Blog

  • CRISPR gene editing efficiency triples with DNA-wrapped nanoparticles

    CRISPR gene editing efficiency triples with DNA-wrapped nanoparticles

    image: ©Nathan Devery| iStock

    Northwestern scientists developed DNA-wrapped nanoparticles that triple CRISPR efficiency, paving the way for more effective gene editing therapies

    Northwestern University researchers have unveiled a major breakthrough in CRISPR technology, demonstrating that DNA-wrapped nanoparticles can deliver gene editing tools with three times the efficiency of current methods. The discovery marks a step forward in making gene editing therapies safer, faster, and more accessible.

    The study will be published in the Proceedings of the National Academy of Sciences on September 5th.

    Lipid nanoparticle spherical nucleic acids: An exciting discovery

    Lipid nanoparticle spherical nucleic acids (LNP-SNAs) are tiny structures that carry the complete set of CRISPR editing tools – Cas9 enzymes, guide RNA, and a DNA repair template – wrapped in a dense, protective shell of DNA. The DNA coating not only guides LNP-SNAs to specific organs and tissues but also facilitates their entry into cells.

    The scientists conducted lab tests on various human and animal cell types. They found that the new CRISPR delivery system, involving DNA-wrapped nanoparticles, entered cells up to three times more effectively than the standard lipid particle delivery systems used for COVID-19 vaccines. This impressive efficiency, coupled with significantly reduced toxicity, and a threefold boost in gene editing efficiency, is a cause for excitement and anticipation in the scientific community. 

    “CRISPR is a potent tool that could correct defects in genes to decrease susceptibility to disease and even eliminate disease itself,” said nanotechnology and nanomedicine pioneer Chad A. Mirkin, who led the new study. “But it’s difficult to get CRISPR into the cells and tissues that matter. Reaching and entering the right cells — and the right places within those cells — requires a minor miracle. By using SNAs to deliver the machinery required for gene editing, we aimed to maximize CRISPR’s efficiency and expand the number of cell and tissue types that we can deliver it to.”

    The study highlights the importance of how a nanomaterial’s structure can determine its potency. It underlies the emerging field of structural nanomedicine, pioneered by Northwestern’s Chad A. Mirkin and his colleagues.

    CRISPR’s biggest hurdle: Inefficient delivery into cells and nuclei

    While CRISPR can disable genes, repair mutations, and add new functions once inside a cell, it cannot enter on its own and instead relies on delivery vehicles such as viral vectors or lipid nanoparticles (LNPs). 

    While viruses are efficient, they can trigger an immune response in the human body, leading to painful or even dangerous side effects. In contrast, lipid nanoparticles are safer but less efficient, often becoming trapped in endosomes or cellular compartments. The new CRISPR delivery system, involving DNA-wrapped nanoparticles, offers a safer alternative, providing reassurance and confidence in its potential for gene editing therapies.

    “Only a fraction of the CRISPR machinery actually makes it into the cell, and even a smaller fraction makes it all the way into the nucleus,” Mirkin said. “Another strategy is to remove cells from the body, inject the CRISPR components, and then put the cells back in. As you can imagine, that’s extremely inefficient and impractical.”

    DNA-wrapped support for CRISPR

    To overcome CRISPR delivery problems, Mirkin’s team turned to SNAs, which are globular, instead of linear, forms of DNA and RNA previously invented in Mirkin’s lab at Northwestern.

    The spherical genetic material surrounds a nanoparticle core, which can be packed with cargo. Roughly 50 nanometers in diameter, the tiny structures possess a proven ability to enter cells for targeted delivery. Seven SNA-based therapies are already in human clinical trials, including a Phase 2 clinical trial for Merkel cell carcinoma being developed by Flashpoint Therapeutics, a clinical-stage biotechnology startup.

    In the new study, Mirkin’s team began with an LNP core that carried the CRISPR machinery inside. The team then decorated the particle’s surface with a dense layer of short strands of DNA, as DNA can interact with a cell’s surface receptors, and cells readily absorb SNAs. The DNA can also be engineered to make delivery more selective.

    “Simple changes to the particle’s structure can dramatically change how well a cell takes it up,” Mirkin said. “The SNA architecture is recognized by almost all cell types, so cells actively take up the SNAs and rapidly internalize them.”

    Three-fold improved performance

    After successfully synthesizing LNP-SNAs with CRISPR cargo, Mirkin and his team added them to cellular cultures, which included skin cells, white blood cells, human bone marrow stem cells, and human kidney cells. 

    The team observed and measured several key factors:

    • How efficiently the cells internalised the particles
    • Whether the particles were toxic to cells
    • If the particles successfully deliver a gene

    They also analysed the cells’ DNA to determine if CRISPR had made the desired gene edits, finding that across every factor, the system successfully delivered CRISPR machinery and enabled complex genetic modifications.

    In the future, Mirkin plans further to validate the system in multiple in vivo disease models. Because the platform is modular, researchers can adapt it for a wide range of systems and therapeutic applications.

    “CRISPR could change the whole field of medicine,” Mirkin said. “But how we design the delivery vehicle is just as important as the genetic tools themselves. By marrying two powerful biotechnologies — CRISPR and SNAs — we have created a strategy that could unlock CRISPR’s full therapeutic potential.”

    Continue Reading

  • Late-Stage Incidence Rates Continue to Increase Rapidly as Mortality Declines Slow

    Late-Stage Incidence Rates Continue to Increase Rapidly as Mortality Declines Slow

    The American Cancer Society study shows the highest mortality rates are among Black and Native American men

    ATLANTA, Sept. 2, 2025 /PRNewswire/ — Today, the American Cancer Society (ACS) released Prostate Cancer Statistics, 2025, a report on current prostate cancer occurrence and outcomes in the United States. According to the study, prostate cancer incidence rates have reversed from a decline of 6.4% per year during 2007 through 2014 to an increase of 3.0% annually during 2014 through 2021, with the steepest increase (4.6%-4.8% per year) for advanced-stage diagnoses. Simultaneously, mortality declines slowed from 3%-4% per year during the 1990s and 2000s to 0.6% per year over the past decade. These major findings are to be published today in the journal CA: A Cancer Journal for Clinicians.

    The report also shows wide disparities. Mortality rates for prostate cancer are two times higher for Black men compared to White men, versus 67% higher incidence rates. Likewise, Native American men have 12% higher prostate cancer mortality than White men, despite 13% lower incidence.

    “Our research highlighting the continued increases in prostate cancer incidence and persistent racial disparities underscores the need for redoubled efforts to understand the etiology of prostate cancer and optimize early detection,” said Tyler Kratzer, MPH, associate scientist, cancer surveillance research at the American Cancer Society, and lead author of the study. “At age 50, per ACS guidelines, all men should have a conversation with their healthcare provider about the benefits and harms of screening, but Black men and those with a family history of prostate cancer should have that conversation at age 45.”

    Prostate cancer is the most common cancer diagnosis among men in the U.S., accounting for  30% of male cancers in 2025, and is the second leading cancer death in men behind lung cancer. This year, ACS estimates there will be 313,780 new cases of prostate cancer and 35,770 deaths. For the report, researchers analyzed population-based cancer incidence data through 2021 and mortality data through 2023 collected by the National Cancer Institute and the Centers for Disease Control and Prevention.

    According to study authors, distant‐stage disease is increasing in men of every age, including by nearly 3% per year in those younger than 55 years and 6% per year in men 55 years and older. The five-year relative survival rate for distant-stage prostate cancer is only 38%, but approaches 100% for earlier-stage diagnoses.

    Other key findings from the report include:

    • American Indian and Alaska Native men are the most likely to be diagnosed with distant stage disease (12% versus 8% among White men).
    • Prostate cancer mortality ranges from 36.9 deaths per 100,000 among Black men to 8.8 among Asian American and Pacific Islander men. American Indian and Alaska Native men have the second-highest mortality rates (20.6), with White (18.4) and Hispanic (15.4) men ranking 3rd and 4th among broadly defined racial and ethnic groups.
    • Prostate cancer mortality varies by state, with the highest death rates in Washington D.C. (27.5 deaths per 100,000) and Mississippi (24.8 deaths per 100,000), which have a high proportion of Black residents.

    “Our report underscores the need to redouble efforts to optimize early diagnosis that minimizes overdetection and to ensure that these strategies reach Black and Native American communities in particular,” said Rebecca Siegel, MPH, senior scientific director, cancer surveillance research at the American Cancer Society and senior author of the report. “All men deserve the same opportunity to survive this common cancer.”

