Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Publication year range
1.
Am J Chin Med ; 52(1): 275-289, 2024.
Article in English | MEDLINE | ID: mdl-38291583

ABSTRACT

This study intends to explore the effects of Cucurbitacin B (CuB) and KIF20A on esophageal carcinoma (ESCA). Data were downloaded from the Cancer Genome Atlas (TCGA) database. The expression properties of KIF20A have been confirmed by GEPIA and ualcan from TCGA. The expression of KIF20A was determined using western blotting in ECA109 and KYSE150 cells after transfection with KIF20A, KIF20A siRNA, or numerical control siRNA (si-NC). Then, different concentrations of CuB were used to treat ECA109 and KYSE150 cells. CCK-8 and colony formation assays were used to measure cell viability, and a Transwell assay was utilized to assess cell migration and invasion ability. N-cadherin, E-cadherin, snail, p-Janus kinase 2 (JAK2), JAK2, p-signal transducer and activator of transcription 3 (STAT3), and STAT3 expression levels were evaluated using western blot. KIF20A was higher expressed in ESCA than in normal cells, and its overexpression was associated with squamous cell carcinoma, TNM stage, and lymph nodal metastasis of ESCA patients. In ECA109 and KYSE150 cells, increased KIF20A facilitated cell proliferation, migration, and invasion, whereas the knockdown of KIF20A can reverse these effects with N-cadherin. Snail expression diminished and E-cadherin increased. Similarly, CuB treatment could inhibit cell proliferation, migration, and invasion concentration dependently. Furthermore, KIF20A accelerated the expression of p-JAK2 and p-STAT3, while the application of CuB inhibited KIF20A expression and attenuated the activation of the JAK/STAT3 pathway. These findings revealed that CuB could inhibit the growth, migration, and invasion of ESCA through downregulating the KIF20A/JAK/STAT3 signaling pathway, and CuB could serve as an essential medicine for therapeutic intervention.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Triterpenes , Humans , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Cell Line, Tumor , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Signal Transduction/genetics , Carcinoma, Squamous Cell/genetics , Cell Proliferation/genetics , Cell Movement/genetics , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Cadherins/genetics , Cadherins/metabolism , Gene Expression Regulation, Neoplastic , Kinesins/genetics , Kinesins/metabolism , Kinesins/pharmacology
2.
Adv Drug Deliv Rev ; 201: 115052, 2023 10.
Article in English | MEDLINE | ID: mdl-37567502

ABSTRACT

Small interfering RNAs (siRNAs) are among the most promising therapeutic platforms in many life-threatening diseases. Owing to the significant advances in siRNA design, many challenges in the stability, specificity and delivery of siRNA have been addressed. However, safety concerns and dose-limiting toxicities still stand among the reasons for the failure of clinical trials of potent siRNA therapies, calling for a need of more comprehensive understanding of their potential mechanisms of toxicity. This review delves into the intrinsic and delivery related toxicity mechanisms of siRNA drugs and takes a holistic look at the safety failure of the clinical trials to identify the underlying causes of toxicity. In the end, the current challenges, and potential solutions for the safety assessment and high throughput screening of investigational siRNA and delivery systems as well as considerations for design strategies of safer siRNA therapeutics are outlined.


Subject(s)
High-Throughput Screening Assays , Humans , RNA, Small Interfering/therapeutic use , RNA Interference
3.
J Prim Care Community Health ; 14: 21501319231172709, 2023.
Article in English | MEDLINE | ID: mdl-37191000

ABSTRACT

INTRODUCTION: The UK Government partnered with industry to tackle cardiovascular disease (CVD) in the first NHS population health agreement. The ambition was to prevent 150 000 strokes, heart attacks and dementia cases over the next 10 years with a new siRNA LDL-C lowering therapy (Inclisiran) delivered within Integrated Care Services by primary care to support a comprehensive approach to lipid management. Following the approval of inclisiran, and guidance published by the National Institute for Health & Care Excellence (NICE) on its use, this paper has been created by a UK general practice to share real-world observations of cases and the potential service benefits of rolling out this innovative drug treatment. The process of identifying patients at risk of atherosclerotic cardiovascular disease (ASCVD) and lessons learned from implementing in practice is also addressed. Workstreams were developed to rapidly roll out a low clinical burden enhanced lipid management program incorporating siRNA LDL-C lowering therapy into primary care practice. APPROACH/METHOD: (1) Multi-disciplinary team (MDT) education program based on freely available Academic Health Science Network (AHSN), National Institute for Health & Care Excellence (NICE), and commercial materials. (2) Automated searches using a software program were run to identify "at-risk" patients alongside manual case-finding in everyday clinics. (3) Patients were invited for review using multi-channel modalities. (4) Where appropriate, treatment was commenced after consent was obtained. (5) Automated recall systems are used to ensure follow-up; initially at 3 months, then every 6 months. DISCUSSION AND CONCLUSIONS: Enhanced lipid management as a secondary prevention measure is achievable in line with national guidance and objectives. The methodology and education/training processes used in combination with reconstructing the management process can help practice staff realize the program benefits, which in turn can lead to a shift in behavior where all staff embed manual case-finding of high-risk patients into everyday consultations and reviews; enabling rapid identification of eligible patients. Taking a multi-disciplinary, holistic approach to new initiatives reduces service burden, particularly for GPs. Leveraging resources from the AHSN and others removes additional training pressures often associated with new initiatives and provides a wealth of educational material to support primary care MDT upskilling.


