiently block SARS-CoV-2 infection.
- Author
-
Hariharan VN, Shin M, Chang CW, O'Reilly D, Biscans A, Yamada K, Guo Z, Somasundaran M, Tang Q, Monopoli K, Krishnamurthy PM, Devi G, McHugh N, Cooper DA, Echeverria D, Cruz J, Chan IL, Liu P, Lim SY, McConnell J, Singh SP, Hildebrand S, Sousa J, Davis SM, Kennedy Z, Ferguson C, Godinho BMDC, Thillier Y, Caiazzi J, Ly S, Muhuri M, Kelly K, Humphries F, Cousineau A, Parsi KM, Li Q, Wang Y, Maehr R, Gao G, Korkin D, McDougall WM, Finberg RW, Fitzgerald KA, Wang JP, Watts JK, and Khvorova A
- Subjects
- Humans, Animals, Mice, RNA, Small Interfering genetics, SARS-CoV-2 genetics, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Oligonucleotides, Lung, COVID-19 therapy
- Abstract
The continuous evolution of SARS-CoV-2 variants complicates efforts to combat the ongoing pandemic, underscoring the need for a dynamic platform for the rapid development of pan-viral variant therapeutics. Oligonucleotide therapeutics are enhancing the treatment of numerous diseases with unprecedented potency, duration of effect, and safety. Through the systematic screening of hundreds of oligonucleotide sequences, we identified fully chemically stabilized siRNAs and ASOs that target regions of the SARS-CoV-2 genome conserved in all variants of concern, including delta and omicron. We successively evaluated candidates in cellular reporter assays, followed by viral inhibition in cell culture, with eventual testing of leads for in vivo antiviral activity in the lung. Previous attempts to deliver therapeutic oligonucleotides to the lung have met with only modest success. Here, we report the development of a platform for identifying and generating potent, chemically modified multimeric siRNAs bioavailable in the lung after local intranasal and intratracheal delivery. The optimized divalent siRNAs showed robust antiviral activity in human cells and mouse models of SARS-CoV-2 infection and represent a new paradigm for antiviral therapeutic development for current and future pandemics.
- Published
- 2023
- Full Text
- View/download PDF
3. Durability of transgene expression after rAAV gene therapy.
- Author
-
Muhuri M, Levy DI, Schulz M, McCarty D, and Gao G
- Subjects
- Animals, Gene Transfer Techniques, Genetic Therapy, Humans, Transgenes, Dependovirus genetics, Genetic Vectors genetics
- Abstract
Recombinant adeno-associated virus (rAAV) gene therapy has the potential to transform the lives of patients with certain genetic disorders by increasing or restoring function to affected tissues. Following the initial establishment of transgene expression, it is unknown how long the therapeutic effect will last, although animal and emerging human data show that expression can be maintained for more than 10 years. The durability of therapeutic response is key to long-term treatment success, especially since immune responses to rAAV vectors may prevent re-dosing with the same therapy. This review explores the non-immunological and immunological processes that may limit or improve durability and the strategies that can be used to increase the duration of the therapeutic effect., Competing Interests: Declaration of interests D.I.L., M.S., and D.M. are employees of Pfizer Inc. and may own stock/options in the company. M.M. and G.G declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
4. Membranous Bubbles: High-Purity and High-Titer Exosomes as the Potential Solution for Adeno-Associated Viruses to Evade Neutralization?
- Author
-
Muhuri M and Gao G
- Subjects
- Genetic Vectors, Dependovirus genetics, Exosomes
- Published
- 2021
- Full Text
- View/download PDF
5. Modulating immune responses to AAV by expanded polyclonal T-regs and capsid specific chimeric antigen receptor T-regulatory cells.
