Category: 3. Business

  • Outrage in Paris as Shein prepares to open its first permanent store | Shein

    Outrage in Paris as Shein prepares to open its first permanent store | Shein

    The online fast-fashion retailer Shein will open its first permanent, bricks-and-mortar store in the world in Paris this week, amid political outrage, fury from workers and warnings from city hall that it will damage the French capital’s progressive image.

    The Singapore-based clothing company, which was founded in China, has built a massive online business despite criticism over its factory working conditions and the environmental impact of low-cost, throwaway fashion.

    Shein, which has previously trialled temporary pop-up stores, will on Wednesday open a permanent shop on the sixth floor of Paris’s prestigious BHV department store, a historic building that has stood opposite Paris’s city hall since 1856. There are about 23 million Shein customers in France, one of its biggest European markets.

    But with vast banners for Shein draped across the building, the brand’s arrival has sparked outrage over the promotion of fast fashion.

    The office of the French minister for small businesses said Shein’s Paris presence sent “a bad signal that should be avoided”. Several leading independent French fashion brands have pulled their products from the BHV store in protest.

    “There would be no sense being sold in the same shop as Shein,” Guillaume Alcan, a co-founder of the French ethical footwear brand Odaje, told Le Monde. Disneyland Paris abandoned plans to open a Christmas pop-up store in BHV and pulled out of creating themed window displays for the end-of-year holidays, saying “conditions were no longer in place” to “calmly hold Christmas events” at the location.

    Shein branding on the outside of the department store building. Photograph: Abdul Saboor/Reuters

    After Shein’s arrival was announced, a French state-owned bank pulled out of talks with the operator of the department store to buy the building. Paris city hall blocked plans for a Paris rugby stadium to carry the BHV logo.

    BHV staff have staged strikes and street protests in recent weeks.

    Nicolas Bonnet-Oulaldj, Paris’s Communist deputy mayor in charge of commerce, said of Shein’s arrival: “We are totally against this. It is the complete opposite of Paris’s policy to develop independent shops and support products that are made in France.”

    Ian Brossat, a Communist party senator in Paris, said: “Shein coming to BHV is a real provocation, particularly since the national assembly and senate recently approved a law to restrict ‘ultra fast-fashion’.”

    Shein, which has defended its labour and environmental policies, has said its presence in France will attract younger shoppers and boost other high street businesses. It will also open permanent shops in the French cities of Dijon, Reims, Grenoble, Angers and Limoges inside Galeries Lafayette department stores, which are operated by the same group that manages Paris’s BHV.

    skip past newsletter promotion

    The row intensified on Monday after the French finance minister, Roland Lescure, threatened to ban Shein in France if it resumed selling “childlike” sex dolls. France’s anti-fraud unit reported the presence of the dolls on Shein’s e-commerce site this weekend.

    “These horrible items are illegal,” Lescure told the BFM TV channel, promising a judicial investigation. Shein told Reuters: “The products in question were immediately removed from the platform as soon as we became aware of these major shortcomings.”

    France has already fined Shein three times in 2025 for a total of €191m (£167m). The biggest fine, of €150m, was imposed for failing to comply with online cookie legislation. The company is contesting this. Other fines were issued for false advertising, misleading information and not declaring the presence of plastic microfibres in its products.

    The European Commission is investigating Shein over risks linked to illegal products. Shein said at the start of the investigation earlier this year that it welcomed “efforts that enhance trust and safety for European consumers when shopping online”. In May, the company said it had “intensified its product safety and quality controls”.

    Continue Reading

  • UK Regulator Targets Delays in Rare Disease Treatments – Medscape

    1. UK Regulator Targets Delays in Rare Disease Treatments  Medscape
    2. Major change for rare disease treatments on way, signals MHRA  GOV.UK
    3. MHRA set to overhaul the UK’s rare disease drug regulatory pathway  European Pharmaceutical Review
    4. New drugs to be approved faster so patients don’t wait years  The Times
    5. MHRA reforms will speed up path for rare therapies  Healthcare Management Magazine

    Continue Reading

  • Economic historian to discuss underestimated gains from transportation investments: For Journalists

    Economic historian to discuss underestimated gains from transportation investments: For Journalists

    EVANSTON, Ill. — Richard Hornbeck, an economic historian at the University of Chicago Booth School of Business, will discuss how flawed economic models underestimate the true impact of transportation investments during the 2025 Leon N. Moses Distinguished Lecture in Transportation at Northwestern University this week.

    Hosted by the Northwestern University Transportation Center (NUTC), the lecture, “Amplified gains from transportation infrastructure investments,” will take place at 7:30 p.m. on Wednesday, Nov. 5 at 700 University Place in Evanston. The free lecture is open to the public but registration is encouraged.

    Hornbeck is the V. Duane Rath Professor of Economics at the University of Chicago, where he studies the historical development of the U.S. economy. In his lecture, he will argue for a new framework that accounts for real-world imperfections often overlooked in traditional economic models of infrastructure investment. These models typically assume the economy functions efficiently. But, in reality, markets can be uncompetitive, and firms often struggle to access financing.

    Hornbeck will illustrate this flaw using the example of U.S. railroad expansion in the late 1800s. Ignoring broader economic distortions led to an understatement of the railroads’ contribution to economic growth. By reworking these frameworks, economists can more accurately estimate the benefits from future transportation investments.

    In addition to his position at the University of Chicago, Hornbeck is a research associate at the National Bureau of Economic Research, affiliated with programs on the development of the American economy, development economics, and environmental and energy economics. Prior to joining Chicago Booth in 2015, Hornbeck was the Dunwalke Associate Professor of American History in the economics department at Harvard University. He received an Alfred P. Sloan Fellowship in 2014 and was selected for the 2009 Review of Economic Studies Tour.

    The Leon N. Moses Distinguished Lecture in Transportation was named in honor of the late Professor Leon N. Moses for his significant contributions to the field of transportation economics and regional science and for his long and dedicated service to the NUTC.

    The NUTC is one of the world’s leading interdisciplinary education and research institutions, serving industry, government and the public. Founded in 1954 to make substantive and enduring contributions to the movement of materials, people, energy and information, the center stands at the forefront of transportation research and education, bringing together academic researchers, students and business affiliates in open exploration of transportation and supply chain operations.

    Continue Reading

  • Glofitamab/Polatuzumab Vedotin Exhibits Response Durability in R/R LBCL

    Glofitamab/Polatuzumab Vedotin Exhibits Response Durability in R/R LBCL

    The addition of polatuzumab vedotin-piiq (Polivy) to glofitamab-gxbm (Columvi) displayed durable responses and efficacy among patients with relapsed/refractory large B-cell lymphoma (LBCL) and patients with high-grade BCL (HGBCL), according to findings from a phase 1b/2 study (NCT03533283) published in the Journal of Clinical Oncology.1

    Efficacy data from the trial revealed that after a median follow-up of 32.7 months (range, 0-55) for the total population (n = 129), the objective response rate (ORR) as assessed by an independent review committee (IRC) was 78.3%, with 59.7% attaining a complete response (CR). Additionally, similar results were found with the investigator-assessed ORR and CR rate, at 84.2% and 61.4%, respectively. Furthermore, after a median follow-up of 19.2 months (range, 0-43) among patients with HGBCL (n = 44), the ORR and CR rate was 79.5% and 65.9%.

    Across all patients, the median duration of response (DOR) was 26.4 months (95% CI, 10.9-44.3), with a 24-month rate of 51.9%. For the 77 patients who attained a CR, the median duration of CR (DOCR) was 37.8 months (95% CI, 24.1-not estimable [NE]) after a median follow up of 25.5 months (range, 0-48), with a 24-month rate of 63.9%. Among patients with HGBCL, the median DOCR was not reached.

    Additionally, the median progression-free survival (PFS) per IRC assessment was 12.3 months (95% CI, 8.8-27.7) for the overall population and 16.3 months (95% CI, 9.1-NE) for those with HGBCL. The 24-month PFS rate in the overall population was 41.8%. Additionally, the median overall survival (OS) and event-free survival (EFS) in the overall population was 33.8 months (95% CI, 20.6-NE) and 11.5 months (95% CI, 8.5-17.7), respectively; the OS data were immature at the clinical cutoff date, with a 24-month OS rate of 54.3%.

    “[Glofitamab/polatuzumab vedotin] demonstrated high efficacy, with frequent and durable responses in patients with heavily pretreated [relapsed/refractory] LBCL, spanning various histologies and including individuals who received previous CAR T-cell therapy,” lead study author Martin Hutchings, MD, PhD, staff specialist in the Department of Hematology at the Finsen Centre, National Hospital, Copenhagen University Hospital in Denmark, wrote in the publication with study coinvestigators. “A particularly important finding was the high activity in patients with HGBCL. The safety profile remained manageable. Long-term follow-up data on [glofitamab/polatuzumab vedotin] in patients with [relapsed/refractory] LBCL, including HGBCL, will offer valuable insights into its promising potential as a therapeutic option for patients with [relapsed/refractory] LBCL.”

    The open-label phase 1b/2 trial enrolled patients with relapsed/refractory LBCL, including diffuse large B-cell lymphoma (DLBCL), HGBCL, transformed follicular lymphoma, and primary mediastinal LBCL. The study was expanded to include 44 patients with HGBCL based on a high unmet need in this subgroup.

    Patients received pretreatment with 1000 mg of obinutuzumab (Gazyva) on cycle 1, day 1 at 7 days prior to the first glofitamab dose to help mitigate the frequency of cytokine release syndrome (CRS). Polatuzumab vedotin was given intravenously at 1.8 mg/kg on cycle 1, day 2 and day 1 of cycles 2 to 6 once daily for each 21-day cycle. Intravenous glofitamab was given as step-up dosing during cycle 1, at 2.5 mg on day 8 and 10 mg on day 15; the target dose of 30 mg was given on day 1 of cycles 2 to 12 for each 21-day cycle.

    Following the first glofitamab dose, patients were hospitalized for 24 hours. A fixed duration of 6 cycles for polatuzumab vedotin and 12 cycles for glofitamab was given in the absence of disease progression, unacceptable toxicities, or withdrawal of consent. Patients with responses or stable disease were followed until disease progression, and those with progressive disease were followed for survival after an end-of-study visit.