    The American Cancer Society Cancer Action Network (ACS CAN), the advocacy affiliate of the American Cancer Society, supports the Prostate-Specific Antigen Screening for High-risk Insured Men (PSA Screening for HIM) Act, federal legislation that would waive cost-sharing requirements such as deductibles, copayments, and coinsurance for prostate cancer screening tests for men with the highest risk of prostate cancer.

    “Out-of-pocket costs such as co-pays can be a barrier to accessing early detection,” said Lisa A. Lacasse, president of ACS CAN. “No one should be at a disadvantage against cancer. The PSA Screening for HIM Act will help remove a major obstacle that can prevent those at high risk for the disease from getting the screening tests they need to find prostate cancer at the earliest, most treatable stage. We urge the House and the Senate to pass this legislation to help reduce prostate cancer disparities and save more lives.”https://www.cancer.org/about-us/who-we-are/executive-leadership/lisa-lacasse-bio.html

    Other ACS researchers contributing to the study include Natalia Mazzitelli, MPH, Jessica Star, MPH, Dr. William Dahut, and Dr. Ahmedin Jemal.

    Additional ACS Resources:

    About the American Cancer Society
    The American Cancer Society is a leading cancer-fighting organization with a vision to end cancer as we know it, for everyone. For more than 110 years, we have been improving the lives of people with cancer and their families as the only organization combating cancer through advocacy, research, and patient support. We are committed to ensuring everyone has an opportunity to prevent, detect, treat, and survive cancer. To learn more, visit cancer.org or call our 24/7 helpline at 1-800-227-2345. Connect with us on Facebook, X, and Instagram. 

    SOURCE American Cancer Society


    Continue Reading

  • Mi-Lnc70 regulates the progression of murine pancreatic β-cell Line a

    Mi-Lnc70 regulates the progression of murine pancreatic β-cell Line a

    Introduction

    Insulinoma is a type of pancreatic endocrine tumor that often causes hypoglycemia due to persistent hyperinsulinemia.1 Accumulating evidence has shown that the expression of certain long non-coding RNAs (lncRNAs) in β-cells is regulated by extracellular glucose concentration-, indicating that lncRNAs may be involved in the regulation of insulin secretion.2–6 The mapping of some pancreatic lncRNA genes has uncovered their association with human diabetes. Moreover, specific lncRNAs have been found to be dysregulated in pancreatic islets with type 2 diabetes (T2D), suggesting that lncRNAs may serve as potential regulatory factors in diabetes research.2,7 These findings imply that lncRNAs may play a role in the synthesis or secretion of insulin in insulinoma.

    Mutations or aberrant expression of protein-coding genes, as well as mutations and dysregulation of lncRNAs, play a significant role in the pathogenesis of cancer. Genome-wide association studies (GWAS) of tumor samples have identified a considerable number of lncRNAs associated with various types of cancer. LncRNAs may exhibit tumor suppressive or promoting functions, and their abnormal expression and mutations are closely related to tumorigenesis, metastasis, and tumor staging. They also have diagnostic and prognostic value.8–11 For instance, a study revealed that lncRNA NUTF2P3-001 elevated K-Ras expression after hypoxia induction and facilitated the proliferation of Panc-1 and Bxpc-3 cell lines.12 The long non-coding RNA (lncRNA) HOTTIP has been shown to enhance the expression of HOXA genes by binding to the WDR5/MLL1 complex. This interaction plays a crucial role in promoting the proliferation, survival, and migration of pancreatic cancer cells.13

    Mi-Lnc70 is an islet-specific lncRNA located between the hepatic nuclear factor 1A (Hnf1a) and signal peptide peptidase-like 3 (Sppl3) genes on mouse chromosome 5, and it is a partial antisense transcript of Hnf1a. During pancreatic maturation in mice, the expression of Mi-Lnc70 gradually increases, although its expression level is not regulated by glucose.9 HNF1A has been identified as a tumor suppressor in pancreatic cancer,14–16 capable of inhibiting the proliferation and migration of pancreatic cancer cells.16–18 Moreover, HNF1A is crucial for maintaining the function of human pancreatic α and β cells.19,20 Mutations or deletions in HNF1A lead to pancreatic β cell dysfunction and are closely associated with the development of diabetes.21–23 Given the significant role of HNF1A in pancreatic cell function and its proximity to Mi-Lnc70, this study aims to explore the biological function of Mi-Lnc70 in the mouse pancreatic β-cell line (MIN6), providing a foundation for elucidating the molecular mechanisms related to pancreatic tumors.

    Material and Methods

    Cell Line and Cell Culture

    The murine pancreatic β-cell line MIN6 (RRID: CVCL_0431) was obtained from Xiubin Liang’s laboratory in the Department of Pathophysiology at Nanjing Medical University and used for experimental studies. MIN6 cells were maintained in DMEM (Gibco) supplemented with 15% fetal bovine serum (FBS, Hyclone) and 50 μmol/L β-mercaptoethanol (Sigma) at 37°C in a humidified incubator with 5% CO₂. Prior to cell seeding, culture dishes were coated with gelatin (Sigma). Mouse embryonic fibroblast (MEF) cells were cultured in DMEM supplemented with 10% FBS. Cells were passaged at a 1:3 ratio using 0.05% trypsin (Gibco) and the medium was changed daily. All cell line usage and animal procedures were conducted in strict accordance with the guidelines established by the Inner Mongolia University Animal Care and Use Committee and were approved under license number IMU-MOUSE-2018-012.

    Cell Transfection

    Cells were transfected with Mi-Lnc70-specific antisense locked nucleic acid (LNA) GapmeR using HiPerFect Transfection Reagent (QIAGEN), with nontargeting siRNA serving as a negative control. Twenty-four hours post-transfection, the culture medium was replaced with an appropriate volume of complete culture medium, and the cells were incubated for an additional 72 hours. Cell status was subsequently examined under a microscope. The sequences of the siRNAs targeting Mi-Lnc70 are detailed in Table S1.

    RNA Extraction and Real-Time PCR

    Total RNA was extracted from cells using Trizol reagent (TransGene Biotech) according to the manufacturer’s instructions. The extracted RNA was reverse transcribed into cDNA using the PrimeScript RT Reagent Kit with gDNA Eraser (RR047A, Takara). For miRNA reverse transcription, the TransScript® miRNA First-Strand cDNA Synthesis SuperMix (TransGene Biotech) was used. Quantitative real-time PCR was performed using TB GREEN™ Premix Ex Taq™ II (Takara) on a 7500 Real-Time PCR System (ABI Biosystems). The mouse U6 gene was used as the housekeeping gene for normalization. The relative expression levels of target genes were determined using the comparative CT method (ΔΔCT). Primer sequences used in the study are listed in Table S2.

    Cell Counting Kit (CCK)-8 Assay

    For the cell proliferation assay, 1×10⁴ cells suspended in 100 μL of culture medium were seeded into each well of a 96-well plate. After cell attachment, 100 μL of CCK-8 reagent mixture (prepared by mixing 10 μL of CCK-8 reagent with 90 μL of DMEM) was added to each well. The plates were then incubated at 37°C in the dark for 2 hours. The absorbance at 450 nm was measured using a microplate reader (Molecular Devices). Data were analyzed using GraphPad Prism version 8.0.

    Cell Cycle Assay

    Cells in the logarithmic growth phase were seeded into a 24-well plate at a density of 9×10⁵ cells/mL. After 24 hours of culture, cells were transfected as described previously. At 72 hours post-transfection, MIN6 cells were collected, washed twice with cold DPBS, and immediately fixed in cold 75% ethanol at 4°C for a minimum of 4 hours. Following fixation, cells were washed once with DPBS and stained with a propidium iodide (PI) solution containing 50 μg/mL PI and 100 μg/mL RNase A. Cells were then incubated at 4°C for 30 minutes in the dark. Flow cytometric analysis was performed within 1 hour to determine the cell cycle distribution.

    Apoptosis Assay

    Cells in the logarithmic growth phase were seeded into 24-well plates at a density of 9×10⁵ cells/mL. After 24 hours of culture, cells were transfected with Mi-Lnc70-specific siRNA. At 72 hours post-transfection, cell pellets were collected. Apoptosis was assessed using the Annexin V-FITC Apoptosis Detection Kit (Solarbio) according to the manufacturer’s instructions. Briefly, cells were resuspended in binding buffer, stained with Annexin V-FITC and propidium iodide (PI), and analyzed by flow cytometry within 1 hour to determine the apoptosis rate.

    Wound Healing Assay

    MIN6 cells were seeded in 24-well plates at a density of 9×10⁵ cells per well and cultured for 24 hours. A linear scratch was created in the cell monolayer using a 100 μL micropipette tip. Non-adherent cells were removed by washing with DPBS. Subsequently, cells were transfected with Mi-Lnc70-specific siRNA as described previously. Images of the scratch wounds were captured at 0 hours and 72 hours using a microscope. The wound-healing rate was calculated using the following formula: 100% × [(wound width at 0 h − wound width at 72 h) / wound width at 0 h].