- Author
-
Arjomandnejad M, Sylvia K, Blackwood M, Nixon T, Tang Q, Muhuri M, Gruntman AM, Gao G, Flotte TR, and Keeler AM
- Abstract
Immune responses to adeno-associated virus (AAV) capsids limit the therapeutic potential of AAV gene therapy. Herein, we model clinical immune responses by generating AAV capsid-specific chimeric antigen receptor (AAV-CAR) T cells. We then modulate immune responses to AAV capsid with AAV-CAR regulatory T cells (Tregs). AAV-CAR Tregs in vitro display phenotypical Treg surface marker expression, and functional suppression of effector T cell proliferation and cytotoxicity. In mouse models, AAV-CAR Tregs mediated continued transgene expression from an immunogenic capsid, despite antibody responses, produced immunosuppressive cytokines, and decreased tissue inflammation. AAV-CAR Tregs are also able to bystander suppress immune responses to immunogenic transgenes similarly mediating continued transgene expression, producing immunosuppressive cytokines, and reducing tissue infiltration. Taken together, AAV-CAR T cells and AAV-CAR Tregs are directed and powerful immunosuppressive tools to model and modulate immune responses to AAV capsids and transgenes in the local environment., Competing Interests: M.A., A.M.K., and T.R.F. have submitted a patent (US2020029527). T.R.F. serves as a paid consultant for Ferring Ventures, which is unrelated to the work described here. A.M.K. has an SRA with Kriya Therapeutics, which is unrelated to this work. G.G. is a scientific co-founder of Voyager Therapeutics, Adrenas Therapeutics, and Aspa Therapeutics, and holds equity in the companies. G.G. is inventor on patents related to AAV-based gene therapy, some of which were licensed to commercial entities but are unrelated to this work., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
6. Vectored Immunotherapeutics for Infectious Diseases: Can rAAVs Be The Game Changers for Fighting Transmissible Pathogens?
- Author
-
Zhan W, Muhuri M, Tai PWL, and Gao G
- Subjects
- Humans, Vaccines, Communicable Diseases therapy, Dependovirus, Genetic Vectors, Immunotherapy methods
- Abstract
Conventional vaccinations and immunotherapies have encountered major roadblocks in preventing infectious diseases like HIV, influenza, and malaria. These challenges are due to the high genomic variation and immunomodulatory mechanisms inherent to these diseases. Passive transfer of broadly neutralizing antibodies may offer partial protection, but these treatments require repeated dosing. Some recombinant viral vectors, such as those based on lentiviruses and adeno-associated viruses (AAVs), can confer long-term transgene expression in the host after a single dose. Particularly, recombinant (r)AAVs have emerged as favorable vectors, given their high in vivo transduction efficiency, proven clinical efficacy, and low immunogenicity profiles. Hence, rAAVs are being explored to deliver recombinant antibodies to confer immunity against infections or to diminish the severity of disease. When used as a vaccination vector for the delivery of antigens, rAAVs enable de novo synthesis of foreign proteins with the conformation and topology that resemble those of natural pathogens. However, technical hurdles like pre-existing immunity to the rAAV capsid and production of anti-drug antibodies can reduce the efficacy of rAAV-vectored immunotherapies. This review summarizes rAAV-based prophylactic and therapeutic strategies developed against infectious diseases that are currently being tested in pre-clinical and clinical studies. Technical challenges and potential solutions will also be discussed., Competing Interests: GG is a scientific co-founder of Voyager Therapeutics and Aspa Therapeutics, and holds equity in these companies. GG is an inventor on patents with potential royalties licensed to Voyager Therapeutics, Aspa Therapeutics, and other biopharmaceutical companies. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Zhan, Muhuri, Tai and Gao.)
- Published
- 2021
- Full Text
- View/download PDF
7. Novel Combinatorial MicroRNA-Binding Sites in AAV Vectors Synergistically Diminish Antigen Presentation and Transgene Immunity for Efficient and Stable Transduction.