Subject(s)
Cardiovascular Diseases , Humans , Cardiovascular Diseases/prevention & control , Cholesterol, LDL , State Medicine , RNA, Small Interfering/therapeutic use , Primary Health Care
4.
Altern Ther Health Med ; 29(1): 130-136, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35986740

ABSTRACT

Context: Lung cancer is one of the most common forms of cancer. Autophagy and apoptosis play an important role in the development of lung cancer. Researchers have found upregulation of GRP78 expression in cancer cells of various types. Objective: The study intended to explore the mechanism of G protein-coupled receptor 78(GPR78) in regulating autophagy and drug resistance in non-small cell lung cancer (NSCLC). Design: The research team performed a laboratory study. Setting: The study took place in the Department of Thoracic Surgery at Hainan General Hospital of the Hainan Affiliated Hospital of Hainan Medical University in Haikou, Hainan, China. Intervention: The research team cultured immortalized, normal, human bronchial epithelial cells C3 (HBEC3) lines and HBEC4 lines in a serum medium without keratinocytes and infected the expression of GPR78 in knockdown A549 cells using lentiviral agents. The team divided the cells into a control group and a shRNA-GPR78 group, the intervention group. The lentiviral silencing vector expressing shRNA targets human GPR78#1 and GPR78 #2aadam10. Outcome Measures: The research team analyzed the mRNA expression of GPR78 in the NSCLC cell lines H1975, H1299, and A549 and in HBEC3 and HBEC4 using a real time-polymerase chain reaction (RT-PCR) and measured the proliferation of A549 cells at 0h, 24h, 48h, 72h, and 96h using yellow tetrazolium salt (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). The team also analyzed the migration and invasion ability of cells using wound healing and Transwell tests as well as measured the protein expression of the autophagy-related factors Beclin-1, microtubule-associated protein light chain 3-I/II (LC3-I/LC3-II), ubiquitin-binding protein p62 and c-Jun N-terminal kinase (JNK) using a Western blot test. The team also analyzed the protein expressions of caspase-9, caspase-3, and caspase-12 related to apoptosis using a Western blot. To detect the cell viability induced by cisplatin, the team used a Cell Counting Kit 8 (CCK-8) at the concentrations of 1µM, 3µM and 10µM. Results: The mRNA expression of GPR78 in the H1975, H1299, and A549 cell lines was significantly higher than that in the HBEC3 and HBEC4 cell lines (P < .05). At 48h, 72h, and 96h, the A549 cell proliferation in the shRNA-GPR78 group was significantly lower than that of the control group (P < .05). The cell migration and invasion of cells in the shRNA-GPR78 group was significantly lower than that in the control group (P < .05), and the cell viability of the shRNA-GPR78 group was significantly lower than that of control group (P < .05). The expression of Beclin-1 and JNK protein in shRNA-GPR78 group was significantly higher than that in the control group (P < .05), and the expression of LC3-I/LC3-II and p62 protein in shRNA-GPR78 group was significantly lower than that in the control group (P < .05). The protein expressions of caspase-9, caspase-3, and caspase-12 in the shRNA-GPR78 group were significantly higher than those of the control group (P < .05), and the protein activities of RhoA and Rac1 in the shRNA-GPR78 group were significantly lower than those in the control group (P < .05). Conclusion: NSCLC upregulated GPR78. The knockdown of GPR78 can attenuate the proliferation, migration, and invasion of NSCLC cells and increase the apoptosis and autophagy of NSCLC cells that cisplatin has induced. Therefore, targeting GPR78 may be a promising treatment strategy for NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Cisplatin/pharmacology , Cisplatin/therapeutic use , Caspase 3/therapeutic use , Caspase 9 , Beclin-1 , Caspase 12/therapeutic use , Cell Line, Tumor , Apoptosis , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Cell Proliferation , Autophagy , Drug Resistance , RNA, Messenger
5.
Arthritis Res Ther ; 24(1): 234, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36253807