    Transwell Migration and Invasion Assay

    Transwell assays were conducted using a transwell chamber with a pore diameter of 8 μm (Corning). MIN6 cells, transfected with Mi-Lnc70-specific siRNA, were resuspended in serum-free medium and seeded into the upper chamber at a density of 5×10⁴ cells in 100 μL. The lower chamber was filled with complete medium containing 10% fetal bovine serum as a chemoattractant. After 20–24 hours of incubation at 37°C in a 5% CO₂ incubator, cells that had migrated to the lower surface of the membrane were fixed with absolute ethanol and stained with DAPI (Beyotime) for 2–3 minutes at room temperature. Cells remaining on the upper surface were gently removed with a cotton swab. Images were captured using an inverted light microscope (Nikon). For the invasion assay, the procedure was identical to the migration assay, except that the upper chamber was coated with Matrigel to mimic the extracellular matrix.

    Cell Total Protein Extraction and Western-Blot

    Cells were collected and lysed in lysis buffer (Thermo Fisher Scientific) supplemented with phenylmethylsulfonyl fluoride (Beyotime) on ice for 30 minutes. The lysates were then heated at 100 °C for 10 minutes, followed by centrifugation at 4°C for 10 minutes at 12000×g. The supernatant was collected, and protein concentration was determined using the BCA assay (Thermo Fisher Scientific). Total protein was separated by 10% SDS-PAGE and transferred to PVDF membranes (Millipore). Membranes were blocked with 5% skim milk powder in TBST for 1 hour at room temperature, and then incubated with primary antibodies overnight at 4°C. After washing with TBST, membranes were incubated with secondary antibodies for 1 hour at room temperature. Target protein bands were visualized using an enhanced chemiluminescence (ECL) detection system (Thermo Fisher Scientific) and captured using an imaging analysis system (Bio-Rad). The primary and secondary antibodies used in this study are listed in Tables S3 and S4.

    Immunofluorescence

    MIN6 cells were washed once with DPBS and fixed in 4% paraformaldehyde (PFA; Solarbio) for 30 minutes at room temperature. After fixation, cells were permeabilized with 0.5% Triton X-100 (Solarbio) for 30 minutes. Subsequently, cells were blocked for 2 hours at room temperature using a blocking solution containing 2% BSA, 2% goat serum, 2% skim milk powder, and 0.15 M glycine in DPBS. The samples were then incubated with primary antibodies overnight at 4°C. Following three washes with DPBS, the samples were incubated with secondary antibodies for 1 hour at room temperature. After two additional washes with DPBS, the samples were stained with DAPI (0.5 mg/mL; Beyotime) for 3 minutes at room temperature. The samples were then washed twice with DPBS and mounted onto slides using antifading Mounting Medium (Solarbio). Fluorescent signals were visualized using a Nikon confocal laser-scanning microscope (Nikon, A1). The primary and secondary antibodies used in this study are listed in Tables S5 and S6.

    Statistical Analyses

    Data are presented as the mean ± standard deviation (SD) from at least three independent experiments. Statistical significance between two groups was determined using Student’s t-test, whereas one-way analysis of variance (ANOVA) was employed to analyze data from more than three groups. Significance levels were defined as follows: *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.

    Results

    Expression Analysis of Pancreatic Islet-Specific lncRNAs in MIN6 Cells

    To elucidate the role of lncRNAs in insulinoma, we initially utilized real-time fluorescence quantitative PCR to measure the expression levels of several pancreatic islet-specific lncRNAs in the mouse pancreatic β-cell line MIN6. Our results revealed that Mi-Lnc70 exhibited a significantly higher expression level compared to other lncRNAs such as Mi-Lnc72, Mi-Lnc75, Mi-Lnc76, and Mi-Lnc80 (Figure 1A and B). This finding indicates that Mi-Lnc70 may exert a crucial regulatory function in mouse pancreatic β-cells. Therefore, we selected Mi-Lnc70 for further investigation.

    Figure 1 The expression of pancreatic islet-specific lncRNAs was analyzed in MIN6 cells. (A) Morphological in MEF and MIN6 cells during culture. Scale bars: 100 μm. (B) The relative expression of pancreatic islet-specific Mi-LncRNAs in the MIN6 cells. ****p < 0.0001. (C) The relative expression of Mi-Lnc70 in the MIN6 cells under different transfection conditions is presented below. GapmeR-1#/2#/3# are different siRNAs designed for Mi-Lnc70, and GapmeRs mix is a mixture of GapmeR-1#, GapmeR-2# and Gapme-R3#. *p < 0.05, ****p < 0.0001.

    To explore the role of Mi-Lnc70 in pancreatic β-cells, we employed GapmeR antisense oligonucleotides to specifically knockdown Mi-Lnc70 (si-Lnc70) and used a nonsense oligonucleotide as a negative control (NC). Post-transfection, the expression level of Mi-Lnc70 in the si-Lnc70 group was significantly reduced compared to both the NC group and the blank control group (Figure 1C).

    Inhibition of MIN6 Cell Proliferation by Knocking Down Mi-Lnc70

    To elucidate the role of Mi-Lnc70 in mouse pancreatic β cells, we first investigated the effect of Mi-Lnc70 knockdown on the proliferation of MIN6 cells in vitro. The CCK-8 assay was used to assess the impact of Mi-Lnc70 on cell proliferation. We constructed a Mi-Lnc70 knockdown group in the MIN6 cell line and found that the proliferation ability of the si-Mi-Lnc70 group was significantly lower than that of the negative control (NC) group and the blank control group (Figure 2A).

    Figure 2 The Mi-Lnc70 construct was found to promote the proliferation of MIN6 cells. (A) The activity of MIN6 cells transfected with NC and si-Lnc70. (B) The detection of apoptosis in MIN6 cells transfected with NC and si-Lnc70. (C) The ratio of early apoptotic cells to late apoptotic cells in (B). (D) The cell cycle of MIN6 cells transfected with NC and si-Lnc70 was also analyzed.

    Apoptosis detection in the MIN6 cell line after transfection revealed that the proportion of late apoptotic cells (Q2: 31.9%) increased, while the proportion of live cells decreased following Mi-Lnc70 knockdown (Figure 2B and C). Furthermore, flow cytometry analysis of the cell cycle demonstrated an increased proportion of cells in the G2/M phase after Mi-Lnc70 knockdown, indicating G2/M phase arrest (Figure 2D). Collectively, these results indicate that down-regulation of Mi-Lnc70 induces G2/M phase arrest in MIN6 cells, thereby reducing their proliferation ability and promoting apoptosis.

    Knockdown of Mi-Lnc70 Reduces the Migration and Invasion Abilities of MIN6 Cells

    To further investigate the function of Mi-Lnc70 in mouse pancreatic β cells, we examined the effects of Mi-Lnc70 knockdown on the migration, invasion, and wound-healing abilities of MIN6 cells in vitro. The Transwell assay was employed to assess the migration and invasion capabilities of MIN6 cells. As depicted in Figure 3A and B, the migration capacity of the si-Mi-Lnc70 group exhibited a substantial decline. Specifically, it was diminished by approximately 39.7% relative to the blank control group and by approximately 42.3% in comparison with the negative control (NC) group. Consistent with the migration results, the invasive capacity of the MIN6 cell line within the si-Mi-Lnc70 group was likewise markedly attenuated. Quantitatively, it was decreased by approximately 66.8% when juxtaposed with the blank control group and by approximately 66.7% relative to the negative control (NC) group, as illustrated in Figure 3C and D. Additionally, wound-healing assays revealed that Mi-Lnc70 knockdown led to a decreased wound-healing capacity in MIN6 cells (Figure 3E and F). Collectively, these findings suggest that downregulation of Mi-Lnc70 results in reduced migration, invasion, and wound-healing abilities in MIN6 cells.

    Figure 3 Mi-Lnc70 enhanced the migration and invasion ability of MIN6 cells. (A and B) Transwell assays were employed to assess the migratory capacity of MIN6 cells transfected with NC and si-Lnc70. (C and D) Transwell invasion assays demonstrated the invasion capacity of MIN6 cells transfected with NC and si-Lnc70. ***p < 0.001. (E and F) Wound healing assay of MIN6 cells transfected with NC and si-Lnc70. ****p < 0.0001.