- Author
-
Muhuri M, Zhan W, Maeda Y, Li J, Lotun A, Chen J, Sylvia K, Dasgupta I, Arjomandnejad M, Nixon T, Keeler AM, Manokaran S, He R, Su Q, Tai PWL, and Gao G
- Subjects
- Animals, Antigen-Presenting Cells immunology, Binding Sites, Female, Genetic Therapy methods, Male, Mice, Mice, Inbred C57BL, Transgenes, Antigen Presentation immunology, Dependovirus, Genetic Vectors, MicroRNAs, Transduction, Genetic methods
- Abstract
Recombinant adeno-associated virus (rAAV) platforms hold promise for in vivo gene therapy but are undermined by the undesirable transduction of antigen presenting cells (APCs), which in turn can trigger host immunity towards rAAV-expressed transgene products. In light of recent adverse events in patients receiving high systemic AAV vector doses that were speculated to be related to host immune responses, development of strategies to mute innate and adaptive immunity is imperative. The use of miRNA binding sites (miR-BSs) to confer endogenous miRNA-mediated regulation to detarget transgene expression from APCs has shown promise for reducing transgene immunity. Studies have shown that designing miR-142BSs into rAAV1 vectors were able to repress costimulatory signals in dendritic cells (DCs), blunt the cytotoxic T cell response, and attenuate clearance of transduced muscle cells in mice to allow sustained transgene expression in myofibers with negligible anti-transgene IgG production. In this study, we screened individual and combinatorial miR-BS designs against 26 miRNAs that are abundantly expressed in APCs, but not in skeletal muscle. The highly immunogenic ovalbumin (OVA) transgene was used as a proxy for foreign antigens. In vitro screening in myoblasts, mouse DCs, and macrophages revealed that the combination of miR-142BS and miR-652-5pBS strongly mutes transgene expression in APCs but maintains high myoblast and myocyte expression. Importantly, rAAV1 vectors carrying this novel miR-142/652-5pBS cassette achieve higher transgene levels following intramuscular injections in mice than previous detargeting designs. The cassette strongly inhibits cytotoxic CTL activation and suppresses the Th17 response in vivo . Our approach, thus, advances the efficiency of miRNA-mediated detargeting to achieve synergistic reduction of transgene-specific immune responses and the development of safe and efficient delivery vehicles for gene therapy., Competing Interests: GG is a scientific co-founder of Voyager Therapeutics, Adrenas Therapeutics, and Aspa Therapeutics, and holds equity in these companies. GG is an inventor on patents with potential royalties licensed to Voyager Therapeutics, Aspa Therapeutics, and other biopharmaceutical companies. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Muhuri, Zhan, Maeda, Li, Lotun, Chen, Sylvia, Dasgupta, Arjomandnejad, Nixon, Keeler, Manokaran, He, Su, Tai and Gao.)
- Published
- 2021
- Full Text
- View/download PDF
8. Docosanoic acid conjugation to siRNA enables functional and safe delivery to skeletal and cardiac muscles.
- Author
-
Biscans A, Caiazzi J, McHugh N, Hariharan V, Muhuri M, and Khvorova A
- Subjects
- Animals, Disease Models, Animal, Fatty Acids chemistry, Heart drug effects, Heart physiopathology, Heart Diseases genetics, Heart Diseases pathology, Heart Diseases therapy, Humans, Mice, Muscle, Skeletal drug effects, Muscular Diseases genetics, Muscular Diseases pathology, Muscular Diseases therapy, Myocardium pathology, Myostatin antagonists & inhibitors, Oligonucleotides chemistry, Oligonucleotides genetics, RNA, Small Interfering chemistry, RNA, Small Interfering genetics, Fatty Acids pharmacology, Gene Transfer Techniques, Myostatin genetics, Oligonucleotides pharmacology, RNA, Small Interfering pharmacology
- Abstract
Oligonucleotide therapeutics hold promise for the treatment of muscle- and heart-related diseases. However, oligonucleotide delivery across the continuous endothelium of muscle tissue is challenging. Here, we demonstrate that docosanoic acid (DCA) conjugation of small interfering RNAs (siRNAs) enables efficient (~5% of injected dose), sustainable (>1 month), and non-toxic (no cytokine induction at 100 mg/kg) gene silencing in both skeletal and cardiac muscles after systemic injection. When designed to target myostatin (muscle growth regulation gene), siRNAs induced ~55% silencing in various muscle tissues and 80% silencing in heart, translating into a ~50% increase in muscle volume within 1 week. Our study identifies compounds for RNAi-based modulation of gene expression in skeletal and cardiac muscles, paving the way for both functional genomics studies and therapeutic gene modulation in muscle and heart., Competing Interests: Declaration of interests A.K. owns stock of RXi Pharmaceuticals and Advirna. The other authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