ABSTRACT

BACKGROUND: Abnormal proliferation of fibroblast-like synoviocytes (FLSs) in the synovial lining layer is the primary cause of synovial hyperplasia and joint destruction in rheumatoid arthritis (RA). Currently, the relationship between metabolic abnormalities and FLS proliferation is a new focus of investigation. However, little is known regarding the relationship between amino acid metabolism and RA. METHODS: The concentrations of amino acids and cytokines in the synovial fluid of RA (n = 9) and osteoarthritis (OA, n = 9) were detected by LC-MS/MS and CBA assay, respectively. The mRNA and protein expression of cationic amino acid transporter-1 (CAT-1) were determined in FLSs isolated from RA and OA patients by real-time PCR and western blotting. MTT assay, cell cycle, apoptosis, invasion, and cytokine secretion were determined in FLSs knocked down of CAT-1 using siRNA or treated with D-arginine under normoxic and hypoxic culture conditions. A mouse collagen-induced arthritis (CIA) model was applied to test the therapeutic potential of blocking the uptake of L-arginine in vivo. RESULTS: L-rginine was upregulated in the synovial fluid of RA patients and was positively correlated with the elevation of the cytokines IL-1ß, IL-6, and IL-8. Further examination demonstrated that CAT-1 was the primary transporter for L-arginine and was overexpressed on RA FLSs compared to OA FLSs. Moreover, knockdown of CAT-1 using siRNA or inhibition of L-arginine uptake using D-arginine significantly suppressed L-arginine metabolism, cell proliferation, migration, and cytokine secretion in RA FLSs under normoxic and hypoxic culture conditions in vitro but increased cell apoptosis in a dose-dependent manner. Meanwhile, in vivo assays revealed that an L-arginine-free diet or blocking the uptake of L-arginine using D-arginine suppressed arthritis progression in CIA mice. CONCLUSION: CAT-1 is upregulated and promotes FLS proliferation by taking up L-arginine, thereby promoting RA progression.


Subject(s)
Arginine , Arthritis, Experimental , Arthritis, Rheumatoid , Cationic Amino Acid Transporter 1 , Synoviocytes , Animals , Mice , Amino Acids/metabolism , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/drug therapy , Cationic Amino Acid Transporter 1/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Chromatography, Liquid , Cytokines/metabolism , Fibroblasts/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Mice, Inbred CBA , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Synovial Membrane/metabolism , Synoviocytes/metabolism , Tandem Mass Spectrometry
6.
Molecules ; 27(19)2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36234773

ABSTRACT

Akebia saponin D (ASD) is derived from the Dipsacus asper Wall. ex Henry, which is a traditional Chinese medicine commonly used to treat rheumatic arthritis (RA). However, the in-depth mechanism of the anti-inflammatory effect of ASD is still unclear. This study aimed to preliminarily explore the anti-inflammatory effect of ASD and the underlying mechanisms from the perspective of DNA methylation and inflammation-related pathways. We found that ASD significantly reduced the production of multiple inflammatory mediators, including nitric oxide (NO) and prostaglandin E2 (PGE2), in LPS-induced RAW264.7 cells. The expression of DNA methyltransferase (DNMT) 3b and inducible nitric oxide synthase (iNOS) was also obviously inhibited by the ASD treatment. The protein and mRNA levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were also significantly inhibited by ASD. ASD inhibited the macrophage M1 phenotype, inhibited the high level of DNMT3b, and downregulated the signal transducer and activator of the transcription 3 (STAT3) pathway to exert its anti-inflammatory activity. Furthermore, DNMT3b siRNA and Nrf2 siRNA significantly promoted the anti-inflammatory effect of ASD. Our study demonstrates for the first time that ASD inhibits the IL-6-STAT3-DNMT3b axis and activates the nuclear factor-E2-related factor 2 (Nrf2) signaling pathway to achieve its inhibitory effect on inflammatory reactions.


Subject(s)
Interleukin-6 , NF-E2-Related Factor 2 , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , DNA/therapeutic use , Dinoprostone/metabolism , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Methyltransferases/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger , RNA, Small Interfering/therapeutic use , STAT3 Transcription Factor , Saponins , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Int J Mol Sci ; 23(20)2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36293521

ABSTRACT

Angiogenesis inhibitor drugs have been explored as important pharmacological agents for cancer therapy, including hepatocellular carcinoma. These agents have several drawbacks, such as drug resistance, nonspecific toxicity, and systemic side effects. Therefore, combination therapy of the drug and small interfering RNA could be a promising option to achieve high therapeutic efficacy while allowing a lower systemic dose. Therefore, we studied adding an alpha-fetoprotein siRNA (AFP-siRNA) incorporated on polymeric nanoparticles (NPs) along with angiogenesis inhibitor drugs. The AFP siRNA-loaded NPs were successfully synthesized at an average size of 242.00 ± 2.54 nm. Combination treatment of AFP-siRNA NPs and a low dose of sunitinib produced a synergistic effect in decreasing cell viability in an in vitro hepatocellular carcinoma (HCC) model. AFP-siRNA NPs together with sorafenib or sunitinib greatly inhibited cell proliferation, showing only 39.29 ± 2.72 and 44.04 ± 3.05% cell viability, respectively. Moreover, quantitative reverse transcription PCR (qRT-PCR) demonstrated that AFP-siRNA incorporated with NPs could significantly silence AFP-mRNA expression compared to unloaded NPs. Interestingly, the expression level of AFP-mRNA was further decreased to 28.53 ± 5.10% when sunitinib was added. Therefore, this finding was considered a new promising candidate for HCC treatment in reducing cell proliferation and enhancing therapeutic outcomes.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , RNA, Small Interfering/therapeutic use , alpha-Fetoproteins/genetics , Sorafenib/pharmacology , Sorafenib/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Sunitinib/therapeutic use , Cell Line, Tumor , Polymers/therapeutic use , RNA, Messenger
8.
J Nanobiotechnology ; 20(1): 96, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35236356