    Among the overall and HGBCL populations, the median age was 67.0 years (range, 23-84) and 66.5 years (range, 24-84). Most patients were male (63.6% vs 63.6%), had an ECOG performance status of 0 or 1 (94.6% vs 93.2%), and had Ann Arbor stage III/IV disease (76.7% vs 72.7%). Patients most often had an Internal Prognostic Index (IPI) score of 2 (30.2% vs 29.5%), 3 (24.0% vs 20.5%), and 4 (23.3% vs 25.0%).

    A total of 72.9% vs 70.5% of the overall and HGBCL groups had extranodal involvement at study start, and 29.5% vs 22.7% had bulky disease. Most patients were negative for double- or triple-hit lymphoma (45.7% vs 27.3%), had received 2 or more lines of treatment (58.9% vs 45.5%), and were refractory to the first line of previous therapy (62.0% vs 70.5%) or any previous therapy (79.1% vs 81.8%).

    The primary end point of the trial was IRC-assessed ORR per PET-CT scan per Lugano 2014 criteria. Secondary end points included investigator-assessed ORR, DOR, DOCR, PFS, EFS, OS, and safety.

    Regarding safety, 99.2% of patients experienced any-grade adverse effects (AEs), the most common of which were CRS (43.4%), neutropenia (41.9%), peripheral neuropathy (26.4%), diarrhea (24.0%), COVID-19 infection (23.3%), and pyrexia (20.2%). Additionally, 58.9% of patients experienced grade 3 or 4 AEs; the most common included neutropenia (32.6%), COVID-19 infection or pneumonia (9.4%), anemia (8.5%), thrombocytopenia (8.5%), and tumor flare (7.0%). Grade 5 AEs occurred in 9.3% of patients, and serious AEs occurred in 61.2%.

    AEs led to discontinuation in 14.7% of patients, including 12.4% discontinuing glofitamab and 8.5% discontinuing polatuzumab vedotin. Only 1 instance of grade 3 and grade 5 CRS was observed, with serious CRS occurring in 29.4% of patients.

    The time to CRS onset after the 30 mg once daily dose was 36.2 hours (range, 18.5-55.9). The most common CRS management strategies included tocilizumab (Actemra; 33.9%), intravenous fluids (23.2%), low-flow oxygen (19.6%), and corticosteroids (14.3%).

    Reference

    Hutchings M, Sureda A, Bosch F, et al. Efficacy and safety of glofitamab plus polatuzumab vedotin in relapsed/refractory large B-cell lymphoma including high-grade B-cell lymphoma: results from a phase Ib/II trial. J Clin Oncol. Published October 20, 2025. doi:10.1200/JCO-25-00992

    Continue Reading

  • Telegraph bidder reported for potential breach of editorial independence rules | Telegraph Media Group

    Telegraph bidder reported for potential breach of editorial independence rules | Telegraph Media Group

    The boss of the US private equity group bidding for the Daily Telegraph has been reported to the UK government for potentially breaching rules protecting the newspaper’s editorial independence, after allegedly threatening to “go to war” with the title’s newsroom.

    The Guardian understands that the independent directors of Telegraph Media Group (TMG) have alerted the Department for Culture, Media and Sport (DCMS) about supposed comments made by RedBird Capital’s Gerry Cardinale to the Telegraph’s editor, Chris Evans. The government department is thought to be considering if there has been a breach of the legislation.

    The title’s former editor Charles Moore disclosed in his Telegraph column last month that inquiries by the paper’s journalists into RedBird Capital’s bid had prompted Cardinale to “threaten he would go to war with our entire newsroom”.

    Moore added there had been “apparent media briefings” that Evans would be removed and replaced as editor – although that was followed up by a column the Telegraph published by Cardinale last week.

    In it, the private equity boss said: “We won’t ever compromise the editorial independence of the Telegraph. At RedBird, we are very clear about one fundamental premise: don’t invest in a newspaper if you want to influence it – that will kill the investment thesis and is just bad business.”

    Last year the government introduced a statutory instrument compelling parties to “take all reasonable steps to retain key staff within the Telegraph Media Group business and to ensure that no key staff are removed from their position”.

    The order added: “At all times, the … acquiring entities must keep the secretary of state informed of any material developments relating to the Telegraph Media Group business, which includes details of key staff who leave or join the Telegraph Media Group business.”

    The referral to the culture department follows yet another eventful few weeks in the saga of the acquisition of the Telegraph, which has come under scrutiny as the initial acquisition was funded by foreign state interests. Last week the newspaper linked its presumed new owner to the suspected ringleader of the alleged Chinese spy ring in Westminster.

    The Telegraph’s future has been uncertain since the Barclay family lost its grip on the media group in 2023 in a row about unpaid debts. An organisation connected to Redbird Capital, called Redbird IMI, took control of the newspaper titles later that year.

    However, RedBird IMI was forced to put the papers up for sale in spring 2024 after the then Conservative government passed a law blocking foreign states or associated individuals from owning newspaper assets in the UK. Redbird IMI is in the process of selling TMG to RedBird Capital – which holds various investments, including a stake in the parent company of Liverpool football club.

    While a quarter of RedBird IMI’s funding came from RedBird Capital, the remainder was sourced from International Media Investments (IMI), which is controlled by Abu Dhabi’s Sheikh Mansour bin Zayed al-Nahyan, the vice-president of the United Arab Emirates and owner of Manchester City FC.

    skip past newsletter promotion

    The Labour government eased the ban on foreign governments owning stakes in UK newspapers this year by allowing them to hold up to 15% of titles, ultimately allowing the RedBird Capital offer that would include IMI retaining a 15% Telegraph stake.

    The DCMS and a spokesperson for TMG’s directors declined to comment on the report to DCMS by the Telegraph’s directors.

    A spokesperson for RedBird Capital said: “RedBird is a private equity fund, not a proprietor, and the mandate of the fund is to grow the value of its investments. The way to grow the value of the Telegraph is to grow subscribers and the way to do that is to embrace and support the values that matter most to subscribers – namely, free speech and independent journalism.

    “As such, we have committed to establishing an independent advisory board tasked with upholding the highest standards of journalistic integrity. The deputy chairman of the Telegraph Media Group, Lord Black, has agreed to design its framework.”

    Continue Reading

  • Top 10 US billionaires’ collective wealth grew by $698bn in past year – report | US income inequality

    Top 10 US billionaires’ collective wealth grew by $698bn in past year – report | US income inequality

    The collective wealth of the top 10 US billionaires has soared by $698bn in the past year, according to a new report from Oxfam America published on Monday on the growing wealth divide.

    The report warns that Trump administration policies risk driving US inequality to new heights, but points out that both Republican and Democratic administrations have exacerbated the US’s growing wealth gap.

    Using Federal Reserve data from 1989 to 2022, researchers also calculated that the top 1% of households gained 101 times more wealth than the median household during that time span and 987 times the wealth of a household at the bottom 20th percentile of income. This translated to a gain of $8.35m per household for the top 1% of households, compared with $83,000 for the average household during that 33-year period.

    Meanwhile, over 40% of the US population, including nearly 50% of children, are considered low-income, with family earnings that are less than 200% of the national poverty line.

    When reviewing the 10 countries with the largest economies in the Organisation for Economic Co-operation and Development (OECD), the US has the highest rate of relative poverty, second-highest rate of child poverty and infant mortality, and the second-lowest life expectancy rate.

    “Inequality is a policy choice,” said Rebecca Riddell, senior policy lead for economic justice at Oxfam America. “These comparisons show us that we can make very different choices when it comes to poverty and inequality in our society.”

    The report outlines the way that systems in the US, including the tax code, social safety nets, and worker’s rights and protections, have been slowly dismantled, allowing concentrated wealth to turn into concentrated power.

    Donald Trump’s “one big, beautiful bill”, passed by Congress in May, has been one of the “single largest transfers of wealth upwards in decades”, according to the report, by cutting tax for the wealthy and corporations.

    But over the last few decades, Republicans have not acted alone.

    “Policymakers have been choosing inequality, and those choices have had bipartisan support,” Riddell said. “Policy reforms over the last 40 years, from cuts to taxes and the social safety net, to labor issues and beyond, really had the backing of both parties.”

    Policy recommendations outlined in the report fall into four categories: rebalancing power through campaign finance reform and antitrust policy; using the tax system to reduce inequality through taxes on the wealthy and corporations; strengthening the social safety net; and protecting unions.

    skip past newsletter promotion

    These solutions can be tricky to carry out politically because of long-term stigmatization, particularly of social safety nets and taxation. The report refers to the concept of the “welfare queen” popularized during Ronald Reagan’s presidency in the 1980s, while taxation has always been seen as repressive for all rather than as a tool for addressing inequality.

    “What’s really needed is a different kind of politics,” Riddell said. “One that’s focused on delivering for ordinary people by really rapidly reducing inequality. There are sensible, proven reforms that could go a long way to reversing the really troubling trends we see.”

    The report features interviews with community leaders who are actively working to reduce inequality, even as progress has seemingly stalled on the national stage. In one interview in the report, activists with United Workers Maryland said the current moment seems ripe with opportunity because many Americans are starting to see how the current set-up isn’t working for them, but only for the people at the very top.

    “I think it’s brilliant that they see this as an opportunity,” Riddell said. “I love thinking about this moment as an opportunity to look around us and realize our broader power.”

    This article was amended on 3 November 2025. The Oxfam America report looks at the 10 largest economies in the OECD, not all 38 countries.

    Continue Reading

  • The Change of Osteoimmune Microenvironment in Type 2 Diabetic Mice:A

    The Change of Osteoimmune Microenvironment in Type 2 Diabetic Mice:A

    Introduction

    Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder marked by insulin resistance and relative insulin deficiency, leading to persistent hyperglycemia. Unlike Type 1 diabetes mellitus (T1DM), which typically arises in childhood and involves autoimmune destruction of pancreatic β-cells, T2DM predominantly occurs in adults and is strongly associated with obesity, physical inactivity, and genetic predisposition.1,2 The rising global prevalence of T2DM underscores the urgent need to elucidate its pathophysiology, particularly the interplay between metabolic dysregulation, systemic inflammation, and tissue-specific complications, including those affecting the bone marrow.