    Knockdown of Mi-Lnc70 Reduces the Expression of Pancreas-Related LncRNAs and MiRNAs in MIN6 Cells

    To elucidate the underlying mechanisms, we examined the impact of Mi-Lnc70 knockdown on the expression of pancreas-related lncRNAs and miRNAs in MIN6 cells. Comparative analysis of the relative expression levels of these lncRNAs and miRNAs before and after transfection revealed significant changes in the expression profiles of multiple lncRNAs and miRNAs in MIN6 cells following Mi-Lnc70 knockdown. Among the lncRNAs, the expression levels of Mi-Lnc77, Mi-Lnc87, Mi-Lnc78, and Mi-Lnc85 were upregulated, while Mi-Lnc12 and Mi-Lnc67 showed no significant changes. In contrast, the expression levels of Mi-Lnc71, Mi-Lnc72, Mi-Lnc75, Mi-Lnc76, and Mi-Lnc80 were significantly downregulated (Figure 4A–C).

    Figure 4 Knockdown of Mi-Lnc70 had effects on the expression of pancreas-related lncRNAs and miRNAs in MIN6 cells. (A) The lncRNAs with reduced expression level are presented. *p < 0.05, **p < 0.01, ***p < 0.001. (B) The lncRNAs with unchanged expression level are presented. (C) The lncRNAs with increased expression level are presented. *p < 0.05. (D) The miRNAs with unchanged expression level are presented. (E) The miRNAs with reduced expression level are presented. *p < 0.05, **p < 0.01, ***p < 0.001.

    Furthermore, the knockdown of Mi-Lnc70 in MIN6 cells also affected the expression of pancreas-related miRNAs. While the expression levels of miR-195-3p, miR-15a-5p, and miR-30d-3p remained comparable to those in untransfected cells, the expression levels of miR-23b-3p, miR-124-1-3p, miR-124-1-5p, miR-16-1-5p, miR-7, miR-195-5p, miR-15b-5p, and miR-15b-3p were significantly decreased, with the most pronounced reductions observed for miR-7, miR-195-5p, and miR-15b-3p (Figure 4D and E). These findings suggest that Mi-Lnc70 may regulate the expression of genes involved in the proliferation, apoptosis, migration, and invasion abilities of MIN6 cells by modulating the expression of specific miRNAs, particularly miR-7, miR-195-5p, and miR-15b-3p.

    Knockdown of Mi-Lnc70 Reduces the Synthesis of Secretion-Related Proteins in MIN6 Cells

    To investigate the effect of Mi-Lnc70 knockdown on insulin synthesis in MIN6 cells, we quantified the relative expression levels of insulin and related factors post-transfection. Figure 5A showed that the knockdown of Mi-Lnc70 resulted in a significant downregulation of the expression levels of insulin and its associated factors, which encompass Insulin1, Insulin2, Somatostatin, Pax4, MafA, Isl-1, NeuroD1, Pax6, Pdx1, Nkx6.1, and Glucagon. Immunofluorescence staining revealed diminished expression of PDX1, C-peptide, insulin, glucagon, and somatostatin in MIN6 cells following Mi-Lnc70 knockdown. Notably, a significant reduction in somatostatin expression was observed in MIN6 cells transfected with si-Lnc70 (Figure 5B). Western blot analysis further revealed that the protein expression levels of insulin, glucagon, and PDX1 in Mi-Lnc70 knockdown cell lines were significantly downregulated compared with those in negative control cells and non-transfected cells, as shown in Figure 5C and D. Collectively, these findings indicate that the knockdown of Mi-Lnc70 inhibits the synthesis of PDX1, INSULIN, and GLUCAGON in MIN6 cells (Figure 6).

    Figure 5 Mi-Lnc70 affects the synthesis of islet-related secreted proteins in MIN6 cells. (A) Relative expression levels of insulin and related genes in MIN6 cells transfected with NC and si-Lnc70. *p < 0.05, **p < 0.01, ***p < 0.001. (B) Immunofluorescence staining of insulin and related genes in MIN6 cells transfected with NC and si-Lnc70 (n=3). Nuclei were stained with DAPI. Scale bars, 100 μm. (C and D) Expression of INSULIN, GLUCAGON and PDX1 proteins in MIN6 cells transfected with NC and si-Lnc70. **p < 0.01, ***p < 0.001.

    Figure 6 Mi-Lnc70 Promotes Pancreatic β-Cell Progression and Affects the Synthesis of PDX1, Insulin, and Glucagon. Mi-Lnc70 was found to be significantly increased in MIN6 cells. Knockdown of Mi-Lnc70 significantly inhibited the proliferation, migration, and invasion of MIN6 cells while promoting cell apoptosis and G2/M phase arrest of the cell cycle. Additionally, Mi-Lnc70 knockdown reduced the expression of pancreas-related lncRNAs and miRNAs in MIN6 cells, as well as the synthesis of secretion-related proteins, including PDX1, INSULIN, and GLUCAGON.

    Discussion

    The most significant biological characteristics of tumor cells are their capacities for migration and invasion, which are closely related to the degree of differentiation of the tumor cells. Poorly differentiated tumor cells typically exhibit enhanced proliferative capacity. Previous studies have demonstrated that Malat1 can regulate the proliferation, migration, and invasion of cancer cells by modulating the expression of multiple downstream genes, including cell cycle regulatory genes such as p21, p27, and B-Myb. A reduction in Malat1 expression can result in the arrest of cancer cells in the G2/M phase of the cell cycle.24 In this study, we observed that the downregulation of Mi-Lnc70 in MIN6 cells led to significant inhibition of migration and invasion abilities, decreased wound-healing capacity, increased cell apoptosis, and cell cycle arrest in the G2/M phase. These findings suggest that Mi-Lnc70 plays a crucial role in regulating the biological behavior of MIN6 cells, potentially through mechanisms similar to those involving Malat1.

    The miRNAs are pivotal regulators of gene expression, influencing multiple cellular pathways and the development and progression of cancer.25 Long non-coding RNAs (lncRNAs) can act as competing endogenous RNAs (ceRNAs) or natural miRNA sponges, significantly impacting gene expression through post-transcriptional regulation. This regulatory mechanism involves competition for binding to shared miRNAs, leading to mutual regulation. Data from human pancreatic ductal adenocarcinoma (PDAC) tissues and adjacent normal tissues have shown a significant inverse correlation between miR-216a and Malat1. Overexpression of miR-216a or knockout of Malat1 has been found to affect the cell cycle of pancreatic cancer cells, with miR-216a overexpression inducing effects similar to those of si-Malat1, including G2/M phase arrest.26 Additionally, lncRNA HNF1A-AS1 can interact with miRNAs and play a role in various cancers. HNF1A-AS1 can competitively bind to miRNAs (such as miR-149-5p, miR-30b-3p, miR-124) to relieve the inhibitory effects of these miRNAs on target genes, thereby promoting the expression of the target genes.27–29 Conversely, HNF1A-AS1 can also synergize with miRNAs (such as miR-32-5p, miR-30b-5p, miR-363) to relieve the inhibition of downstream target genes, promoting the expression of common downstream target genes.30–32 In this study, we observed a significant positive correlation between certain pancreatic development-related miRNAs and Mi-Lnc70, including miR-23b-3p, miR-124-1-5p, miR-7, miR-195-5p, miR-15b-5p, and miR-15b-3p. We hypothesize that miR-23b-3p, miR-124-1-5p, miR-7, miR-195-5p, miR-15b-5p, and miR-15b-3p may bind to Mi-Lnc70 and regulate each other. However, Mi-Lnc70 may function as a ceRNA or may cooperate with miRNAs to regulate downstream genes. Therefore, the specific mechanisms by which Mi-Lnc70 interacts with miR-23b-3p, miR-124-1-5p, miR-7, miR-195-5p, miR-15b-5p, and miR-15b-3p in MIN6 cells remain to be elucidated. Future studies should focus on deciphering these interactions to better understand the regulatory roles of Mi-Lnc70 in pancreatic β-cell function and tumorigenesis.