ABSTRACT

BACKGROUND: Despite extensive investigations on photothermal therapy, the clinical application is restricted due to poor stability, low therapeutic efficacy of photothermal therapy agents and its affinity loss in the multistep synthesis of delivery carriers. To address this, we designed an IR792-MCN@ZIF-8-PD-L1 siRNA (IM@ZP) nanoparticle drug delivery system. IM@ZP was prepared by in situ synthesis and physical adsorption, followed by characterization. Photothermal conversion ability of IM@ZP was assessed by irradiation of near-infrared (NIR) laser, followed by analysis of its effect on 4T1 cell viability, maturation of dendritic cells (DCs) and the secretion of related cytokines in vitro, and the changes of tumor infiltrating T cells and natural killer (NK) cells in vivo. Subcutaneous 4T1 tumor-bearing mouse and lung metastasis models were established to investigate the role of IM@ZP in killing tumor and inhibiting metastasis in vivo. RESULTS: IM@ZP was uniform nanoparticles of 81.67 nm with the characteristic UV absorption peak of IR792, and could effectively adsorb PD-L1 siRNA. Under the irradiation of 808 nm laser, IM@ZP exhibited excellent photothermal performance. IM@ZP could be efficiently uptaken by 4T1 cells, and had high transfection efficiency of PD-L1 siRNA. Upon NIR laser irradiation, IM@ZP effectively killed 4T1 cells, upregulated HSP70 expression, induced DC maturation and increased secretion of TNF-α and IL-6 in vitro. Moreover, in vivo experimental results revealed that IM@ZP enhanced photothermal immunotherapy as shown by promoted tumor infiltrating CD8 + and CD4 + T cells and NK cells, and inhibited tumor growth and lung metastasis. CONCLUSION: Together, biocompatible IM@ZP nanoparticles result in high photothermal immunotherapy efficiency and may have a great potential as a delivery system for sustained cancer therapy.


Subject(s)
Nanoparticles , Triple Negative Breast Neoplasms , Animals , B7-H1 Antigen , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Delivery Systems , Humans , Immunotherapy , Lasers , Mice , Phototherapy/methods , RNA, Small Interfering/therapeutic use , Triple Negative Breast Neoplasms/drug therapy
9.
Biomaterials ; 282: 121404, 2022 03.
Article in English | MEDLINE | ID: mdl-35134700

ABSTRACT

Diagnosis-guided synergistic treatment based on innovative nanomaterials is of great significance for the development of anti-cancer therapies. However, the low delivery efficiency of therapeutic gene and the inability to trigger release on demand are still major obstacles impeding its wide application. Herein, we report an ultra-fast one-step method within 2 min to prepare a smart carrier, liposome-coated Prussian blue @ gold nano-flower, which is named LPAR after linking with tumor-targeting peptide. The versatile LPAR not only can respond to near-infrared (NIR) light, achieve the selective delivery and the controlled release of siRNA targeting the mutant gene of Kras at its codon-12 from Glycine (G) to Aspartic acid (D) (named as G12D mutant gene) in the malignant pancreatic tumors, but also efficiently convert the absorbed NIR light into the heat to realize gene-photothermal synergistic therapy both in vitro and in vivo. Theoretical simulation results reveal that the outstanding photothermal conversion efficiency of LPAR is mainly due to its higher electric field intensity and power density distributions. Furthermore, the LPAR possesses the capabilities for triple-modal imaging. Therefore, the developed NIR light-responsive LPAR has the potential to be served as a tumor-targeted nano-delivery system for imaging-guided synergistic therapy of cancers.


Subject(s)
Nanoparticles , Neoplasms , Cell Line, Tumor , Doxorubicin/therapeutic use , Gold/therapeutic use , Humans , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Phototherapy , RNA, Small Interfering/therapeutic use
10.
J Control Release ; 343: 314-325, 2022 03.
Article in English | MEDLINE | ID: mdl-35085700