    Chronic low-grade inflammation is increasingly recognized as a central contributor to T2DM progression. Immune cells, such as monocytes, macrophages, and neutrophils, mediate this inflammatory state, exacerbating insulin resistance and metabolic dysfunction. The bone marrow (BM), as the primary hematopoietic organ, serves both as a source and a regulator of these immune cells. Consequently, alterations in BM cellular composition and function can profoundly influence systemic inflammation, immune responses, and metabolic homeostasis.3,4 Notably, T2DM has been associated with an expansion of pro-inflammatory monocytes (eg, Cd36+ subsets) and skewing of macrophage polarization towards a pro-inflammatory phenotype, suggesting that immune dysregulation in the BM may contribute to both chronic inflammation and impaired bone remodeling.5,6 Furthermore, changes in the BM microenvironment, including stromal cell activity and extracellular matrix remodeling, can disturb hematopoiesis and immune responses, linking local BM alterations to systemic complications of T2DM such as cardiovascular disease and kidney injury.7–9

    Bone health is tightly connected to BM immune regulation, a relationship encapsulated by the concept of osteoimmunology. Immune cells within the BM, including monocytes, neutrophils, and B cells, influence osteoclast and osteoblast activity, thereby modulating bone formation and resorption. In T2DM, chronic hyperglycemia, metabolic stress, and inflammatory cytokines disrupt the balance of osteoclasts and osteoblasts, increasing the risk of osteoporosis and fracture.10–12 For instance, AP-1–mediated suppression of osteoclast activity has been described in diabetic bone, underscoring how molecular immune signals can reshape skeletal remodeling.13 Importantly, emerging evidence indicates that immune-mediated shifts in BM neutrophils, B cells, and monocytes not only impair local bone turnover but also exacerbate systemic inflammation and metabolic dysfunction, further connecting osteoimmune interactions to broader T2DM pathophysiology. Clinically, such immune-driven bone fragility has been associated with higher fracture incidence, delayed bone healing, and poorer outcomes after orthopedic surgery in diabetic patients, making this research area of significant translational relevance. Despite these insights, detailed mechanisms linking specific BM immune subsets to T2DM-related bone pathology remain poorly understood. A deeper understanding of how immune dysregulation contributes to T2DM and its complications is crucial for the development of targeted prevention and treatment strategies of bone health.14,15

    Technological advances such as single-cell RNA sequencing (scRNA-seq) now provide a transformative approach to dissect cellular heterogeneity and functional states at unprecedented resolution. This technology is particularly suited for studying the BM microenvironment in T2DM, where multiple immune and stromal cell types interact dynamically, and conventional bulk RNA-seq approaches cannot resolve subset-level changes or intercellular signaling networks.16,17 Previous scRNA-seq studies16,17 in diabetic models have explored general shifts in myeloid and lymphoid compartments; however, they often lack detailed subset annotation, functional state analysis, or ligand-receptor interaction characterization, leaving gaps in understanding BM immune dysregulation in T2DM.18,19 Thus, the precise mechanisms linking BM immune alterations to systemic inflammation and bone pathology in T2DM remain incompletely understood.

    To address these knowledge gaps, the present study leverages scRNA-seq to provide a comprehensive analysis of BM cells in T2DM mice. Specifically, our objectives are to: (1) profile immune cell heterogeneity, including neutrophil, monocyte, B cell, T cell, and dendritic cell subsets; (2) identify T2D-specific alterations in cellular proportions and functional states; and (3) characterize intercellular communication within the osteoimmune microenvironment through ligand-receptor analysis. By systematically mapping these changes, we highlight nuanced T2D-specific reprogramming of the BM immune landscape, uncover altered pathways such as THBS, CLEC, and IL-6 signaling, and provide mechanistic insights into how BM immune dysregulation contributes to systemic inflammation, insulin resistance, and bone remodeling defects in T2DM. Collectively, this study not only extends beyond prior research by integrating cellular, molecular, and intercellular dimensions of BM pathology, but also lays the foundation for future translation of scRNA-seq findings into therapeutic strategies that may alleviate immune dysfunction and skeletal complications in T2DM.

    Materials and Methods

    High-Throughput Gene Expression Data

    The high-throughput single-cell RNA sequencing data were downloaded from the GEO database (https://www.ncbi.nlm.nih.gov/geo/) under the accession number GSE212726. The dataset is based on the GPL24247 platform (Illumina NovaSeq 6000, Mus musculus) and includes two samples: whole bone marrow cells extracted from the femur and tibia of wild-type mice (Group WT) and those from T2DM mice (Group DM). These samples were selected for in-depth analysis in this study. This dataset was selected due to its comprehensive annotation, availability of metadata, and complete profiling of bone marrow immune cells. Other public datasets (eg, db/db or diet-induced models) were also screened through GEO and ArrayExpress using the terms: “diabetes”, “bone marrow”, “single-cell RNA-seq”, “mouse”, and “immune.” Inclusion criteria required (a) availability of raw counts or processed matrices, (b) annotation of bone marrow tissue origin, (c) specification of diabetes induction method, and (d) sufficient sequencing depth and metadata. While GSE212726 was prioritized, future studies will incorporate additional murine and human datasets for validation and translational extension.

    Cell Clustering Analysis, Visualization, and Annotation

    To ensure data quality, Low-quality cells (<200 detected genes or >10% mitochondrial transcripts) and potential doublets (>25,000 UMIs) were excluded. Additional doublet detection was performed using DoubletFinder v2.0. Data normalization and regression were performed with Seurat v5.0.3, generating scaled data from UMI counts and mitochondrial content. For cell normalization and regression, we utilized the Seurat package (version 5.0.3, https://satijalab.org/seurat/). The process involved using the expression matrix, unique molecular identifier (UMI) counts per sample, and mitochondrial content to generate scaled data. Principal Component Analysis (PCA) was performed on the top 2,000 highly variable genes, and the first 10 principal components were selected for constructing t-SNE and UMAP plots. To address batch effects across samples, we applied the fastMNN function from the scran package (version 1.12.1) with parameters set to k=5, d=50, and approximate = TRUE, leveraging the mutual nearest neighbor method. An unsupervised cell clustering analysis was performed using a graph-based method with a resolution of 0.4, focusing on the top 10 PCA components. The identification of marker genes was conducted via the FindAllMarkers function, utilizing the Wilcoxon rank sum test algorithm with the following criteria: (1) log fold change (logFC) greater than 0.25; (2) p-value less than 0.05; (3) minimum percentage (min.pct) exceeding 0.1. To refine cell type characterization, clusters identified as the same cell type were selected for additional re-tSNE analysis, followed by graph-based clustering and marker identification.

    Pseudo-Time Analysis

    Pseudotime analysis was performed using Monocle 2 [Monocle2](http://cole−trapnell−lab.github.io/monocle−release), utilizing the DDR-Tree algorithm with default settings. Before running the Monocle analysis, we selected marker genes identified from the Seurat clustering results and utilized raw expression counts of filtered cells. Following the pseudotime analysis, Branch Expression Analysis Modeling (BEAM)20 was employed to investigate genes responsible for determining cell fate at specific branch points.

    Cell-Cell Communication Analysis

    We used CellphoneDB v 2.1.7,21 a comprehensive database of ligands, receptors, and their interactions, to systematically examine cell-cell communication molecules. For clusters at various time points, we annotated membrane, secreted, and peripheral proteins. We identified significant interactions by evaluating the mean interaction scores and cell communication significance, considering only those interactions with a p-value less than 0.05. These interactions were based on the normalized cell matrix obtained through Seurat’s normalization process. Normalized Seurat matrices were input, and results highlight potential T2D-specific signaling (eg, THBS, CLEC, IL-6). These predictions are computational and require experimental confirmation (eg, blocking antibodies, co-culture assays).

    Differential Expression Genes (DEGs) Analysis

    To identify differentially expressed genes (DEGs) among samples, we utilized the FindMarkers function, which employs the Wilcoxon rank-sum test algorithm. The criteria for selecting DEGs were as follows: a logFC greater than 1, an adjusted p-value below 0.05, and a minimum expression percentage (min.pct) above 0.1.

    Gene Ontology Analysis

    To understand the biological significance of marker genes and DEGs, we conducted Gene Ontology (GO) analysis. GO annotations were sourced from NCBI (http://www.ncbi.nlm.nih.gov/), UniProt (http://www.uniprot.org/), and the Gene Ontology database (http://www.geneontology.org/). Fisher’s exact test was applied to identify significantly enriched GO categories, and we adjusted p-values using False Discovery Rate (FDR) correction.22

    Pathway Analysis

    Pathway analysis was conducted to identify key pathways associated with marker genes and DEGs by leveraging the KEGG database.23 Significant pathways were selected using Fisher’s exact test, with p-values and FDR serving as thresholds for significance. Particular attention was given to osteoimmunology-relevant pathways.24

    Quantitative Real-Time Reverse Transcription Polymerase Chain Reaction (qRT–PCR)

    Type 2 diabetes mellitus (T2DM) was induced in C57BL/6J mice by feeding a high-fat diet (HFD, D12492, Research Diets; 60% kcal from fat) for 8 weeks, followed by a single low-dose intraperitoneal injection of streptozotocin (STZ, 100 mg/kg, Sigma). This HFD+STZ combination model is widely used to mimic the pathophysiology of T2DM, as HFD induces insulin resistance and low-dose STZ causes partial β-cell dysfunction. Successful induction of T2DM was confirmed by measuring fasting blood glucose levels (>11.1 mmol/L), impaired glucose tolerance tests (GTT), and increased body weight compared with WT controls. Control mice received a standard chow diet. Bone marrow samples from three mice per group, were harvested from the femur and tibia for s qRT–PCR. Although a formal power analysis was not conducted prior to sample collection due to the high cost and complexity of scRNA-seq, we acknowledge this as a limitation and note that future studies with larger cohorts will be needed to further validate these findings. We acknowledge that the small sample size limits statistical power, and STZ-induced diabetes may have cytotoxic effects independent of hyperglycemia; complementary models such as db/db or diet-induced T2D will be needed in future studies. For validation experiments, key genes, including FOSB, RELB, IL1B, MAP3K7, PPP3R1, TNF, TGFBR2, and SOCS3, were selected for qRT-PCR analysis using RNA extracted from the bone marrow of diabetic (DM) and control (WT) groups. Purified DNA-free RNA was isolated using the RNeasy Mini Kit, reverse transcribed using the One Step PrimeScript® miRNA cDNA Synthesis Kit (Takara, Japan, D350A), and amplified using the SYBR® Premix Ex Taq™ II Kit (Takara, Japan, DRR820A), following the manufacturer’s instructions. Gene expression levels were calculated using the ΔΔCT method after normalization to the expression of the GAPDH housekeeping gene. Primer sequences used for qRT-PCR are provided in Supplementary Table 1. Each reaction was performed in triplicate technical replicates for each of the three biological samples per group. Melt curve analysis was conducted to confirm amplification specificity.