9. Engineering adeno-associated viral vectors to evade innate immune and inflammatory responses.
- Author
-
Chan YK, Wang SK, Chu CJ, Copland DA, Letizia AJ, Costa Verdera H, Chiang JJ, Sethi M, Wang MK, Neidermyer WJ Jr, Chan Y, Lim ET, Graveline AR, Sanchez M, Boyd RF, Vihtelic TS, Inciong RGCO, Slain JM, Alphonse PJ, Xue Y, Robinson-McCarthy LR, Tam JM, Jabbar MH, Sahu B, Adeniran JF, Muhuri M, Tai PWL, Xie J, Krause TB, Vernet A, Pezone M, Xiao R, Liu T, Wang W, Kaplan HJ, Gao G, Dick AD, Mingozzi F, McCall MA, Cepko CL, and Church GM
- Subjects
- Animals, Gene Transfer Techniques, Genetic Therapy, Immunity, Innate, Mice, Swine, Dependovirus genetics, Genetic Vectors
- Abstract
Nucleic acids are used in many therapeutic modalities, including gene therapy, but their ability to trigger host immune responses in vivo can lead to decreased safety and efficacy. In the case of adeno-associated viral (AAV) vectors, studies have shown that the genome of the vector activates Toll-like receptor 9 (TLR9), a pattern recognition receptor that senses foreign DNA. Here, we engineered AAV vectors to be intrinsically less immunogenic by incorporating short DNA oligonucleotides that antagonize TLR9 activation directly into the vector genome. The engineered vectors elicited markedly reduced innate immune and T cell responses and enhanced gene expression in clinically relevant mouse and pig models across different tissues, including liver, muscle, and retina. Subretinal administration of higher-dose AAV in pigs resulted in photoreceptor pathology with microglia and T cell infiltration. These adverse findings were avoided in the contralateral eyes of the same animals that were injected with the engineered vectors. However, intravitreal injection of higher-dose AAV in macaques, a more immunogenic route of administration, showed that the engineered vector delayed but did not prevent clinical uveitis, suggesting that other immune factors in addition to TLR9 may contribute to intraocular inflammation in this model. Our results demonstrate that linking specific immunomodulatory noncoding sequences to much longer therapeutic nucleic acids can "cloak" the vector from inducing unwanted immune responses in multiple, but not all, models. This "coupled immunomodulation" strategy may widen the therapeutic window for AAV therapies as well as other DNA-based gene transfer methods., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
10. Oncolytic Virus Alphavirus M1: A New and Promising Weapon to Fight Cancer.
- Author
-
Muhuri M and Gao G
- Subjects
- Humans, Alphavirus genetics, Neoplasms genetics, Neoplasms therapy, Oncolytic Virotherapy, Oncolytic Viruses genetics
- Published
- 2021
- Full Text
- View/download PDF
11. Overcoming innate immune barriers that impede AAV gene therapy vectors.
- Author
-
Muhuri M, Maeda Y, Ma H, Ram S, Fitzgerald KA, Tai PW, and Gao G
- Subjects
- Adaptive Immunity, Animals, Genetic Vectors therapeutic use, Humans, Dependovirus immunology, Genetic Therapy, Genetic Vectors immunology, Immunity, Innate
- Abstract
The field of gene therapy has made considerable progress over the past several years. Adeno-associated virus (AAV) vectors have emerged as promising and attractive tools for in vivo gene therapy. Despite the recent clinical successes achieved with recombinant AAVs (rAAVs) for therapeutics, host immune responses against the vector and transgene product have been observed in numerous preclinical and clinical studies. These outcomes have hampered the advancement of AAV gene therapies, preventing them from becoming fully viable and safe medicines. The human immune system is multidimensional and complex. Both the innate and adaptive arms of the immune system seem to play a concerted role in the response against rAAVs. While most efforts have been focused on the role of adaptive immunity and developing ways to overcome it, the innate immune system has also been found to have a critical function. Innate immunity not only mediates the initial response to the vector, but also primes the adaptive immune system to launch a more deleterious attack against the foreign vector. This Review highlights what is known about innate immune responses against rAAVs and discusses potential strategies to circumvent these pathways.
- Published
- 2021
- Full Text
- View/download PDF
12. Is smaller better? Vaccine targeting recombinant receptor-binding domain might hold the key for mass production of effective prophylactics to fight the COVID-19 pandemic.
- Author
-
Muhuri M and Gao G
- Subjects
- Betacoronavirus, COVID-19, Humans, SARS-CoV-2, Coronavirus Infections, Influenza Vaccines, Pandemics, Pneumonia, Viral, Severe Acute Respiratory Syndrome
- Published
- 2020
- Full Text
- View/download PDF
13. MicroRNA-138 is a Prognostic Biomarker for Triple-Negative Breast Cancer and Promotes Tumorigenesis via TUSC2 repression.