ABSTRACT

Rheumatoid arthritis (RA) is a common inflammatory disease and its treatment is largely limited by drug ineffectiveness or severe side effects. In RA progression, multiple signalling pathways, such as hypoxia-inducible factor (HIF)-1α, nuclear factor kappa B (NF-κB), and mitogen-activated protein kinase (MAPK) pathways, act synergistically to maintain the inflammatory response. To downregulate HIF-1α, NF-κB, and MAPK expression, we proposed HIF-1α siRNA-loaded calcium phosphate nanoparticles encapsulated in apolipoprotein E3-reconstituted high-density lipoprotein (HIF-CaP-rHDL) for RA therapy. Here, we evaluated the potential of CaP-rHDL nanoparticles in RA therapy using a murine macrophage line (RAW 264.7) and a collagen-induced arthritis (CIA) mouse model. The CaP-rHDL nanoparticles showed significant anti-inflammatory effects along with HIF-1α knockdown and NF-κB and MAPK signalling pathway inhibition in lipopolysaccharide-activated macrophages. Moreover, they inhibited receptor activator of NF-κB ligand (RANKL)-induced osteoclast formation. In CIA mice, their intravenous administration resulted in high accumulation at the arthritic joint sites, and HIF-CaP-rHDL effectively suppressed inflammatory cytokine secretion and relieved bone erosion, cartilage damage, and osteoclastogenesis. Thus, HIF-CaP-rHDL demonstrated great potential in RA precision therapy by inhibiting multiple inflammatory signalling pathways.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Nanoparticles , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , NF-kappa B , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use
11.
Methods Mol Biol ; 2323: 267-280, 2021.
Article in English | MEDLINE | ID: mdl-34086287

ABSTRACT

Spontaneous tumor regression following bacterial infection has been observed for hundreds of years. These observations along with anecdotal medical findings in 1890s led to the development of Coley's "toxins," consisting of killed Streptococcus pyogenes and Serratia marcescens bacteria, as the first cancer immunotherapy. The use of this approach, however, was not widely accepted at the time especially after the introduction of radiation therapy as a treatment for cancer in the early 1900s. Over the last 30-40 years there has been renewed interest in the use of bacteria to treat human solid tumors. This is based on the observation that various nonpathogenic anaerobic bacteria can infiltrate and replicate within solid tumors when given intravenously. Bacteria tested as potential anticancer agents include the Gram-positive obligate anaerobes Bifidobacterium and Clostridium, as well as the gram-negative facultative anaerobe Salmonella. Recent advances in synthetic biology and clinical success in cancer immunotherapy provide renewed momentum for developing bacteria-based cancer immunotherapy for cancer treatment and should allow greater potential for the development of novel therapeutic approaches for this devastating disease.


Subject(s)
Biological Therapy/methods , Neoplasms/therapy , RNA Interference , Synthetic Biology/methods , Animals , Cell Line, Tumor , Clinical Trials, Phase I as Topic , Colonic Neoplasms/microbiology , Colonic Neoplasms/therapy , Escherichia coli/genetics , Escherichia coli/physiology , Female , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Humans , Immunotherapy/methods , Immunotherapy/trends , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasms/microbiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Real-Time Polymerase Chain Reaction/methods , Remission Induction , Salmonella typhimurium/genetics , Salmonella typhimurium/physiology , Species Specificity , Specific Pathogen-Free Organisms , Synthetic Biology/trends , Xenograft Model Antitumor Assays
12.
Mol Pharm ; 18(6): 2105-2121, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33945284

ABSTRACT

The ongoing pandemic of global concern has killed about three million humans and affected around 151 million people worldwide, as of April 30, 2021. Although recently approved vaccines for COVID-19 are engendering hope, finding new ways to cure the viral pandemic is still a quest for researchers worldwide. Major pandemics in history have been of viral origin, such as SARS, MERS, H1NI, Spanish flu, and so on. A larger emphasis has been on discovering potential vaccines, novel antiviral drugs, and agents that can mitigate the viral infection symptoms; however, a relatively new area, RNA interference (RNAi), has proven effective as an antiviral agent. The RNAi phenomenon has been largely exploited to cure cancer, neurodegenerative diseases, and some rare diseases. The U.S. Food and Drug Administration has recently approved three siRNA products for human use that garner significant hope in siRNA therapeutics for coronaviruses. There have been some commentaries and communications addressing this area. We have summarized and illustrated the significance and the potential of the siRNA therapeutics available as of April 30, 2021 to combat the ongoing viral pandemic and the emerging new variants such as B.1.1.7 and B.1.351. Numerous successful in vitro studies and several investigations to address the clinical application of siRNA therapeutics provide great hope in this field. This seminal Review describes the significance of siRNA-based therapy to treat diverse viral infections in addition to the current coronavirus challenge. In addition, we have thoroughly reviewed the patents approved for coronaviruses, the major challenges in siRNA therapy, and the potential approaches to address them, followed by innovation and prospects.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Pandemics/prevention & control , RNA, Small Interfering/therapeutic use , SARS-CoV-2/genetics , Antiviral Agents/history , COVID-19/epidemiology , COVID-19/history , COVID-19/virology , Clinical Trials as Topic , Drug Approval , Drug Evaluation, Preclinical , History, 20th Century , History, 21st Century , Humans , Mutation , Patents as Topic , RNA, Small Interfering/history , SARS-CoV-2/pathogenicity
13.
Aging (Albany NY) ; 13(2): 1872-1882, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33440348