    Previous studies have demonstrated that a fraction of lncRNAs are primary functional targets of islet-specific transcription factors and key cis-regulatory determinants of transcriptional programs in islet cells. Additionally, a considerable proportion of lncRNAs exhibit lineage-specific expression patterns. MafA and Pdx1 play pivotal roles in maintaining the development and maturation of pancreatic β cells. By binding to the insulin promoter region, they regulate related transcription factors, thereby influencing the synthesis and secretion of insulin.33,34 For instance, lncRNA MALAT1 can inhibit the expression of PDX1 and insulin secretion by reducing H3 histone acetylation of the PDX1 promoter, thereby inducing β-cell dysfunction.35 Downregulation of lncRNA HOTTIP regulates insulin secretion and the cell cycle in islet β cells via the inhibition of the MEK/ERK pathway.36 Moreover, lncRNA PLUTO can regulate the transcription of PDX1 in mature pancreatic β cells.37,38 Our results showed that downregulation of Mi-Lnc70 led to decreased expression of miRNAs such as miR-7, miR-195-5p, miR-15b-5p, and miR-15b-3p, as well as decreased expression of downstream factors such as PDX1, MafA, and Nkx6.1, ultimately resulting in reduced insulin synthesis. Therefore, we speculate that Mi-Lnc70 may bind to miRNAs such as miR-7, miR-195-5p, miR-15b-5p, and miR-15b-3p, acting as a ceRNA or coordinately regulating the expression of downstream factors such as PDX1 and MafA in MIN6 cells. This regulatory mechanism may ultimately lead to a reduction in insulin synthesis. Future studies should focus on elucidating the specific interactions between Mi-Lnc70 and these miRNAs, as well as their downstream targets, to better understand the regulatory roles of Mi-Lnc70 in pancreatic β-cell function and insulinoma pathogenesis.

    In summary, our study demonstrates that the downregulation of Mi-Lnc70 significantly inhibits the migration, invasion, and wound-healing capacities of MIN6 cells while simultaneously promoting cell apoptosis. Additionally, the expression of pancreas-related genes, as well as multiple lncRNAs and miRNAs, is concomitantly reduced. These findings highlight the crucial regulatory role of Mi-Lnc70 in MIN6 cells. However, it is important to acknowledge that in vitro models have inherent limitations. Therefore, the precise mechanisms by which Mi-Lnc70 regulates the mouse pancreatic β-cell lineage require further validation through in vivo studies. Future research should focus on elucidating the detailed molecular pathways and interactions involving Mi-Lnc70 in pancreatic β-cell function and tumorigenesis, ultimately providing a more comprehensive understanding of its role in insulinoma.

    Conclusion

    Mi-Lnc70 plays an important role in the proliferation, migration, and endocrine-related gene expression in pancreatic MIN6 cells, particularly in the synthesis of PDX1, INSULIN, and GLUCAGON.

    Abbreviations

    lncRNAs, long non-coding RNAs; CCK-8, cell counting kit-8; miRNAs, micro RNAs; PDX1, pancreatic and duodenal homeobox 1; EMT, epithelial-mesenchymal transition; PI3K/ AKT, phosphoinositide 3-kinase/protein kinase B; K-Ras, kirsten rat sarcoma virus; APC, adenomatous polyposis coli; ERK, extra-cellular signal regulated kinase; MAPK, mitogen-activated protein kinase; PKC, protein kinase C; T2D, type 2 diabetes; NUTF2P3-001, nuclear transport factor 2 pseudogene 3-001; MALAT1, metastasis associated lung adenocarcinoma transcript 1; HOTTIP, HOXA distal transcript antisense RNA; HOXA, homebox A10; WDR5/MLL1, WD repeat-containing protein 5/mixed lineage leukemia 1; Sppl3, signal peptide peptidase-like 3; Hnf1a, hepatic nuclear factor 1A; B-Myb, Myb-related protein B; PDAC, pancreatic ductal adenocarcinoma; PAX4, paired box 4; PAX6, paired box 6; Nkx6.1, NK6 Homeobox 1; Isl-1, Islet-1; NeuroD1, neurogenic differentiation 1; MafA, MAF BZIP transcription factor A.

    Author Contributions

    All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

    Data Sharing Statement

    The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding authors.

    Consent for Publication

    All authors have provided their consent for publication.

    Acknowledgments

    We thank Dr. Xiubin Liang of the Department of Pathophysiology of Nanjing Medical University for supplying the MIN6 cells. This paper has been uploaded to China National Knowledge Infrastructure (CNKI) as a thesis: https://www.cnki.net/.

    Funding

    This work was financially supported by the National Natural Science Foundation of China (81471001).

    Disclosure

    The authors declared that there is no competing interest in this work.

    References

    1. Fu J, Zheng H, Cui Q, et al. Nfe2l1-silenced insulinoma cells acquire aggressiveness and chemoresistance. Endocrine-Related Cancer. 2018;25(3):185–200. doi:10.1530/ERC-17-0458

    2. Moran I, Akerman I, van de Bunt M, et al. Human beta cell transcriptome analysis uncovers lncRNAs that are tissue-specific, dynamically regulated, and abnormally expressed in type 2 diabetes. Cell Metab. 2012;16(4):435–448. doi:10.1016/j.cmet.2012.08.010

    3. Yang W, Lyu Y, Xiang R, Yang J. Long noncoding RNAs in the pathogenesis of insulin resistance. Int J Mol Sci. 2022;23(24):16054. doi:10.3390/ijms232416054

    4. Sathishkumar C, Prabu P, Mohan V, Balasubramanyam M. Linking a role of lncRNAs (long non-coding RNAs) with insulin resistance, accelerated senescence, and inflammation in patients with type 2 diabetes. Human Genomics. 2018;12(1):41. doi:10.1186/s40246-018-0173-3

    5. Formichi C, Nigi L, Grieco GE, et al. Non-coding RNAs: novel players in insulin resistance and related diseases. Int J Mol Sci. 2021;22(14):7716. doi:10.3390/ijms22147716

    6. Zhong G, Yang Q, Wang Y, Liang Y, Wang X, Zhao D. Long noncoding RNA X-inactive specific transcript (lncRNA XIST) inhibits hepatic insulin resistance by competitively binding microRNA-182-5p. Immunity, Inflammation and Disease. 2023;11(11):e969. doi:10.1002/iid3.969

    7. Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014;15(1):7–21. doi:10.1038/nrg3606

    8. Bhan A, Soleimani M, Mandal SS. Long noncoding RNA and cancer: a new paradigm. Cancer Res. 2017;77(15):3965–3981. doi:10.1158/0008-5472.CAN-16-2634

    9. Bartonicek N, Maag JL, Dinger ME. Long noncoding RNAs in cancer: mechanisms of action and technological advancements. Mol Cancer. 2016;15(1):43. doi:10.1186/s12943-016-0530-6

    10. Kornfeld JW, Bruning JC. Regulation of metabolism by long, non-coding RNAs. Front Genet. 2014;5:57. doi:10.3389/fgene.2014.00057

    11. Vitiello M, Tuccoli A, Poliseno L. Long non-coding RNAs in cancer: implications for personalized therapy. Cell Oncology. 2015;38(1):17–28. doi:10.1007/s13402-014-0180-x

    12. Bermont F, Hermant A, Benninga R, et al. Targeting mitochondrial calcium uptake with the natural flavonol kaempferol, to promote metabolism/secretion coupling in pancreatic beta-cells. Nutrients. 2020;12(2):538. doi:10.3390/nu12020538

    13. Cheng Y, Jutooru I, Chadalapaka G, Corton JC, Safe S. The long non-coding RNA HOTTIP enhances pancreatic cancer cell proliferation, survival and migration. Oncotarget. 2015;6(13):10840–10852. doi:10.18632/oncotarget.3450

    14. Hoskins JW, Jia J, Flandez M, et al. Transcriptome analysis of pancreatic cancer reveals a tumor suppressor function for HNF1A. Carcinogenesis. 2014;35(12):2670–2678. doi:10.1093/carcin/bgu193

    15. Kalisz M, Bernardo E, Beucher A, et al. HNF1A recruits KDM6A to activate differentiated acinar cell programs that suppress pancreatic cancer. THE EMBO Journal. 2020;39(9):e102808. doi:10.15252/embj.2019102808

    16. Luo Z, Li Y, Wang H, et al. Hepatocyte nuclear factor 1A (HNF1A) as a possible tumor suppressor in pancreatic cancer. PLoS One. 2015;10(3):e0121082. doi:10.1371/journal.pone.0121082

    17. Yu Y, Liang S, Zhou Y, Li S, Li Y, Liao W. HNF1A/CASC2 regulates pancreatic cancer cell proliferation through PTEN/Akt signaling. Journal of Cellular Biochemistry. 2019;120(3):2816–2827. doi:10.1002/jcb.26395

    18. Gong Y, Dai HS, Shu JJ, Liu W, Bie P, Zhang LD. LNC00673 suppresses proliferation and metastasis of pancreatic cancer via target miR-504/ HNF1A. Journal of Cancer. 2020;11(4):940–948. doi:10.7150/jca.32855

    19. Qian MF, Bevacqua RJ, Coykendall VM, et al. HNF1α maintains pancreatic α and β cell functions in primary human islets. JCI Insight. 2023;8(24). doi:10.1172/jci.insight.170884.