- Author
-
Nama S, Muhuri M, Di Pascale F, Quah S, Aswad L, Fullwood M, and Sampath P
- Subjects
- Animals, Disease-Free Survival, Female, Humans, MCF-7 Cells, Mice, Mice, Inbred NOD, Mice, SCID, Survival Rate, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Carcinogenesis genetics, Carcinogenesis metabolism, Gene Expression Regulation, Neoplastic, MicroRNAs genetics, MicroRNAs metabolism, RNA, Neoplasm genetics, RNA, Neoplasm metabolism, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms mortality, Tumor Suppressor Proteins biosynthesis, Tumor Suppressor Proteins genetics
- Abstract
Breast cancer manifests as a spectrum of subtypes with distinct molecular signatures, and different responses to treatment. Of these subtypes, triple-negative breast cancer (TNBC) has the worst prognoses and limited therapeutic options. Here we report aberrant expression of microRNA-138 (miR-138) in TNBC. Increased miR-138 expression is highly specific to this subtype, correlates with poor prognosis in patients, and is functionally relevant to cancer progression. Our findings establish miR-138 as a specific diagnostic and prognostic biomarker for TNBC. OncomiR-138 is pro-survival; sequence-specific miR-138 inhibition blocks proliferation, promotes apoptosis and inhibits tumour growth in-vivo. miR-138 directly targets a suite of pro-apoptotic and tumour suppressive genes, including tumour suppressor candidate 2 (TUSC2). miR-138 silences TUSC2 by binding to a unique 5'-UTR target-site, which overlaps with the translation start-site of the transcript. Over-expression of TUSC2 mimics the phenotype of miR-138 knockdown and functional rescue experiments confirm that TUSC2 is a direct downstream target of miR-138. Our report of miR-138 as an oncogenic driver in TNBC, positions it as a viable target for oligonucleotide therapeutics and we envision the potential value of using antimiR-138 as an adjuvant therapy to alleviate this therapeutically intractable cancer.
- Published
- 2019
- Full Text
- View/download PDF
14. Circumventing cellular immunity by miR142-mediated regulation sufficiently supports rAAV-delivered OVA expression without activating humoral immunity.
- Author
-
Xiao Y, Muhuri M, Li S, Qin W, Xu G, Luo L, Li J, Letizia AJ, Wang SK, Chan YK, Wang C, Fuchs SP, Wang D, Su Q, Nahid MA, Church GM, Farzan M, Yang L, Wei Y, Desrosiers RC, Mueller C, Tai PW, and Gao G
- Subjects
- Animals, Antibody Formation, Antigen-Presenting Cells, CD8-Positive T-Lymphocytes, Cytokines metabolism, Dendritic Cells immunology, Gene Transfer Techniques, Genetic Therapy methods, Genetic Vectors, Homeodomain Proteins, Lymphocyte Activation, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscles immunology, Muscles pathology, T-Lymphocytes, Cytotoxic metabolism, Dependovirus genetics, Immunity, Cellular immunology, Immunity, Humoral immunology, MicroRNAs genetics, MicroRNAs metabolism, Ovalbumin immunology
- Abstract
Recombinant adeno-associated virus (rAAV)-mediated gene delivery can efficiently target muscle tissues to serve as "biofactories" for secreted proteins in prophylactic and therapeutic scenarios. Nevertheless, efficient rAAV-mediated gene delivery is often limited by host immune responses against the transgene product. The development of strategies to prevent anti-transgene immunity is therefore crucial. The employment of endogenous microRNA (miRNA)-mediated regulation to detarget transgene expression from antigen presenting cells (APCs) has shown promise for reducing immunogenicity. However, the mechanisms underlying miRNA-mediated modulation of anti-transgene immunity by APC detargeting are not fully understood. Using the highly immunogenic ovalbumin (OVA) protein as a proxy for foreign antigens, we show that rAAV vectors containing miR142 binding sites efficiently repress co-stimulatory signals in dendritic cells, significantly blunt the cytotoxic T cell response, allow for sustained transgene expression in skeletal myoblasts, and attenuate clearance of transduced muscle cells in mice. Furthermore, the blunting of humoral immunity against circulating OVA correlates with detargeting of OVA expression from APCs. This demonstrates that incorporating APC-specific miRNA binding sites into rAAV vectors provides an effective strategy for reducing transgene-specific immune response. This approach holds promise for clinical applications where the safe and efficient delivery of a prophylactic or therapeutic protein is desired.