ABSTRACT

OBJECTIVE: This study aimed to investigate the effects of multiwalled carbon nanotubes (MWNTs) co-delivering sorafenib (Sor) and epidermal growth factor receptor (EGFR) siRNA (MWNT/Sor/siRNA) on tumor growth in liver cancer (LC). RESULTS: MWNT/Sor/siRNA was proved to possess increased Sor release, high siRNA stability, and enhanced cellular uptake. In addition, MWNT treatment has few effects on cell proliferation and apoptosis in HepG2 cells; however, MWNT/Sor/siRNA treatment significantly inhibited clone number and induced cell apoptosis, which shows a more favorable antitumor effect than MWNT/Sor and free Sor and free siRNA in HepG2 cells. Moreover MWNT/Sor/siRNA treatment has the most significant antitumor effect in vivo. CONCLUSIONS: MWNT/Sor/siRNA exhibited a superior antitumor effect in vitro and in vivo. METHODS: The MWNT/Sor and MWNT/Sor/siRNA were prepared, and then the morphologies of MWNT/Sor/siRNA were analyzed. In vitro Sor release assay, siRNA stability and cellular uptake of MWNT/Sor/siRNA were performed as well. Next, the effects of MWNT, free Sor, free siRNA, MWNT/Sor and MWNT/Sor/siRNA were evaluated by colony-forming assay, and cell apoptosis assay in HepG2 cells. Meanwhile, the level of EGFR and proteins associated with apoptosis was tested. Furthermore, the anti-tumor effects of MWNT/Sor/siRNA on LC xenograft mice were also unraveled.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Delivery Systems , ErbB Receptors/genetics , Liver Neoplasms/drug therapy , Nanotubes, Carbon , RNA, Small Interfering/therapeutic use , Sorafenib/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Liver Neoplasms/genetics , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Small Interfering/administration & dosage , Sorafenib/administration & dosage
14.
Dtsch Med Wochenschr ; 146(2): 92-101, 2021 01.
Article in German | MEDLINE | ID: mdl-33465805

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of morbidity and mortality. The fact that elevated levels of low-density lipoprotein-cholesterol (LDL-C) play a causal role in the development of ASCVD is now well accepted, given the results of numerous epidemiological and genetic studies, as well as randomized controlled clinical trials. Statins have become a primary therapeutic cornerstone in ASCVD prevention since they have been shown to reduce CV events by reducing levels of LDL-C. But despite the proven efficacy and safety of statin therapy, several aspects indicate a substantial need for additional or alternative LDL-C lowering therapies. These aspects include not only a high variability in individual response to therapy, but also possible side effects, potentially reducing adherence to treatment. Most importantly, an elevated risk for cardiovascular (CV) events remains in a large proportion of high-risk patients, especially in those with persistent elevation of LDL-C levels despite a maximum tolerated dose of statin therapy. Also, large clinical trials currently investigate a potential CV benefit of drug therapies targeting elevated levels of triglycerides and lipoprotein (a), respectively.


Subject(s)
Atherosclerosis/drug therapy , Hypolipidemic Agents , Cholesterol, LDL/blood , Dicarboxylic Acids/therapeutic use , Fatty Acids/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Humans , Oligonucleotides, Antisense/therapeutic use , RNA, Small Interfering/therapeutic use
15.
Am J Reprod Immunol ; 85(2): e13349, 2021 02.
Article in English | MEDLINE | ID: mdl-32966664

ABSTRACT

Exosomes are unique biogenic nanocarriers of endocytic origin that are generated from most of the cells and found in biofluids like milk, plasma, saliva, and urine. Bovine milk represents the largest and an economic source for the production of exosomes. In recent past, the utility of the milk exosomes as drug carriers is intensified. Exosomes are emerging for delivery of both small and large therapeutics due to their biocompatibility. In this article, we highlighted the various exosomal isolation techniques, physicochemical properties, their biodistribution, and utility of milk exosomes in delivering the small drug molecules and siRNA to combat cancer.


Subject(s)
Biological Therapy/methods , Drug Delivery Systems/methods , Exosomes/metabolism , Milk/metabolism , Neoplasms/therapy , Animals , Antineoplastic Agents/therapeutic use , Humans , Nanostructures , RNA, Small Interfering/therapeutic use
16.
Reproduction ; 161(1): 11-19, 2021 01.
Article in English | MEDLINE | ID: mdl-33112285

ABSTRACT

Endometriosis is a common gynecological disease in reproductive-age women. Although the hormone-dependent therapy is the first line treatment for endometriosis, it is not a curative regimen and associated with severe side-effects, which significantly decrease the life quality of affected individuals. To seek a target for treatment of endometriosis, we focused on plasma membrane proteins that are elevated in ectopic cells and exert beneficial effects in cell growth and survival. We performed bioinformatics analysis and identified the neurotrophic receptor tyrosine kinase 2 (NTRK2) as a potential candidate for treatment. The expression levels of NTRK2 were markedly upregulated in the lesions of clinical specimen as well as in the mouse endometriotic-like lesion. Mechanistic investigation demonstrated that upregulation of NTRK2 is induced by hypoxia in a hypoxia-inducible factor 1 alpha-dependent manner. Knockdown of NTRK2 or administration of ANA-12, a selective antagonist of NTRK2, significantly induced endometriotic stromal cells death, suggesting it may be a potential therapeutic agent. In vivo study using surgery-induced endometriosis mice model showed ANA-12 (1.5 mg/kg body weight) treatment induced apoptosis of endometriotic cells and caused the regression of ectopic lesions. Taken together, our findings suggest a possible mechanism responsible for the aberrant expression of NTRK2 in endometriotic lesions and this may be involved in the pathogenesis of endometriosis.