    20. Nhj N, Ghosh S, Bok CM, et al. HNF4A and HNF1A exhibit tissue specific target gene regulation in pancreatic beta cells and hepatocytes. Nature Communications. 2024;15(1):4288. doi:10.1038/s41467-024-48647-w

    21. Miyachi Y, Miyazawa T, Ogawa Y. HNF1A mutations and beta cell dysfunction in diabetes. Int J Mol Sci. 2022;23(6):3222. doi:10.3390/ijms23063222

    22. Chen Y, Jia J, Zhao Q, et al. Novel loss-of-function variant in HNF1a induces β-cell dysfunction through endoplasmic reticulum stress. Int J Mol Sci. 2022;23(21).

    23. Nocera D, Menniti M, Belviso S, et al. Functional characterization of p.Pro409His variant in HNF1A, a hypomorphic mutation involved in pancreatic β-cell dysfunction. Acta diabetologica. 2019;56(8):883–888. doi:10.1007/s00592-019-01298-6

    24. Zhang Y, Tang X, Shi M, Wen C, Shen B. MiR-216a decreases MALAT1 expression, induces G2/M arrest and apoptosis in pancreatic cancer cells. Biochem Biophys Res Commun. 2017;483(2):816–822. doi:10.1016/j.bbrc.2016.12.167

    25. Hussen BM, Hidayat HJ, Salihi A, Sabir DK, Taheri M, Ghafouri-Fard S. MicroRNA: a signature for cancer progression. Biomed Pharmacother. 2021;138:111528. doi:10.1016/j.biopha.2021.111528

    26. Kato M, Wang L, Putta S, et al. Post-transcriptional up-regulation of Tsc-22 by Ybx1, a target of miR-216a, mediates TGF-beta-induced collagen expression in kidney cells. J Biol Chem. 2010;285(44):34004–34015. doi:10.1074/jbc.M110.165027

    27. Liu L, Chen Y, Li Q, Duan P. lncRNA HNF1A-AS1 modulates non-small cell lung cancer progression by targeting miR-149-5p/Cdk6. Journal of Cellular Biochemistry. 2019;120(11):18736–18750. doi:10.1002/jcb.29186

    28. Liu HT, Ma RR, Lv BB, et al. LncRNA-HNF1A-AS1 functions as a competing endogenous RNA to activate PI3K/AKT signalling pathway by sponging miR-30b-3p in gastric cancer. British Journal of Cancer. 2020;122(12):1825–1836. doi:10.1038/s41416-020-0836-4

    29. Guo X, Zhang Y, Liu L, Yang W, Zhang Q. HNF1A-AS1 regulates cell migration, invasion and glycolysis via modulating miR-124/MYO6 in colorectal cancer cells. OncoTargets and Therapy. 2020;13:1507–1518. doi:10.2147/OTT.S231249

    30. Wu J, Li R, Li L, et al. MYC-activated lncRNA HNF1A-AS1 overexpression facilitates glioma progression via cooperating with miR-32-5p/SOX4 axis. Cancer Medicine. 2020;9(17):6387–6398. doi:10.1002/cam4.3186

    31. Ma C, Wang H, Zong G, et al. EGR1 modulated LncRNA HNF1A-AS1 drives glioblastoma progression via miR-22-3p/ENO1 axis. Cell Death Discovery. 2021;7(1):350. doi:10.1038/s41420-021-00734-3

    32. Bi Y, Mao Y, Su Z, Du J, Ye L, Xu F. Long noncoding RNA HNF1A-AS1 regulates proliferation and apoptosis of glioma through activation of the JNK signaling pathway via miR-363-3p/MAP2K4. Journal of Cellular Physiology. 2021;236(2):1068–1082. doi:10.1002/jcp.29916

    33. Cesana M, Cacchiarelli D, Legnini I, et al. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 2011;147(2):358–369. doi:10.1016/j.cell.2011.09.028

    34. Ndlovu MN, Van Lint C, Van Wesemael K, et al. Hyperactivated NF-kappaB and AP-1 transcription factors promote highly accessible chromatin and constitutive transcription across the interleukin-6 gene promoter in metastatic breast cancer cells. Mol Cell Biol. 2009;29(20):5488–5504. doi:10.1128/MCB.01657-08

    35. Ding H, Wang F, Shi X, et al. LncRNA MALAT1 induces the dysfunction of β cells via reducing the histone acetylation of the PDX-1 promoter in type 1 diabetes. Experimental and Molecular Pathology. 2020;114:104432. doi:10.1016/j.yexmp.2020.104432

    36. Xu X, Tian J, Li QY. Downregulation of HOTTIP regulates insulin secretion and cell cycle in islet β cells via inhibiting MEK/ERK pathway. European Review for Medical and Pharmacological Sciences. 2018;22(15):4962–4968. doi:10.26355/eurrev_201808_15636

    37. Streicher SA, Lim U, Park SL, et al. Genome-wide association study of pancreatic fat: the multiethnic cohort adiposity phenotype study. PLoS One. 2021;16(7):e0249615. doi:10.1371/journal.pone.0249615

    38. Akerman I, Tu Z, Beucher A, et al. Human pancreatic β cell lncRNAs control cell-specific regulatory networks. Cell Metab. 2017;25(2):400–411. doi:10.1016/j.cmet.2016.11.016

    Continue Reading

  • Telluride 2025: The 6 best films we saw at the film festival

    Telluride 2025: The 6 best films we saw at the film festival

    p]:text-cms-story-body-color-text clearfix”>

    Jesse Plemons in the movie “Bugonia.”

    (Atsushi Nishijima / Focus Features)

    Jesse Plemons is never one to chew scenery. Even when handed a role that edges on madness, he doesn’t go big. Instead, he goes deep, building tension quietly from the inside out. And in Yorgos Lanthimos’ uncategorizable, darkly comic sci-fi thriller, Plemons — reuniting with the director after playing three characters in last year’s “Kinds of Kindness” — delivers one of his most riveting performances yet. As Teddy, a rumpled, reclusive beekeeper convinced that a pharma CEO (Emma Stone) is an alien from the planet Andromeda, Plemons channels paranoia, grief and righteousness into something both absurd and unnervingly sincere. The “I do my own research” archetype could easily veer into “SNL” sketch territory but he plays it heartbreakingly straight, creating a chillingly familiar portrait of a man lost in an algorithmic maze of internet rabbit holes and desperate for clarity in a world that no longer makes sense. Teddy enlists his younger cousin Don (Aidan Delbis, an autistic first-time actor in a mesmerizing turn) to help him abduct Stone’s steely executive, drawing him into the mission in a misguided effort to protect him. Even as things spiral into chaos, Plemons (a 2022 supporting actor Oscar nominee for Jane Campion’s “The Power of the Dog”) roots the performance in a warped but recognizably human emotional logic. The result captures the anxious, conspiratorial spirit of 2025 with eerie precision, proving once again that Plemons doesn’t need to raise his voice to deliver a performance that speaks volumes. — Josh Rottenberg

    Continue Reading

  • Marshall adds a subwoofer and compact soundbar to its Heston TV audio lineup

    Marshall adds a subwoofer and compact soundbar to its Heston TV audio lineup

    Marshall’s family of TV audio gear is growing. The company has announced the Heston Sub 200 subwoofer and Heston 60 compact soundbar, both of which are now available for pre-order. The $700 Heston 60 is a smaller take on the first soundbar Marshall introduced earlier this year, the Heston 120, offering Dolby Atmos and DTS-X for more intimate spaces. With the Heston Sub 200, a $600 subwoofer that pairs with both soundbars, Marshall touts “a sound that can be truly felt.” Both products come in Cream and Black, and sport Marshall’s classic guitar amp styling. They ship September 23.

    The Heston 60 has 7 Class D amplifiers — two 25W and five 5W — with a total power output of 56W. It has two woofers and five full range drivers. The soundbar offers wireless and wired connectivity, with Bluetooth (5.3) and Wi-Fi, as well as an HDMI 2.1 port (eARC), a 3.5mm aux port, an RCA input and USB-C. It also supports AirPlay 2, Google Cast, Spotify Connect and Tidal Connect.

    The Heston 200, Heston 120 and Heston 60

    (Marshall)

    The Heston Sub 200 packs two 5.25 inch subwoofers and two 120W Class D amplifiers, with a peak total power output of 236W. Marshall says it’ll deliver deep, room-shaking bass. The subwoofer has Bluetooth and wired input (RCA mono).

    The Heston 60 and Heston Sub 200 will be available from Marshall starting September 23, and will roll out to select other retailers on September 30.