    Statistical Analysis

    All statistical analyses and data visualizations were conducted using R software (version 4.3.3). GraphPad Prism software (version 8.1.0; GraphPad Software Inc., USA) was used to perform statistical analyses and generate graphs.25 One-way analysis of variance (ANOVA) was applied to compare gene expression levels and inflammatory factor concentrations. For comparisons between two groups, unpaired two-tailed Student’s t-tests were used. For multiple group comparisons, one-way ANOVA followed by Tukey’s post hoc test was applied. Proportional changes were assessed with chi-square tests. Results were presented as means ± standard errors (SEM), and a p-value of less than 0.05 was considered statistically significant.

    Results

    Profiles of Bone Marrow Cells in WT and T2D Mice

    The high-throughput scRNA-seq data were downloaded under accession number GSE212726. In total, 10,420 cells from WT mice and 11,577 cells from T2D mice were collected. After filtering low-quality cells and duplicates, 9,360 cells from WT and 10,885 cells from T2D mice were retained for analysis (Figure 1A). Unbiased clustering identified 12 distinct cell populations: monocytes (primarily expressing Ass1, Trem2, and Lgals1), basophils (primarily expressing Prss34, Mcpt8, and Gata2), BM-neutrophils (primarily expressing Orm1, Ffar2, and Pla2g7), T lymphocytes (T cells, primarily expressing Cd3e, Icos, and Cd5), B lymphocytes (B cells, primarily expressing Cd79a, Iglc2, and Ms4a1), stem cells (primarily expressing Myl10, Pdzk1ip1, and Krt18), mesenchymal stem cells (MSCs, primarily expressing Hist1h1d and Top2a), hematopoietic stem cells (HSCs, primarily expressing Pdk4, Apoe, and Tgm2), neutrophil-myeloid progenitors (NMP, primarily expressing Mpo, Elane, and Gstm1), dendritic cells (DCs, primarily expressing Siglech, Bst2, and Irf8), NK cells (primarily expressing Klra8, Ncr1, and Adamts14), and erythrocytes (primarily expressing Hba-a2) (Figure 1BF).

    Figure 1 Single-cell RNA sequencing provide a comprehensive atlas of bone marrow cells in WT and T2D mice. (A) Study overview. (B-C) Clustering of bone marrow cells into twelve distinct clusters. (B) UMAP of unbiased clustering of all bone marrow cells in groups WT and DM; (C) UMAP of unbiased clustering and cell annotation of bone marrow cells in group WT and DM, respectively. (D) The proportions of each cell in groups WT and DM;*:p < 0.05. (E) Key cell type marker genes of 12 cell clusters, the redder the color, the higher the expression. (F) Cluster signature genes highlighted on left. Expression of top differentially expressed genes (rows) across the cells (columns), the warmer the color, the higher the expression.

    Cell proportion analysis revealed significant differences between WT and T2D mice. B cells (9.16% → 19.78%), MSCs (2.50% → 4.80%), monocytes (3.72% → 4.92%), and HSCs (1.36% → 4.00%) increased, while BM-neutrophils (66.56% → 54.73%), DCs (3.25% → 1.71%), basophils (1.68% → 0.89%), T cells (2.08% → 1.37%), and NMPs (6.94% → 5.82%) decreased. Stem cells, erythrocytes, and NK cells exhibited slight decreases. The top increases and decreases were observed in B cells and BM-neutrophils, respectively (Figure 1D).

    Heterogeneity of BM-Neutrophils in the Bone Marrow of T2D Mice

    M-neutrophils exhibited the largest proportional reduction. Six subsets were identified: BM-neutrophil_0 (primarily expressing Mpo, Elane, and Gstm1), BM-neutrophil_1 (primarily expressing Slfn5, Mpeg1, and Wfdc17), BM-neutrophil_2 (primarily expressing Retnlg, Ceacam10, and Mmp9), BM-neutrophil_3 (primarily expressing Orm1, Tmem216, and Zmpste24), BM-neutrophil_4 (primarily expressing Chit1, Fcnb, and Mogat2), and BM-neutrophil_5 (primarily expressing Clec4d, Vnn3, and Cstdc4) (Figure 2AD).

    Figure 2 The characteristics of BM-neutrophils between groups WT and DM. (A) The tSNE results of BM-neutrophil cluster among all cells. (B) UMAP of BM-neutrophil cluster among all cells and six BM-neutrophil subsets, and UMAP of unbiased clustering and cell annotation of BM-neutrophil subsets in groups WT and DM. (C) The proportion of BM-neutrophil subsets in groups WT and DM;*:p < 0.05. (D) Key marker genes of BM-neutrophil subsets. UMAP of key marker genes of BM-neutrophil subsets, along with the corresponding distribution of expression levels among 12 clusters and 6 BM-neutrophil subsets respectively. Key marker genes included Mpo, Slfn5, Retnlg, Orm1, Chit1 and Clec4d.

    Proportional changes were as follows: BM-neutrophil_0 (50.19% → 39.72%), BM-neutrophil_1 (13.21% → 23.87%), BM-neutrophil_2 (15.47% → 11.01%), BM-neutrophil_3 (11.06% → 13.95%), BM-neutrophil_4 (5.75% → 5.64%), BM-neutrophil_5 (4.32% → 5.81%). Statistical significance was assessed with chi-square tests (p < 0.05 for all major changes) (Figure 2C).

    Among the DEGs across the six neutrophil subsets, Arfgef1, Mrpl15, Ppplr42, Rblcc1, Rpl7, Sgk3, Tcea1, and Tram1 exhibited the most significant changes in expression levels. MPO, a heme protein crucial for neutrophil azurophilic granules,26 was predominantly expressed in BM-neutrophil_0. Slfn5, a Schlafen family protein induced by interferon,27 was mainly expressed in BM-neutrophil_1 and BM-neutrophil_0. Retnlg, also known as resistin-like gamma,28 was expressed in BM-neutrophil_2 and BM-neutrophil_5. Orm1, an acute phase plasma protein,29 was primarily found in BM-neutrophil_3 and BM-neutrophil_4. Chit1, a conserved chitinase secreted by activated macrophages,30 was mainly expressed in BM-neutrophil_4. Clec4d, a receptor for trehalose-6,6’-dimycolate,31 was expressed in both BM-neutrophil_2 and BM-neutrophil_5 (Figure 2D).

    Differential directions and Monocle pseudotime trajectory expression pattern of 6 BM-neutrophil subsets were presented with a Heatmap presenting relative expressions of markers of BM-neutrophils along inferred trajectories in 6 BM-neutrophil subsets (Figure 3A and B).

    Figure 3 Differential trajectory of BM-neutrophil subsets and enrichment of DEGs. (A-D) Differential directions of 6 BM-neutrophil subsets: (A) Heatmap presenting relative expressions of markers of BM-neutrophils along inferred trajectories in 6 BM-neutrophil subsets. The red and blue branches correspond to the two differential directions. (B) Monocle pseudotime trajectory expression pattern of 6 BM-neutrophil subsets. (C) Pseudotime trajectories of the BM-neutrophil subsets. (D) The heatmap of branch point 1 shown in Figure 3C. (E) The top 8 key DEGs in these two cell fate. (F) Signature genes of BM-neutrophil subsets. Subset signature genes highlighted on left. Expression of signature genes (rows) across the cells (columns), the warmer the color, the higher the expression. (G) GO analysis of BM-neutrophil_1. Bubble diagram of upregulated DEGs of BM-neutrophil_1 enriched in GO analysis. (H) KEGG analysis of BMneutrophil_1. Bubble diagram of upregulated DEGs of BM-neutrophil_1 enriched in KEGG analysis. (I-J) The top 5 DEGs of BM-neutrophils. The top 5 upregulated DEGs between groups WT and DM (I). The top 5 downregulated DEGs between groups WT and DM (J).

    Pseudotime analysis revealed two differentiation trajectories originating from BM-neutrophil_0. DEGs in trajectory 1 (Gm15818, Adhfe1, Arfgef1, Terf1, Rb1cc1, Mcmdc2, Sgk3) were associated with apoptosis regulation, hypoxia response, and cellular stress. Trajectory 2 DEGs (Tcf24, Rdh10, Lypla1, Cops5, Cspp1, Mybl1, Ncoa2, Snhg6, Vcpip1, Lactb2) suggest functional diversification. GO and KEGG enrichment showed upregulation in protein processing, oxidative phosphorylation, TNF signaling, osteoclast differentiation, IL-17 signaling, and lipid metabolism. Pathways labeled as “neurodegeneration” and “ALS” likely reflect shared stress and apoptosis mechanisms relevant to BM dysfunction in T2D (Figure 3CH). Because BM-neutrophil_0 constituted the largest proportion of cells in group DM, we performed GO enrichment and KEGG pathway analyses on the upregulated DEGs identified in the BM-neutrophil_0 cluster, which indicated that the DEGs in BM-neutrophil_0 were primarily associated with the negative regulation of apoptotic processes, negative regulation of neuron apoptosis, and responses to hypoxia. The most common cellular components were the cytoplasm, extracellular region, and plasma membrane, while protein binding, identical protein binding, and hydrolase activity were the predominant molecular functions (Figure 3G). KEGG pathway analysis identified several highly enriched pathways among the DEGs, including neurodegeneration-related diseases, protein processing in the endoplasmic reticulum, amyotrophic lateral sclerosis, lipid and atherosclerosis, diabetic cardiomyopathy, NOD-like receptor signaling, oxidative phosphorylation, osteoclast differentiation, TNF signaling, Fc gamma R-mediated phagocytosis, and IL-17 signaling (Figure 3H). The top five upregulated DEGs were Vcan, Pla2g7, lghg2b, Gm43305, and Cfh, while the top five downregulated DEGs were Prg2, Gata2, TRbc1, Cox6a2, and Lpl (Figure 3I and J).