- Published
- 2019
- Full Text
- View/download PDF
15. C/EBPβ mediates RNA polymerase III-driven transcription of oncomiR-138 in malignant gliomas.
- Author
-
Di Pascale F, Nama S, Muhuri M, Quah S, Ismail HM, Chan XHD, Sundaram GM, Ramalingam R, Burke B, and Sampath P
- Subjects
- Animals, CCAAT-Enhancer-Binding Protein-beta metabolism, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Glioma metabolism, Glioma therapy, HEK293 Cells, Humans, Interleukin Receptor Common gamma Subunit deficiency, Interleukin Receptor Common gamma Subunit genetics, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Oncogenes genetics, Protein Binding, RNA Interference, RNAi Therapeutics methods, Xenograft Model Antitumor Assays methods, CCAAT-Enhancer-Binding Protein-beta genetics, Glioma genetics, MicroRNAs genetics, RNA Polymerase III metabolism, Transcription, Genetic
- Abstract
MicroRNA-138 (miR-138) is a pro-survival oncomiR for glioma stem cells. In malignant gliomas, dysregulated expression of microRNAs, such as miR-138, promotes Tumour initiation and progression. Here, we identify the ancillary role of the CCAAT/enhancer binding protein β (C/EBPβ) as a transcriptional activator of miR-138. We demonstrate that a short 158 bp DNA sequence encoding the precursor of miR-138-2 is essential and sufficient for transcription of miR-138. This short sequence includes the A-box and B-box elements characteristic of RNA Polymerase III (Pol III) promoters, and is also directly bound by C/EBPβ via an embedded 'C/EBPβ responsive element' (CRE). CRE and the Pol III B-box element overlap, suggesting that C/EBPβ and transcription factor 3C (TFIIIC) interact at the miR-138-2 locus. We propose that this interaction is essential for the recruitment of the RNA Pol III initiation complex and associated transcription of the oncomiR, miR-138 in malignant gliomas., (© The Author(s) 2017. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2018
- Full Text
- View/download PDF
16. EGF hijacks miR-198/FSTL1 wound-healing switch and steers a two-pronged pathway toward metastasis.
- Author
-
Sundaram GM, Ismail HM, Bashir M, Muhuri M, Vaz C, Nama S, Ow GS, Vladimirovna IA, Ramalingam R, Burke B, Tanavde V, Kuznetsov V, Lane EB, and Sampath P
- Subjects
- Animals, Blotting, Western, Carcinoma, Squamous Cell metabolism, Cell Proliferation, Female, Genes, Switch physiology, Head and Neck Neoplasms metabolism, Immunoprecipitation, Mass Spectrometry, Mice, Inbred NOD, Cell Transformation, Neoplastic metabolism, Epidermal Growth Factor physiology, Follistatin-Related Proteins physiology, MicroRNAs physiology, Wound Healing physiology
- Abstract
Epithelial carcinomas are well known to activate a prolonged wound-healing program that promotes malignant transformation. Wound closure requires the activation of keratinocyte migration via a dual-state molecular switch. This switch involves production of either the anti-migratory microRNA miR-198 or the pro-migratory follistatin-like 1 (FSTL1) protein from a single transcript; miR-198 expression in healthy skin is down-regulated in favor of FSTL1 upon wounding, which enhances keratinocyte migration and promotes re-epithelialization. Here, we reveal a defective molecular switch in head and neck squamous cell carcinoma (HNSCC). This defect shuts off miR-198 expression in favor of sustained FSTL1 translation, driving metastasis through dual parallel pathways involving DIAPH1 and FSTL1. DIAPH1, a miR-198 target, enhances directional migration through sequestration of Arpin, a competitive inhibitor of Arp2/3 complex. FSTL1 blocks Wnt7a-mediated repression of extracellular signal-regulated kinase phosphorylation, enabling production of MMP9, which degrades the extracellular matrix and facilitates metastasis. The prognostic significance of the FSTL1-DIAPH1 gene pair makes it an attractive target for therapeutic intervention., (© 2017 Sundaram et al.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.