Subject(s)
Endometriosis/drug therapy , Membrane Glycoproteins/metabolism , RNA, Small Interfering/therapeutic use , Receptor, trkB/metabolism , Animals , Choristoma/metabolism , Drug Evaluation, Preclinical , Endometriosis/metabolism , Female , Humans , Membrane Glycoproteins/antagonists & inhibitors , Mice, Inbred C57BL , Molecular Targeted Therapy , Primary Cell Culture , RNA, Small Interfering/pharmacology , Receptor, trkB/antagonists & inhibitors , Stromal Cells/metabolism
17.
ACS Appl Mater Interfaces ; 12(20): 22613-22623, 2020 May 20.
Article in English | MEDLINE | ID: mdl-32338491

ABSTRACT

Small interfering RNA (siRNA)-induced gene therapy has been recognized as a promising avenue for effective cancer treatment, while easy enzymatic degradation, poor transfection efficiency, nonspecific biodistribution, and uncontrolled release hinder its extensive clinical applications. Zeolitic imidazolate frameworks-8 (ZIF-8) have emerged as promising drug carriers without an in-depth exploration in programmable siRNA delivery. Herein, we report a multifunctional PDAs-ZIF-8 (PZ) nanoplatform for delivering siRNA with combined photothermal therapy (PTT) and gene therapy (GT) via the noninvasive guidance of photoacoustic (PA)/near-infrared (IR) dual-modal imaging. The ingenious PZ nanocarriers mediated the tumor-specific accumulation of therapeutic siRNA without undesired degradation and preleakage. The pH-responsive ZIF-8 decomposed in an acidic tumor microenvironment that was accompanied by the release of siRNA payloads for cleaving target mRNA in gene silencing therapy. Meanwhile, the polydopamine nanoparticles (PDAs) could simultaneously serve as a powerful noninvasive PA/IR imaging contrast agent and versatile photothermal agent for diagnosis-guided photogenetherapy. The systematic in vitro and in vivo experimental explorations demonstrated that our PDAs-siRNA-ZIF-8 (PSZ) could greatly enhance the therapeutic efficiency as compared with the corresponding PTT or GT monotherapy. This work holds great potential to advance the development of more intelligent diagnosis and therapeutic strategies, thus supplying promising smart nanomedicines in the near future.


Subject(s)
Antineoplastic Agents/therapeutic use , Contrast Media/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , RNA, Small Interfering/therapeutic use , Animals , Combined Modality Therapy/methods , Contrast Media/radiation effects , Drug Carriers/radiation effects , Gene Silencing/drug effects , Genetic Therapy , Hyperthermia, Induced/methods , Indoles/chemistry , Indoles/radiation effects , Infrared Rays , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/radiation effects , Mice, Inbred BALB C , Nanoparticles/radiation effects , Phototherapy/methods , Polymers/chemistry , Polymers/radiation effects , Xenograft Model Antitumor Assays
18.
Cell Biochem Funct ; 38(7): 955-964, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32157707

ABSTRACT

Long non-coding RNA (lncRNA) LINC00899 is one kind cytoplasmic lncRNA, however, there is rarely little information about its function in physiological process. Here, we demonstrated that lncRNA LINC00899 was upregulated in acute myeloid leukaemia (AML) cells and was quite correlated with poor prognosis of AML patients. High expression of LINC00899 in AML cells could promote cell proliferation and inhibit cell apoptosis, and facilitate the progression of AML consequently both in vitro and in vivo. Besides, LINC00899 acted as a molecular sponge of miR-744-3p. Furthermore, we characterized YY1 as the direct target of miR-744-3p, and LINC00899/miR-744-3p interaction modulated YY1 expression in AML cells. Finally, we verified LINC00899 modulated AML cell proliferation and apoptosis via regulating YY1. Our study revealed novel mechanism about how did lncRNA LINC00899 execute function in AML and thus provided potential therapeutic interventions for AML. SIGNIFICANCE OF THE STUDY: LncRNA LINC00899 is upregulated in AML cells and is correlated with poor prognosis of AML patients. LncRNA LINC00899 mediates cell proliferation and apoptosis of acute myeloid leukaemia cells. Knockdown of LINC00899 inhibited the growth of xenograft glioma tumour in vivo. LINC00899 acts as a molecular sponge of miR-744-3p. YY1 is the downstream target of LINC00899/miR-744-3p signalling.