    Heterogeneity of B Cells

    B cells were classified into precursor B cells (Pre_B, primarily expressing Ly6a, Cxcr5, and Ccr6) and naïve B cells (Naïve_B), primarily express IL2ra, Cd72, and Fam129c) (Figure 4AD). Naïve_B increased from 64.29% to 82.03%, while Pre_B decreased from 35.71% to 17.97% (Figure 4C, p < 0.01). KEGG analysis revealed enrichment in antigen processing, insulin resistance, lipid metabolism, and NOD-like receptor signaling, reflecting functional alterations in immune regulation in T2D (Figure 4E and F). GO enrichment and KEGG pathway analyses were conducted for the upregulated DEGs in B cells, which revealed that the DEGs were predominantly associated with immune system processes, negative regulation of transcription by RNA polymerase II, and innate immune responses. The most common cellular components were the cytoplasm, nucleus, and cytosol, while the major molecular functions included protein binding, identical protein binding, and hydrolase activity (Figure 4E). KEGG pathway analysis identified several enriched pathways, including protein processing in the endoplasmic reticulum, lipid and atherosclerosis, antigen processing and presentation, estrogen signaling, NOD-like receptor signaling, measles, insulin resistance, and cholesterol metabolism (Figure 4F). The top five upregulated genes were Ddit4, Hist1h1b, Ighg2b, Ifi27l2a, and Cfh, while the top five downregulated genes were Ccl3, Klf4, Scd2, Hes1, and Lpl (Figure 4G and H).

    Figure 4 The characteristics of B cells between groups WT and DM. (A) The tSNE results of B cells cluster among all cells. (B) UMAP of the cluster of B cells among all cells, and UMAP of unbiased clustering and cell annotation of the subsets of B cells in groups WT and DM. (C) The proportion of the subsets of B cells in groups WT and DM. (D) Key marker genes of B cells subsets. UMAP of key marker genes of the subsets of B cells, along with the corresponding distribution of expression levels among 12 clusters. Key marker genes included Ly6a, Cxcr5, Il2ra, and Fam129c. (E) GO analysis of Naïve_B. Bubble diagram of upregulated DEGs of Naïve_B enriched in GO analysis. (F) KEGG analysis of Naïve_B. Bubble diagram of upregulated DEGs of Naïve_B enriched in KEGG analysis. (G-H) The top 5 DEGs of B cells. The top 5 upregulated DEGs between groups WT and DM (G). The top 5 downregulated DEGs between groups WT and DM (H).

    Heterogeneity of Monocytes, T Cells, and DCs

    Monocytes were subdivided into five subsets: monocyte_0 (primarily expressing Tprgl, S100a9, and S100a8), monocyte_1 (primarily expressing Ccnd1, Irf8, and Cst3), monocyte_2 (primarily expressing Apoc2, Mafb, and Ccr2), monocyte_3 (primarily expressing mt-Nd2, Mki67, and Zeb2), and monocyte_4 (primarily expressing Gpi1, Srgn, and Cd63) (Figure 5AC). Monocytes (monocyte_0–4) showed shifts: monocyte_0 and monocyte_4 decreased, while monocyte_2 and monocyte_3 increased (Figure 5B). T cells were categorized into four subsets: γδ T cells (Tgd), primarily expressing Cd3e, Cd3d, and Thy1; natural killer cells (NK), primarily expressing Ncr1, Gzma, and Klra8; naïve T cells (T), primarily expressing Cd19, Plaur, and Cd74; and CD4+ T cells (CD4 T), primarily expressing Cd4, Foxp3, and Nrpl (Figure 5DF). Proportional changes revealed that Tgd and naïve T cells decreased, and CD4+ T and NK cells increased (Figure 5E). DCs were classified into conventional DCs (cDC), primarily expressing Fcgr1, Ifi205, and Naaa; plasmacytoid DCs (pDC), primarily expressing Ccr9, Cox6a2, and Tlr7; and another DC subset, primarily expressing Clec9a, Sox4, and Calm. pDCs decreased while cDCs and the other subset slightly increased (Figure 5GI).

    Figure 5 The characteristics of monocytes, T cells and DCs between groups WT and DM. (A) Five monocyte subsets: UMAP of unbiased clustering and cell annotation of the subsets of monocytes in groups WT and DM, and UMAP of unbiased clustering of the subsets of monocytes in groups WT and DM. (B) The proportion of the subsets of monocytes in groups WT and DM. (C) Signature genes of monocyte subsets. Subset signature genes highlighted on left. Expression of signature genes (rows) across the cells (columns), the warmer the color, the higher the expression. (D) Four T cells subsets: UMAP of unbiased clustering and cell annotation of the subsets of T lymphocytes in groups WT and DM, and UMAP of unbiased clustering of the subsets of T lymphocytes in groups WT and DM. (E) The proportion of the subsets of T cells in groups WT and DM. (F) Signature genes of T cells subsets. Subset signature genes highlighted on left. Expression of signature genes (rows) across the cells (columns), the warmer the color, the higher the expression. (G) Four dendritic cell subsets: UMAP of unbiased clustering and cell annotation of the subsets of DCs in groups WT and DM, and UMAP of unbiased clustering of the subsets of DCs in groups WT and DM. (H) The proportion of the subsets of DCs in groups WT and DM. (I) Signature genes of dendritic cell subsets. Subset signature genes highlighted on left. Expression of signature genes (rows) across the cells (columns), the warmer the color, the higher the expression.

    Ratios of Immune Cells

    We explored whether alterations in the ratios of immune cells within the bone marrow of WT and T2D mice. DM/WT ratios highlighted immune imbalances: monocytes/DCs ratio was highest, with BM-neutrophils/T cells and BM-neutrophils/DCs ratios increased, and BM-neutrophils/Naïve_B decreased (Figure 6A and B).

    Figure 6 Identification of the ratio of immune cells between groups WT and DM. (A) The ratio of different immune cells. The ratio of BM-neutrophils/T cells, BM-neutrophils/B cells, BM-neutrophils/DCs, BM-neutrophils/monocytes, monocytes/T cells, monocytes/B cells, monocytes/DCs, BM-neutrophil_1/Naïve_B in groups WT and DM. (B) The rate of DB/WT among different immune cells. The RD/RC of BM-neutrophils/T cells, BM-neutrophils/B cells, BMneutrophils/DCs, BM-neutrophils/monocytes, monocytes/T cells, monocytes/B cells, monocytes/DCs, and BM-neutrophil_1/Naïve_B.

    Cell-to-Cell Communication Analysis in the Bone Marrow of T2D Mice

    To investigate the osteoimmune microenvironment in T2D mice, we conducted a cell-to-cell communication analysis using CellphoneDB. This analysis assessed interactions among various cell types in both WT and DM samples. Gaining insights into these cellular interactions will help clarify the underlying mechanisms of bone pathology and reveal potential targets for therapeutic intervention in T2D-associated bone loss. Figure 7A illustrates the differential number and strength of interactions. CellphoneDB analysis revealed altered interaction strengths in T2D: incoming signals increased in BM-neutrophils, monocytes, MSCs, and decreased in B and T cells; outgoing signals increased in BM-neutrophils and basophils, decreased in T cells (Figure 7B and C indicates that differences in signaling pathways of all cell types between WT and DM groups.

    Figure 7 Cell-to-cell communication analysis using CellphoneDB to evaluate interactions between cell types in the WT and DM samples. (A) Differential number and strength of interactions between cell types in group DM and WT samples, with red indicating up-regulated and blue indicating down-regulated interactions. (B) Changes in cell roles: incoming and outgoing interaction strengths of immune cells. (C) Differences in signaling pathways of all cell types between WT and DM groups. (D) Differences in signaling pathways of T cells, monocytes, MSCs, and BM-Neutrophil between WT and DM groups. (E-F) Altered incoming (E) and outgoing (F) signaling patterns of immune cells in the WT and DM groups, respectively.(G) Changes ligand-receptor relationships between T cells and other immune cells in the DM group.((H) Changes in ligand-receptor relationships between BM-neutrophils and other immune cells in the DM group. (I-J) Relative expression of genes associated with the LCK and THBS signaling pathways cross all cell types in WT and DM groups.

    Next, we examined differences in signaling pathways between WT and DM groups. Top enriched pathways in DM were THBS, VISFATIN, CLEC, IL4, IL6, whereas the top enriched pathways in the WT group were LCK, IFN-II, CD6, ALCAM, and CD86. Key ligand-receptor pairs included Thbs1–Cd47 (THBS pathway) and Clec4d–Fcgr2b (CLEC pathway), providing mechanistic insights into immune cell interactions in T2D (Figure 7DH).

    Specifically, Figure 7D indicates that CLEC was a specific outgoing signaling pathway for T cells in the DM group, while THBS was both an outgoing and incoming specific signaling pathway for monocytes, MSCs, and BM-neutrophils in the DM group. Figure 7E and F depicts the altered incoming and outgoing signaling patterns of immune cells in the WT and DM groups, respectively. To examine the ligand-receptor interactions among immune cells, we focused on the relationships between T cells and various other immune cell types. Our analysis revealed notable communication between T cells and Ccl5-Ccr1, confirming the molecular targets and bidirectional communication potential of T cells with other immune cells in the DM group (Figure 7G). Figure 7G also shows the changes in signaling involving ligand-receptor relationships between T cells and other immune cells in the DM group. Figure 7H illustrates the changes in signaling involving ligand-receptor relationships between BM-neutrophils and other immune cells in the DM group. Finally, Figures 7I and J present the relative expression of genes associated with the LCK and THBS signaling pathways in all cell types between the WT and DM groups.

    Gene Expressions Validation by qRT-PCR Analysis

    To validate gene expression patterns, quantitative real-time PCR (qRT-PCR) analysis was performed. The results demonstrated significantly reduced relative expression of FOSB, RELB, IL1B, MAP3K7, PPP3R1, TNF, TGFBR2, and SOCS3 in T2D bone marrow (Figure 8), consistent with scRNA-seq results, indicating strong concordance between the two methods.

    Figure 8 qRT-PCR Analysis of the expression levels of key genes in bone marrow of T2D mice and control samples.