    Continue Reading

  • Jude Law on being in the driver’s seat for Netflix’s ‘Black Rabbit’

    Jude Law on being in the driver’s seat for Netflix’s ‘Black Rabbit’

    Jude Law and his producing partner Ben Jackson have been on plenty of film sets, absorbing lessons from masters including Anthony Minghella (“Cold Mountain”) and Steven Soderbergh (“Side Effects”).

    But for Law, Jackson and their production company, Riff Raff Entertainment, their new Netflix limited series “Black Rabbit,” debuting Sept. 18, marks a big step: It’s their first episodic TV effort produced from inception.

    Created by Zach Baylin and Kate Susman, the eight-episode series stars Law as the ambitious owner of a New York hot spot whose life is thrown into turmoil when his drug- and gambling-addicted brother, played by Jason Bateman, comes back into the business. It’s a sort of grim and gritty Cain-and-Abel story for the restaurant scene, complete with debt-collecting goons, severed digits and armed robbery.

    “It was a slice of New York life that we felt we both knew,” Law said by video from London. “What I loved was the fraternal heartbeat in its center and the question it raised: How far does one go for someone you love?”

    Founded in 2017, Riff Raff locked in millions of dollars in financing from venture firm Calculus Capital about three years ago to expand its development slate and hire additional executives.

    Law isn’t going to lie. It can be easier for him to sell a movie or TV series he has in development when he’s planning to star in it.

    But simply making vehicles for the “Talented Mr. Ripley” actor isn’t all he and Jackson have in mind for Riff Raff. Of the dozens of projects the firm has in development, only a handful have the actor attached. Upcoming efforts include a Sharon Horgan-scripted romantic drama film, a historical drama from Justin Kuritzkes and a major TV drama by writer Jonathan Glatzer.

    The five-person Riff Raff is based primarily in London but has a growing presence in Los Angeles, where Jackson spends half his time. Previous credits include the 2024 thriller “The Order” and the 2021 drama “True Things.”

    This interview was edited for clarity and length.

    Why did you want to make this show, “Black Rabbit”?”

    Jude Law: “Black Rabbit” is a very good example of what we hoped to build and foster in the company. It came out of relationships that we had forged. So in this example, Zach Baylin, whom we’ve worked with on “The Order,” had mentioned this idea for a show he had with his partner, Kate Susman, and passed it on to Ben. It felt familiar, and yet it felt like there was fresh territory being covered.

    Ben Jackson: It also felt that there could be something incredibly cinematic about this piece as well, which is something that as a company we really want to bring to any TV we do.

    Law: We were very much along for the ride on “The Young Pope” and “The New Pope,” learning as young producers. But with this, being front and center and really being able to steer it and relish what episodic TV offers you. … If you trust the map you have, and you trust the team around you, that’s a very exciting creative situation to be in. And that was a really wonderful experience for Ben and me to flex a new muscle as producers in the driver’s seat.

    What learnings did you pick up from “The Young Pope” and “The New Pope?”

    Law: I mentioned those two because we were fortunate enough to have a producer credit. But I would honestly credit every set and every director that Ben and I have been on a set with for the last 20-odd years together. You learn pretty quickly what you think works and what you think doesn’t work. One of the reasons Ben and I ended up creating the company was the inspiration we drew from our experiences of being on a set with Anthony Minghella or Steven Soderbergh.

    Jackson: There’s no better way of learning anything than doing it. It’s osmosis. Everybody makes mistakes, but you see how things work. The late, great Anthony Minghella — whose set I first worked on with “Cold Mountain” — had this overriding sense of family. Everybody was included. Everybody had a role to play.

    Having Troy Kotsur as a menacing bad guy in the show was pretty interesting, given that I think a lot of people know him as the dad from “CODA.”

    Law: I wish one of us could take responsibility for that brilliant choice. It actually came while Zach was on the Academy campaign trail for “King Richard.” His path crossed with Troy, who was similarly attending those events for “CODA.” I think Troy suggested it. They talked about potential roles and Troy said, “I would love to play a really bad guy.” And that germinated in Zach’s imagination, and he wrote a wonderful role for Troy. But Troy turned up and absolutely delivered. There’s an incredible intensity and stillness, and I think we all learned a huge amount from working with him.

    What’s different about Riff Raff and what you guys are trying to do here with this company?

    Law: Like most other companies, we’re trying to make great work. We’re trying to empower people we believe in and are inspired by to make great work. This has always been, for me personally, an opportunity to stretch my input into filmmaking beyond what I do in front of the camera. At the same time, I really appreciate and love the potential of having a company where it’s allowed me as an actor to hold the reins a little firmer and introduce roles that I think the marketplace might not associate me with. But so many of these projects have their own legs, and our aim is to build a brand that people go to because they enjoy our output, not because it’s attached to me.

    Jackson: I think there’s a danger when you have a talent-led company, where people say, oh, well, if it’s his company, we know what we’re going to get. And I think our goal is to make sure that it’s eclectic.

    With “Black Rabbit,” were you always attached to play Jake, the brother who owns the restaurant?

    Law: I was absolutely attached, but …my focus was in the belief of the whole piece. And you know what it was, even when we went to Jason and we were interested in asking him to come on board and for him to direct, I think I had the idea: “Well, shouldn’t Jason be the brother?” But I wasn’t sure at the time whether I was Vince or Jake. And so, yeah, that conversation was had quite late in the day. You know, there was plenty of time for us to learn our lines and swap costumes.

    Jason’s a good choice because he’s done the dirtbag kind of guy before. Even in “Arrested Development,” he’s got a bit of a mean streak.

    Law: But he does it with an incredible — uh, wink, yeah — humor and humanity, which is a very hard nuance to pull off.

    How much has the company grown over the years?

    Law: Ben really changed it all in that it was sort of being financed by myself. We were doing it over my kitchen table on the hoof. And I think he realized we had to make the leap and really decide whether we were going to make a go of it or not. And it was a vital decision to make, and we were fortunate enough to find funding, and fortunate enough, therefore, to be able to broaden the team. And then obviously, first-look deals give us some firepower to develop and produce, and the team has just empowered us to embark on sourcing the slate.

    Jackson: We’re also very aware of what our limits are. We don’t want to be a company that’s going around going, “Oh, that’s an issue, but we’ll option that,” and then it sits there, going nowhere. It’s a lot of work put into developing anything, and so you’ve all got to be on board. Otherwise, things drift. Momentum is key in this industry. You need to keep things moving.

    What’s the market like right now from your perspective in terms of getting things greenlit?

    Law: I think the problem is nobody knows. You have to have conviction in what you’re trying to say. If you have a writer with a great track record, or a director with a great track record, there’s a certain amount of, “Look, I’ve done it before, I can do it again.” But even then everyone is second-guessing right up to the end. It’s just a case of nudging it forward without losing sight of the reason you all got on board in the first place and not budgeting yourself into a position where you’re putting too much pressure on yourselves.

    I go back to relationships, building relationships with the people you can trust, as distributors, who you know will have your back, or a company like Netflix, which has had our back on this from day one.

    Jackson: You have to be adaptable. I think there’s no finite formula for making a successful film or show. Does it fit the world we live in right now? No one knows. I think what you have to be is you have to be committed to what you’re doing.

    What’s the right level of activity for the company?

    Law: Well, that’s hard to say. But we have a slate of 30-plus projects.

    Having now really got a good foothold with the team that we’ve been allowed to assemble in the U.K., we’re very keen to purchase a more significant foothold in the U.S. Ben spends half his time there, and we’re hoping to just build. I think recognizing the bridge between the U.K. and the U.S. markets is very important.

    Well, studios are making so many of their productions in the U.K. these days.

    Law: So much so that it’s almost impossible to get crew and studio space, so it’s possible that we may end up in Budapest, Prague or Berlin.

    What other projects are you excited about?

    Law: Something that we’ve been developing with Justin Kuritzkes (“Challengers”), based loosely on the documentary “Best of Enemies,” is a very exciting script that we’re putting together. It was interesting, the new book on William F. Buckley Jr. just came out, and all the reviews are talking about his influence on the current Republican Party, which is something we spotted a while back.

    Is there any sort of through line for what you find yourselves drawn to?

    Law: It’s a really good question. I hope that I have demonstrated through the choices I’ve made as an actor that I find most genres and most forms of storytelling interesting. I’ve really relished being in big-budget, heavy IP [intellectual property] pieces and working on tiny independents. I hope the company will reflect that. I hope our signature is quality.

    Stuff we wrote

    Number of the week

    $3.67 billion

    The summer box office totaled an unimpressive $3.67 billion in ticket sales from May through Labor Day, down slightly from last year’s similarly soft tally and way below prepandemic levels, when grosses typically exceeded $4 billion.