Subject(s)
Leukemia, Myeloid, Acute/pathology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Progression , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Mice , Mice, Nude , Prognosis , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Up-Regulation , YY1 Transcription Factor/chemistry , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
19.
Mol Diagn Ther ; 24(1): 49-59, 2020 02.
Article in English | MEDLINE | ID: mdl-31701435

ABSTRACT

In 2018, patisiran was the first-ever RNA interference (RNAi)-based drug approved by the US Food and Drug Administration. Now pharmacology textbooks may include a new drug class that results in the effect first described by Fire and Mello 2 decades ago: post-transcriptional gene silencing by a small-interfering RNA (siRNA). Patients with hereditary transthyretin-mediated amyloidosis (hATTR amyloidosis) present with mutations in the transthyretin (TTR) gene that lead to the formation of amyloid deposits in peripheral nerves and heart. The disease may also affect the eye and central nervous system. The formulation of patisiran comprises the RNAi drug encapsulated into a nanoparticle especially developed to deliver the anti-TTR siRNA into the main TTR producer: the liver. Hepatic cells contain apolipoprotein E receptors that recognize ApoE proteins opsonized in the lipid carrier and internalize the drug by endocytosis. Lipid vesicles are disrupted in the cell cytoplasm, and siRNAs are free to trigger the RNAi-based TTR gene silencing. The silencing process involves the binding of siRNA guide strand to 3'-untranslated region sequence of both mutant and wild-type TTR messenger RNA, which culminates in the TTR mRNA cleavage by the RNA-induced silencing complex (RISC) as the first biochemical drug effect. Patisiran 0.3 mg/kg is administered intravenously every 3 weeks. Patients require premedication with anti-inflammatory drugs and antagonists of histamine H1 and H2 receptors to prevent infusion-related reactions and may require vitamin A supplementation. Following patisiran treatment, TTR knockdown remained stable for at least 2 years. Adverse effects were mild to moderate with unchanged hematological, renal, or hepatic parameters. No drug-related severe adverse effects occurred in a 24-month follow-up phase II open-label extension study. At the recommended dosage of patisiran, Cmax and AUC values (mean ± standard deviation) were 7.15 ± 2.14 µg/mL and 184 ± 159 µg·h/mL, respectively. The drug showed stability in circulation with > 95% encapsulated in lipid particles. Metabolization occurred by ribonuclease enzymes, with less than 1% excreted unchanged in the urine. Patisiran ameliorated neuropathy impairment according to the modified Neuropathy Impairment Score + 7 analysis of the phase III study. The Norfolk Quality of Life-Diabetic Neuropathy score and gait speed improved in 51% of the patisiran-treated group in 18 months. Additionally, the modified body mass index showed positive outcomes. Altogether, the data across phase I-III clinical trials points to patisiran as an effective and safe drug for the treatment of hATTR amyloidosis. It is hoped that real-world data from a larger number of patients treated with patisiran will confirm the effectiveness of this first-approved siRNA-based drug.


Subject(s)
Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/therapy , Genetic Therapy , RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Drug Administration Routes , Drug Evaluation, Preclinical , Gene Silencing , Humans , Oligonucleotides/administration & dosage , Prealbumin/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/adverse effects , RNA, Small Interfering/pharmacokinetics , Treatment Outcome
20.
J Microencapsul ; 36(7): 635-648, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31509450

ABSTRACT

In this study, the anticancer activities of two siRNA carriers were compared using a human lung adenocarcinoma epithelial cell line (A549). Firstly, poly(styrene)-graft-poly(linoleic acid) (PS-g-PLina) and poly(styrene)-graft-poly(linoleic acid)-graft-poly(ethylene glycol) (PS-g-PLina-g-PEG) graft copolymers were synthesized by free-radical polymerization. PS-PLina and PS-PLina-PEG nanoparticles (NPs) were prepared by solvent evaporation method and were then characterized. The size was found as 150 ± 10 nm for PS-PLina and 184 ± 6 nm for PS-PLina-PEG NPs. The NPs were functionalized with poly(l-lysine) (PLL) for c-myc siRNA conjugation. siRNA entrapment efficiencies were found in the range of 4-63% for PS-PLina-PLL and 6-42% for PS-PLina-PEG-PLL NPs. The short-term stability test was realised for 1 month. siRNA release profiles were also investigated. In vitro anticancer activity of siRNA-NPs was determined by MTT, flow cytometry, and fluorescence microscopy analyses. Obtained findings showed that both NPs systems were promising as siRNA delivery tool for lung cancer therapy.


Subject(s)
Adenocarcinoma of Lung/therapy , Lung Neoplasms/therapy , Nanoconjugates/chemistry , Plant Oils/chemistry , RNA, Small Interfering/therapeutic use , A549 Cells , Adenocarcinoma of Lung/genetics , Drug Carriers/chemistry , Humans , Linoleic Acid/chemistry , Lung Neoplasms/genetics , Polyethylene Glycols/chemistry , Polystyrenes/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNAi Therapeutics
SELECTION OF CITATIONS
SEARCH DETAIL