    Discussion

    The advent of scRNA-seq has transformed our understanding of cellular heterogeneity and the complex molecular mechanisms underlying T2DM. By enabling the analysis of individual cells within heterogeneous tissues, scRNA-seq provides a high-resolution view of the cellular and molecular landscape, allowing us to dissect complex immune and stromal interactions within the bone marrow (BM). Our study leverages this technology to analyze BM cells from STZ-induced T2D mice, revealing significant alterations in immune cellular composition, subset functionality, and intercellular communication. These findings offer valuable insights into how immune remodeling contributes to systemic inflammation, insulin resistance, and bone pathology in T2DM.

    In total, twelve distinct cell clusters were identified within the BM, including monocytes, basophils, BM-neutrophils, T cells, B cells, stem cells, MSCs, HSCs, NMP, DCs, NK cells, and erythrocytes. When comparing T2D mice to wild-type controls, significant compositional changes were observed. Notably, T2D mice exhibited a substantial increase in B lymphocytes and MSCs, along with a decrease in BM-neutrophils and DCs, consistent with broad dysregulation of hematopoietic and immune cell populations in diabetes. These shifts may influence systemic inflammation, bone remodeling, and metabolic homeostasis, emphasizing the interconnected nature of immune dysfunction and bone pathology. Since immune cells in the BM are closely linked to both systemic inflammation and skeletal remodeling, these compositional changes may have profound implications for metabolic homeostasis and osteoimmune balance.32–34

    Among the most striking findings was the pronounced decrease in BM-neutrophils, dropping from 66.56% in WT to 54.73% in T2D mice, which suggests altered neutrophil maturation or reprogramming. Neutrophils are central players in innate immunity, contributing to pathogen clearance, inflammation resolution, and bone remodeling via cross-talk with osteoclasts and osteoblasts. Their reduction in T2D may reflect impaired maturation or altered trafficking, consistent with prior studies linking neutrophil dysfunction to increased infection susceptibility and chronic inflammation in diabetes.35–38 This dysfunction may also contribute to systemic insulin resistance. Dysregulated pathways such as CXCL12/CXCR3 could underlie aberrant neutrophil migration within BM, further impairing osteoimmune signaling. Although our computational analyses point to such mechanisms, functional assays—including NETosis, phagocytosis, or migration studies—will be essential to validate these predictions and clarify how neutrophil dysfunction contributes to bone pathology.

    Conversely, B cells exhibited a notable increase in proportion, from 9.16% in WT to 19.78% in T2D mice, particularly within the Naïve_B subset. This expansion highlights adaptive immune activation in response to chronic metabolic stress. Activated B cells are known to produce pro-inflammatory cytokines and pathogenic antibodies, which can exacerbate insulin resistance and contribute to systemic inflammation. Previous studies revealed that B cells modulate immune responses and influence insulin resistance through antibody-mediated pathways and cytokine production, and their dysregulation has been associated with T2D complications, including cardiovascular disease and systemic inflammation.39–42 The observed shift from Pre_B to mature B cell populations may reflect compensatory responses to T2D-induced chronic inflammation and metabolic stress that may exacerbate inflammatory signaling and systemic metabolic dysfunction.43,44 These findings underscore the need to consider B cells as active contributors to T2D pathophysiology. Future studies could explore therapeutic targeting of B cell-derived cytokines, such as CCL4 and CCL5, to mitigate systemic inflammation.

    Monocyte and T cell subsets exhibited significant alterations. Reductions in monocyte_0 and monocyte_4, coupled with increases in monocyte_2 and monocyte_3, suggest altered monocyte-mediated inflammation, likely contributing to chronic low-grade inflammation and insulin resistance. Prior studies have reported Cd36+ monocytes with diminished osteoclast potential in diabetic bone marrow, highlighting the importance of subset-level analyses for linking immune dysregulation to bone remodeling. Our data extend these findings, by providing subset-level resolution of T2D-specific monocyte alterations.45–52 Similarly, T cell analysis revealed a decrease in γδ T cells and an increase in CD4+ T cells, particularly Th1 and Th17 subsets. While γδ T cells contribute to tissue homeostasis and immune surveillance, Th1/Th17 cells drive chronic inflammation and exacerbate metabolic dysregulation.53–57 These results collectively suggest that T2D reshapes both innate and adaptive immune compartments, thereby directly influencing osteoimmune dynamics and bone remodeling capacity.

    In addition, we observed a significant reduction in DCs, particularly plasmacytoid DCs (pDCs). Since pDCs are crucial for type I interferon responses and immune surveillance, their depletion could exacerbate chronic inflammation and impair host defense in T2D.58–63 From an osteoimmunology perspective, DC–B cell crosstalk and DC-mediated regulation of osteoclastogenesis are key determinants of bone homeostasis. Their loss may therefore contribute to T2D-associated bone remodeling defects. Functional assays such as flow cytometry or in vitro osteoclastogenesis studies will be needed to confirm these transcriptomic observations.

    Intercellular communication analysis using CellPhoneDB further highlighted T2D-specific signaling pathways, including THBS, CLEC, and IL-6. These interactions reinforce pro-inflammatory crosstalk between BM-neutrophils, monocytes, and MSCs, creating a feed-forward loop of immune activation. THBS signaling has known crosstalk with TGF-β, which regulates osteoblast-osteoclast balance, linking immune dysregulation to altered bone metabolism in T2D. CLEC signaling was enriched in T cells, while THBS was prominent in monocytes, MSCs, and BM-neutrophils, supporting the notion of immune-mediated modulation of bone remodeling. Together, these cellular and molecular changes illustrate a network of immune dysregulation in the bone marrow that can drive systemic metabolic disturbances and bone remodeling defects in T2D.64 KEGG pathway enrichment revealed terms such as “neurodegeneration” and “amyotrophic lateral sclerosis”. While these may appear unrelated to BM, they likely reflect shared inflammatory mechanisms—including oxidative stress, mitochondrial dysfunction, and apoptosis—common to both neurodegenerative disorders and diabetic BM immune cells. We have prioritized pathways with established osteoimmunology relevance (RANKL/OPG, IL-17, TNF) while noting that neurodegeneration-associated enrichments indicate overlapping inflammatory processes. Our enrichment analyses of signaling pathways related to osteoclast differentiation and inflammation, such as TNF and IL-17 pathways, highlight the role of immune cell interactions in bone pathology and metabolic disorders.65–67 Notably, Mac_OLR1 macrophages, known to support osteoclastogenesis, and Neut_RSAD2 neutrophils, which may impair osteoclast activity under diabetic stress, were transcriptionally linked to pathways such as AP-1 and FoxM1, bridging immune remodeling with T2D-specific skeletal phenotypes.13,68 Previous osteoimmunology studies have shown that Mac_OLR1 macrophages promote osteoclast differentiation through RANKL and cytokine signaling, directly enhancing bone resorption.69 Similarly, Neut_RSAD2 neutrophils can release reactive oxygen species and pro-inflammatory mediators that modulate osteoblast activity, potentially inhibiting bone formation.69 These results strengthen the hypothesis that altered immune–bone signaling underlies diabetic skeletal pathology. However, functional perturbation studies—such as blocking antibodies or ligand–receptor inhibition—are needed to establish causal effects. Integration with spatial transcriptomics would also help clarify the anatomical context of these altered interactions.

    Comparison with other diabetic contexts provides further perspective. In T1D models, neutrophil expansion and B cell reduction are often observed, whereas T2D mice displayed the opposite trend, suggesting divergent immunometabolic adaptations. This underscores the necessity of disease-specific studies to understand immune-bone crosstalk in diabetes.19,70,71 This underscores the importance of disease-specific investigations of osteoimmune regulation. Importantly, the STZ-induced T2D model employed here reflects β-cell injury, which may not fully capture the complexity of human T2D. Incorporating db/db or diet-induced models, alongside human scRNA-seq datasets, would enhance translational relevance and help identify conserved versus model-specific immune alterations.

    Relative to previous scRNA-seq studies16,17 that broadly characterized myeloid and lymphoid compartments, our analysis provides deeper resolution of subset-specific changes and reveals novel ligand–receptor pathways underlying osteoimmune dysfunction in T2D. These findings advance our understanding of the BM microenvironment in diabetes and generate new hypotheses for targeted therapeutic intervention.

    Although our results derive from an STZ-induced T2D mouse model, emerging human scRNA-seq data suggest parallels in immune dysregulation, including alterations in Naïve_B and monocyte subsets. Integrating human data, along with spatial transcriptomics, could map immune–stromal interactions with greater precision. Longitudinal studies across disease progression will be crucial for establishing causal relationships between immune remodeling and bone deterioration. These steps are essential for translating experimental observations into clinically meaningful therapies.

    Several limitations warrant consideration. First, this study relied on a single publicly available dataset (GSE212726) with a small sample size (n=3/group), limiting statistical power and generalizability. Replication in larger and independent cohorts will be essential to confirm the reproducibility. Second, the STZ-induced diabetes model does not fully recapitulate the polygenic, obesity-associated, and insulin resistance–dominated pathophysiology of human T2DM, and STZ itself may exert off-target cytotoxic effects on bone marrow cells. Complementary models such as db/db or diet-induced T2D mice should be incorporated to enhance translational relevance. Third, this study relied solely on transcriptomic data without protein-level or functional validation. Gene expression changes may not directly reflect biological activity, and ligand–receptor interactions were inferred computationally rather than experimentally verified. Future validation through approaches such as flow cytometry, cytokine profiling, and in vitro functional assays will be essential. Similarly, ligand–receptor predictions were based on computational inference rather than experimental verification. Fourth, the analysis was restricted to a single time point (7 months), limiting insight into disease progression. Since immune remodeling in T2D is dynamic, longitudinal profiling would provide deeper understanding of temporal immune and metabolic changes. Finally, the study did not integrate multi-omics approaches. Combining scRNA-seq with single-cell ATAC-seq, proteomics, or spatial transcriptomics could better capture transcriptional regulation, protein activity, and the spatial organization of immune–stromal interactions. Future work should integrate multiple datasets, complementary omics platforms, and longitudinal sampling to strengthen robustness.