    That’s bad. Blame a lack of true blockbusters, streaming, shortened theatrical windows and an over-reliance on aging franchises. Read Samantha Masunaga’s story for more on the winners and losers.

    Finally …

    Listen: Cass McCombs, “Interior Live Oak”; The Beths, “Straight Line Was a Lie.”

    Continue Reading

  • Cannabis May Help You Sleep Better, Researchers Say

    Cannabis May Help You Sleep Better, Researchers Say

    Share on Pinterest
    A new study found that daily cannabis use improved sleep in people with insomnia. Getty Images
    • Cannabis products consumed on a daily basis resulted in better sleep for people with insomnia in a new study.
    • Participants also reported improvements in anxiety and depression as well as pain levels.
    • Cannabis continued to reduce insomnia throughout the 18-month trial, although the effects waned over time.
    • Experts caution that tolerance for cannabis may reduce its value as a long-term sleep aid, and note other health risks associated with chronic cannabis use.

    People with insomnia experienced improved sleep with regular cannabis use in a new study.

    The authors report that participants experienced less insomnia using cannabis products daily throughout the 18-month study period. The findings were published on August 27 in PLOS Mental Health.

    Estimates of the number of people who have difficulty sleeping range from 10%, as cited by the study authors, to as high as 30%.

    Effective medical treatments for insomnia are available, but they often come with side effects and a risk of developing a dependence.

    In the new study, researchers recruited people who had been diagnosed with primary insomnia and who were registered with the UK Medical Cannabis Registry. The final study cohort was 124 participants, 18 years old or older.

    Individuals were each prescribed the same controlled daily dosage of cannabis supplied by the authors of the study.

    Cannabis was consumed in oil or dried flower form, or a combination of both. No external cannabis was allowed during the trial period.

    A total of 11 participants experienced 112 adverse events, including fatigue, dry mouth, and insomnia, though none of these were considered disabling or life threatening.

    At the end of the 18-month trial, participants said that cannabis was still helping them sleep. However, the strongest benefit was observed during the first month of the trial and slowly decreased through the study period.

    In addition to reporting insomnia relief, individuals said they also experienced less pain and that their levels of anxiety and depression improved throughout the trial.

    “Many people with anxiety and depression have trouble falling asleep because of racing thoughts, so that’s where cannabis helps in the short term.”

    Previous research studying the effects of cannabis on sleep quality has not consistently confirmed its value.

    The study followed participants for a year and a half, however, it’s unclear whether cannabis is a viable long-term sleep aid.

    The authors note that one potential issue is how the body develops a tolerance for cannabis, meaning that consuming more and more of it may be necessary to achieve the same result.

    They suggest that the decrease in the effectiveness of cannabis over time is likely related to participants’ growing tolerance.

    “There is evidence indicating the consistent medical use of cannabis is associated with higher usage over time, resulting in increased risk of tolerance and dependence,” Patel said.

    Bonaguro noted that cannabis use often becomes a cycle. “It gives short-term relief of anxiety or depression, but with prolonged use, tolerance builds, dependency develops, and both mood and sleep get worse,” she said.

    The study investigated daily consumption of cannabis products for insomnia, and Patel noted there are side effects associated with frequent use.

    However, as a 2021 review notes, cannabis may compare favorably “in regards to safety and tolerability with other drugs, such as opioids, and it is important to weigh up the benefits of use vs the risk of dependence.”

    The review further notes that the risk of adverse side effects, such as tolerance or dependence, may not outweigh the medicinal benefits of using cannabis for the management of chronic health conditions, “highlighting the need for a medicine with a good safety profile.”

    As for sleep, however, cannabis use may be disruptive for some. Some research suggests that cannabis may affect slow-wave (N3) sleep. There is also suspicion that it fractures REM and deep sleep.

    “One more thing I frequently heard from clients who use it regularly or sporadically is that they can’t remember their dreams,” said Bonaguro. “This makes sense since THC is well-documented to suppress REM sleep, which is when most dreaming occurs.”

    Patel added that chronic cannabis use has also been linked to psychiatric problems in susceptible individuals.

    The authors of the study conclude their report with an expression of their own concern.

    They note the likelihood that insomnia patients will develop a tolerance for cannabis may ultimately limit the substance’s usefulness as a long-term therapy, however successful it proved throughout the relatively short-term of the study period.

    Those successes alone, they argue, merit further randomized controlled trials of cannabis use in treating insomnia.

    Continue Reading

  • Clinical Audit of Acute Oxygen Therapy: Enhancing Patient Care at Atbara Teaching Hospital

    Clinical Audit of Acute Oxygen Therapy: Enhancing Patient Care at Atbara Teaching Hospital


    Continue Reading

  • Beta Blockers After Milder Heart Attacks Unhelpful, Possibly Harmful – Inside Precision Medicine

    1. Beta Blockers After Milder Heart Attacks Unhelpful, Possibly Harmful  Inside Precision Medicine
    2. Common heart attack drug doesn’t work and may raise risk of death for some women, new studies say  CNN
    3. Study finds beta blockers may raise risk of death in women | Tap to know more | Inshorts  Inshorts
    4. Effectiveness of Beta-Blockers in Reducing Mortality and Recurrence After Myocardial Infarction: A Systematic Review of Contemporary and Foundational Evidence  The Cureus Journal of Medical Science
    5. Research on beta blockers shows conflicting results  Semafor

    Continue Reading

  • ‘It’s a big result’ – Esteban Ocon and Ollie Bearman “very satisfied” with double-points finish for Haas at Zandvoort

    ‘It’s a big result’ – Esteban Ocon and Ollie Bearman “very satisfied” with double-points finish for Haas at Zandvoort

    Ollie Bearman managed to recover from a pit lane start to finish an incredible sixth in the Dutch Grand Prix, giving Haas much to celebrate as they brought home a double-points finish, with Esteban Ocon crossing the line in 10th.

    The result was extra impressive when considering their starting positions, but the drivers were keen to stress it was one born out of hard work, with excellent strategy calls contributing to their haul of points in Zandvoort.

    Bearman’s drive was arguably the more surprising, given the youngster opted for a pit lane start to change some power unit components after qualifying just 19th on Saturday. He started on the hard tyre and ran a very long first stint, Haas opting not to pit under the first Safety Car unlike the majority of the field.

    The team needed another Safety Car to make their strategy work, and they got just that when Charles Leclerc and Kimi Antonelli crashed, Bearman diving into the pits for his one and only stop. That left him solidly in the top 10, and not only did he hold station, but the rookie kept close enough to Antonelli to profit when the latter’s time penalties were applied.

    “We got lucky today for sure, but sometimes you need to be in the right place at the right time,” Bearman explained after the race.

    “Good calls on strategy, the car was quick as well which always has to be the case as well. The guys did a good job, gave me a great car. We were a bit unlucky I would say after Qualifying, we had a few misfortunes which meant that we started from last, but to score points today is a very good feeling.”

    Bearman had to work hard for his score, sticking close to his team mate in the first stint as both started to struggle on aging tyres, and then keeping some faster, fresher shod cars at bay. It was his first points score since Bahrain, and his best finish of the season so far.

    “Nice to get back in the points. It has been a long time coming for sure, the team have been working incredibly hard, and I think we are getting there step by step.

    “We’ve got some things to work on and also myself, I’m far from perfect, still making too many mistakes. But we are on the right path and I really have high hopes,” he concluded.

    Team work was key in Zandvoort, with Ocon – who started 18th and was the first Haas driver to breach the top 10 – helping Bearman at points in the race. The Frenchman could have felt aggrieved that his team mate finished ahead, given Ocon was running in front for much of the race – but he stopped one lap earlier than Bearman, and that cost him a handful of places.

    “It’s very positive as a team to have a double-points finish for both of us,” Ocon said. “Got a little bit unlucky with the Safety Car, I think I lost maybe three positions or four. But overall, very satisfied with that. Well done to Ollie, he has done a fantastic race also and that’s big for us, it is a big result.

    “We helped ourselves during the race. We were 10th and 11th during the time and Ollie dropped back from DRS and the team asked me to give him DRS [drag reduction system] and we just kept pushing forward, both of us together.

    “And then I boxed and the Safety Car came at the wrong time so I fell backwards, but Ollie benefited from that. So as a team it was good, definitely strong points. I knew there was something we could play here, even starting from the back, it makes crazy races for sure. But happy with the outcome for the team and hopefully we can do more of that at the next one.”

    Haas remain ninth in the Teams’ Championship, but at least have kept in touch with Kick Sauber ahead, and only need another strong weekend to close the gap in what is increasingly turning into an incredibly tight midfield fight.

    Continue Reading