    Future studies could extend our findings by performing functional validation of key immune cell subsets and signaling pathways identified in T2D bone marrow. For example, in vivo or ex vivo experiments could assess the role of BM-neutrophil_0 and Naïve_B subsets in modulating systemic inflammation and insulin resistance. Targeted depletion or adoptive transfer experiments may clarify their causal contribution to metabolic dysfunction. Additionally, blocking or enhancing specific ligand-receptor interactions, such as THBS, CLEC, or IL-6 pathways, could reveal their functional impact on intercellular communication and bone remodeling. Longitudinal studies in T2D mouse models would also help determine how immune cell dynamics evolve over disease progression and influence bone health. Finally, integrating proteomic or metabolomic analyses could provide mechanistic insights into how altered signaling pathways contribute to chronic inflammation and metabolic dysregulation in T2D.

    Conclusion

    The present study, utilizing single-cell RNA sequencing (scRNA-seq), provides comprehensive insights into immune landscape alterations in the bone marrow of a T2DM mouse model. This approach has revealed significant changes in B cells, monocytes, T cells, and dendritic cells, along with altered intercellular signaling pathways, revealing nuanced T2D-specific reprogramming of the bone marrow immune microenvironment. These alterations are likely to influence osteoimmune interactions, affecting osteoclast and osteoblast activity, and thereby contributing to bone remodeling defects alongside systemic inflammation and insulin resistance. While these findings deepen our understanding of immune-mediated mechanisms in diabetic bone pathology, we acknowledge that mouse models may not fully replicate human T2DM pathology. Future studies should incorporate orthogonal datasets, human validation, longitudinal sampling, functional assays, and spatial transcriptomics to confirm these findings and enhance translational relevance.

    Data Sharing Statement

    The datasets presented in this study can be found in online repositories. The names of the repository/repositories and accession number(s) can be found below: https://www.ncbi.nlm.nih.gov/geo/, accession ID: GSE212726.

    Ethics Approval and Consent to Participate

    All protocols in this study were approved by the Committee on the Ethics of Animal Experiments of Shandong Provincial Hospital affiliated to Shandong First Medical University, in compliance with the Guide for the Care and Use of Laboratory Animals published by the NIH.

    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.

    Funding

    The design, collection, analysis, and interpretation of the data in the study were financially supported by the Jinan Clinical Medical Science and Technology Innovation Plan (NO. 202328065), the Natural Science Foundation of Shandong Province (No. ZR2021QH307; No. ZR2021MH013), and the Shandong Province Major Scientific and Technical Innovation Project (No. 2021SFGC0502). The authors, their immediate families, and any research foundations with which they are affiliated have not received any financial payments or other benefits from any commercial entity related to the subject of this article.

    Disclosure

    The authors declare that they have no competing interests.

    References

    1. Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol. 2018;14(2):88–98. doi:10.1038/nrendo.2017.151

    2. Kahn SE, Cooper ME, Del Prato S. Pathophysiology and treatment of type 2 diabetes: perspectives on the past, present, and future. Lancet. 2014;383(9922):1068–1083. doi:10.1016/S0140-6736(13)62154-6

    3. Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature. 2014;505(7483):327–334. doi:10.1038/nature12984

    4. Liu T, Zhang L, Joo D, et al. NF-kappaB signaling in inflammation. Signal Transduct Target Ther. 2017;2(17023). doi:10.1038/sigtrans.2017.23

    5. Furman D, Campisi J, Verdin E, et al. Chronic inflammation in the etiology of disease across the life span. Nature Med. 2019;25(12):1822–1832. doi:10.1038/s41591-019-0675-0

    6. Kanter JE, Hsu CC, Bornfeldt KE. Monocytes and macrophages as protagonists in vascular complications of diabetes. Front. cardiovasc. med. 2020;7(10). doi:10.3389/fcvm.2020.00010

    7. Tripodo C, Sangaletti S, Piccaluga PP, et al. The bone marrow stroma in hematological neoplasms–a guilty bystander. Nat Rev Clin Oncol. 2011;8(8):456–466. doi:10.1038/nrclinonc.2011.31

    8. Omatsu Y. Cellular niches for hematopoietic stem cells in bone marrow under normal and malignant conditions. Inflamm regen. 2023;43(1):15. doi:10.1186/s41232-023-00267-5

    9. Wu PH, Joseph G, Saeed I, et al. Bone marrow adiposity alterations in type 2 diabetes are sex-specific and associated with serum lipid levels. J Bone Miner Res. 2023;38(12):1877–1884. doi:10.1002/jbmr.4931

    10. Hofbauer LC, Busse B, Eastell R, et al. Bone fragility in diabetes: novel concepts and clinical implications. Lancet Diabetes Endocrinol. 2022;10(3):207–220. doi:10.1016/S2213-8587(21)00347-8

    11. Shanbhogue M, Rosen DM, et al. Type 2 diabetes and the skeleton: new insights into sweet bones. Lancet Diabetes Endocrinol. 2016;4(2):159–173. doi:10.1016/S2213-8587(15)00283-1

    12. Oikawa A, Siragusa M, Quaini F, et al. Diabetes mellitus induces bone marrow microangiopathy. Arterioscler. Thromb. Vasc. Biol. 2010;30(3):498–508. doi:10.1161/ATVBAHA.109.200154

    13. Saito H, Gasser A, Bolamperti S, et al. TG-interacting factor 1 (Tgif1)-deficiency attenuates bone remodeling and blunts the anabolic response to parathyroid hormone. Nat Commun. 2019;10(1):1354. doi:10.1038/s41467-019-08778-x

    14. Saltiel AR, Olefsky JM. Inflammatory mechanisms linking obesity and metabolic disease. J Clin Invest. 2017;127(1):1–4. doi:10.1172/JCI92035

    15. Shaikh SR, Beck MA, Alwarawrah Y, et al. Emerging mechanisms of obesity-associated immune dysfunction. Nat Rev Endocrinol. 2024;20(3):136–148. doi:10.1038/s41574-023-00932-2

    16. Hrovatin K, Fischer DS, Theis FJ. Toward modeling metabolic state from single-cell transcriptomics. Mol Metabol. 2022;57(101396). doi:10.1016/j.molmet.2021.101396

    17. Jovic D, Liang X, Zeng H, et al. Single-cell RNA sequencing technologies and applications: a brief overview. Clin trans med. 2022;12(3):e694. doi:10.1002/ctm2.694

    18. Song F, Lee WD, Marmo T, et al. Osteoblast-intrinsic defect in glucose metabolism impairs bone formation in type II diabetic male mice. eLife. 2023;12. doi:10.7554/eLife.85714

    19. Zhong J, Mao X, Li H, et al. Single-cell RNA sequencing analysis reveals the relationship of bone marrow and osteopenia in STZ-induced type 1 diabetic mice. J Adv Res. 2022:41(145–158.doi: 10.1016/j.jare.2022.01.006.

    20. Liu CC, McCaffrey EF, Greenwald NF, et al. Multiplexed Ion Beam Imaging: insights into Pathobiology. Annu. Rev. Patholy. 2022;17:403–423. doi:10.1146/annurev-pathmechdis-030321-091459

    21. Efremova M, Vento-Tormo M, Teichmann SA, et al. CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nature Protocols. 2020;15(4):1484–1506. doi:10.1038/s41596-020-0292-x

    22. Zhang Q, Dong J, Zhang P, et al. Dynamics of transcription factors in three early phases of osteogenic, adipogenic, and chondrogenic differentiation determining the fate of bone marrow mesenchymal stem cells in rats. Front Cell Develop Biol. 2021;9(768316). doi:10.3389/fcell.2021.768316

    23. Liu F, Dong J, Zhou D, et al. Identification of key candidate genes related to inflammatory osteolysis associated with vitamin e-blended uhmwpe debris of orthopedic implants by integrated bioinformatics analysis and experimental confirmation. J Inflamm Res. 2021;14:3537–3554. doi:10.2147/JIR.S320839

    24. Liu F, Dong J, Zhang P, et al. Transcriptome sequencing reveals key genes in three early phases of osteogenic, adipogenic, and chondrogenic differentiation of bone marrow mesenchymal stem cells in rats. Front Mol Biosci. 2021;8(782054). doi:10.3389/fmolb.2021.782054

    25. Liu N, Dong J, Li L, et al. Novel clinical insights into the pathogenesis of posttraumatic elbow stiffness: an expression profile analysis of contracted joint capsule in human. J Inflamm Res. 2025;18:167–182. doi:10.2147/JIR.S499986

    26. Maiocchi SL, Ku J, Thai T, et al. Myeloperoxidase: a versatile mediator of endothelial dysfunction and therapeutic target during cardiovascular disease. Pharmacol Ther. 2021;221(107711). doi:10.1016/j.pharmthera.2020.107711

    27. Kim ET, Dybas JM, Kulej K, et al. Comparative proteomics identifies Schlafen 5 (SLFN5) as a herpes simplex virus restriction factor that suppresses viral transcription. Nature Microbiology. 2021;6(2):234–245. doi:10.1038/s41564-020-00826-3

    28. Uyar O, Dominguez JM, Bordeleau M, et al. Single-cell transcriptomics of the ventral posterolateral nucleus-enriched thalamic regions from HSV-1-infected mice reveal a novel microglia/microglia-like transcriptional response. J Neuroinflammation. 2022;19(1):81. doi:10.1186/s12974-022-02437-7

    29. Rocanin-Arjo A, Cohen W, Carcaillon L, et al. A meta-analysis of genome-wide association studies identifies ORM1 as a novel gene controlling thrombin generation potential. Blood. 2014;123(5):777–785. doi:10.1182/blood-2013-10-529628

    30. Belien J, Swinnen S, D’Hondt R, et al. CHIT1 at diagnosis predicts faster disability progression and reflects early microglial activation in multiple sclerosis. Nat Commun. 2024;15(1):5013. doi:10.1038/s41467-024-49312-y

    31. Miyake Y, Toyonaga K, Mori D, et al. C-type lectin MCL is an FcRgamma-coupled receptor that mediates the adjuvanticity of mycobacterial cord factor. Immunity. 2013;38(5):1050–1062. doi:10.1016/j.immuni.2013.03.010

    32. Liu N, Dong J, Li L, et al. Osteoimmune interactions and therapeutic potential of macrophage-derived small extracellular vesicles in bone-related diseases. Int J Nanomed. 2023;18:2163–2180. doi:10.2147/IJN.S403192

    33. Zhang Q, Sun W, Li T, et al. Polarization behavior of bone macrophage as well as associated osteoimmunity in glucocorticoid-induced osteonecrosis of the femoral head. J Inflamm Res. 2023;16:879–894. doi:10.2147/JIR.S401968

    34. Liu N, Dong J, Li L, et al. The FUNCTION and mechanism of anti-inflammatory factor metrnl prevents the progression of inflammatory-mediated pathological bone osteolytic diseases. J Inflamm Res. 2024;17:1607–1619. doi:10.2147/JIR.S455790

    35. Guo G, Liu Z, Yu J, et al. Neutrophil function conversion driven by immune switchpoint regulator against diabetes-related biofilm infections. Adv. Mater. 2024;36(8):e2310320. doi:10.1002/adma.202310320

    36. Umehara T, Mori R, Mace KA, et al. Identification of specific miRNAs in neutrophils of type 2 diabetic mice: overexpression of mirna-129-2-3p accelerates diabetic wound healing. Diabetes. 2019;68(3):617–630. doi:10.2337/db18-0313

    37. Wong SL, Demers M, Martinod K, et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nature Med. 2015;21(7):815–819. doi:10.1038/nm.3887

    38. Hughes MJ, McGettrick HM, Sapey E. Shared mechanisms of multimorbidity in COPD, atherosclerosis and type-2 diabetes: the neutrophil as a potential inflammatory target. Eur. respir. rev. 2020;29(155). doi:10.1183/16000617.0102-2019

    39. Minton K. B1 B cells link gut dysbiosis and insulin resistance. Nat Rev Immunol. 2019;19(1):1. doi:10.1038/s41577-018-0096-1

    40. DA W, Winer S, Shen L, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nature Med. 2011;17(5):610–617. doi:10.1038/nm.2353

    41. Harmon DB, Srikakulapu P, Kaplan JL, et al. Protective role for b-1b b cells and igm in obesity-associated inflammation, glucose intolerance, and insulin resistance. Arterioscler. Thromb. Vasc. Biol. 2016;36(4):682–691. doi:10.1161/ATVBAHA.116.307166

    42. Nishimura S, Manabe I, Takaki S, et al. Adipose natural regulatory b cells negatively control adipose tissue inflammation. Cell Metab. 2013;18(5):759–766. doi:10.1016/j.cmet.2013.09.017

    43. Luck H, Khan S, Kim JH, et al. Gut-associated IgA(+) immune cells regulate obesity-related insulin resistance. Nat Commun. 2019;10(1):3650. doi:10.1038/s41467-019-11370-y

    44. Ying W, Wollam J, Ofrecio JM, et al. Adipose tissue B2 cells promote insulin resistance through leukotriene LTB4/LTB4R1 signaling. J Clin Invest. 2017;127(3):1019–1030. doi:10.1172/JCI90350

    45. Julla JB, Girard D, Diedisheim M, et al. Blood monocyte phenotype is a marker of cardiovascular risk in type 2 diabetes. Circ. Res. 2024;134(2):189–202. doi:10.1161/CIRCRESAHA.123.322757

    46. Reddy MA, Amaram V, Das S, et al. lncRNA DRAIR is downregulated in diabetic monocytes and modulates the inflammatory phenotype via epigenetic mechanisms. JCI Insight. 2021;6(11). doi:10.1172/jci.insight.143289

    47. Junqueira C, Crespo A, Ranjbar S, et al. FcgammaR-mediated SARS-CoV-2 infection of monocytes activates inflammation. Nature. 2022;606(7914):576–584. doi:10.1038/s41586-022-04702-4

    48. Westcott DJ, Delproposto JB, Geletka LM, et al. MGL1 promotes adipose tissue inflammation and insulin resistance by regulating 7/4hi monocytes in obesity. J Exp Med. 2009;206(13):3143–3156. doi:10.1084/jem.20091333

    49. Wang H, Guo Z, Xu Y. Association of monocyte-lymphocyte ratio and proliferative diabetic retinopathy in the U.S. population with type 2 diabetes. J Transl Med. 2022;20(1):219. doi:10.1186/s12967-022-03425-4

    50. Kitade H, Sawamoto K, Nagashimada M, et al. CCR5 plays a critical role in obesity-induced adipose tissue inflammation and insulin resistance by regulating both macrophage recruitment and M1/M2 status. Diabetes. 2012;61(7):1680–1690. doi:10.2337/db11-1506

    51. Merad M, Martin JC. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat Rev Immunol. 2020;20(6):355–362. doi:10.1038/s41577-020-0331-4

    52. Petit V, Parcelier A, Mathe C, et al. TRIM33 deficiency in monocytes and macrophages impairs resolution of colonic inflammation. EBioMedicine. 2019;44:60–70. doi:10.1016/j.ebiom.2019.05.037

    53. Li P, Li K, Yuan W, et al. 1alpha,25(OH)(2)D(3) ameliorates insulin resistance by alleviating gammadelta T cell inflammation via enhancing fructose-1,6-bisphosphatase 1 expression. Theranostics. 2023;13(15):5290–5304. doi:10.7150/thno.84645

    54. Tao L, Liu H, Gong Y. Role and mechanism of the Th17/Treg cell balance in the development and progression of insulin resistance. Mol Cell Biochem. 2019;459(1–2):183–188. doi:10.1007/s11010-019-03561-4

    55. Bertola A, Ciucci T, Rousseau D, et al. Identification of adipose tissue dendritic cells correlated with obesity-associated insulin-resistance and inducing Th17 responses in mice and patients. Diabetes. 2012;61(9):2238–2247. doi:10.2337/db11-1274

    56. Savage TM, Fortson KT, de Los Santos-Alexis K, et al. Amphiregulin from regulatory T cells promotes liver fibrosis and insulin resistance in non-alcoholic steatohepatitis. Immunity. 2024;57(2):303–318e306. doi:10.1016/j.immuni.2024.01.009

    57. Li Y, Lu Y, Lin SH, et al. Insulin signaling establishes a developmental trajectory of adipose regulatory T cells. Nat Immunol. 2021;22(9):1175–1185. doi:10.1038/s41590-021-01010-3

    58. Moller SH, Wang L, Ho PC. Metabolic programming in dendritic cells tailors immune responses and homeostasis. Cell. Mol. Immunol. 2022;19(3):370–383. doi:10.1038/s41423-021-00753-1

    59. Ye Y, Gaugler B, Mohty M, et al. Plasmacytoid dendritic cell biology and its role in immune-mediated diseases. Clin. Transl. Immunol. 2020;9(5):e1139. doi:10.1002/cti2.1139

    60. Macdougall CE, Wood EG, Solomou A, et al. Constitutive activation of beta-catenin in conventional dendritic cells increases the insulin reserve to ameliorate the development of type 2 diabetes in mice. Diabetes. 2019;68(7):1473–1484. doi:10.2337/db18-1243

    61. Hernandez-Garcia E, Cueto FJ, Cook ECL, et al. Conventional type 1 dendritic cells protect against age-related adipose tissue dysfunction and obesity. Cell. Mol. Immunol. 2022;19(2):260–275. doi:10.1038/s41423-021-00812-7

    62. Kelly B, O’Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015;25(7):771–784. doi:10.1038/cr.2015.68

    63. Hanc P, Gonzalez RJ, Mazo IB, et al. Multimodal control of dendritic cell functions by nociceptors. Science. 2023;379(6639):eabm5658. doi:10.1126/science.abm5658

    64. Thimmappa PY, Vasishta S, Ganesh K, et al. Neutrophil (dys)function due to altered immuno-metabolic axis in type 2 diabetes: implications in combating infections. Human Cell. 2023;36(4):1265–1282. doi:10.1007/s13577-023-00905-7

    65. Yoshitaka T, Ishida S, Mukai T, et al. Etanercept administration to neonatal SH3BP2 knock-in cherubism mice prevents TNF-alpha-induced inflammation and bone loss. J Bone Miner Res. 2014;29(5):1170–1182. doi:10.1002/jbmr.2125

    66. Li J, Ayoub A, Xiu Y, et al. TGFbeta-induced degradation of TRAF3 in mesenchymal progenitor cells causes age-related osteoporosis. Nat Commun. 2019;10(1):2795. doi:10.1038/s41467-019-10677-0

    67. Li JY, D’Amelio P, Robinson J, et al. IL-17A Is increased in humans with primary hyperparathyroidism and mediates pth-induced bone loss in mice. Cell Metab. 2015;22(5):799–810. doi:10.1016/j.cmet.2015.09.012

    68. Yang Y, Ren D, Peng B, et al. Promotion of inflammatory response in mice with diabetes periodontitis: regulation of forkhead box protein M1 silencing to mediate activator protein-1 via reactive oxygen species production. CytoJournal. 2024;21(72). doi:10.25259/Cytojournal_143_2024

    69. Yang W, Wang Y, Mo K, et al. Single-cell RNA sequencing reveals multiple immune cell subpopulations promote the formation of abnormal bone microenvironment in osteoporosis. Sci Rep. 2024;14(1):29493. doi:10.1038/s41598-024-80993-z

    70. Sundararaghavan V, Mazur MM, Evans B, et al. Diabetes and bone health: latest evidence and clinical implications. Ther. Adv. Musculoskelet. Dis. 2017;9(3):67–74. doi:10.1177/1759720X16687480

    71. Huang J, Xiao Y, Zheng P, et al. Distinct neutrophil counts and functions in newly diagnosed type 1 diabetes, latent autoimmune diabetes in adults, and type 2 diabetes. Diabetes/Metab Res Rev. 2019;35(1):e3064. doi:10.1002/dmrr.3064

    Continue Reading

  • AI agents, tech circularity: What’s ahead for platforms in 2026

    AI agents, tech circularity: What’s ahead for platforms in 2026

    What you’ll learn: 

    Experts outlined four emerging trends that show where platforms are heading next: 

    • It’s time to adapt platforms for agentic AI. A new generation of autonomous “agents” can already buy, sell, and negotiate on behalf of their users — a shift that could transform how digital markets operate.
    • AI can accelerate technical debt.  As firms experiment with AI tools that write code, companies are discovering a hidden cost: flawed or poorly integrated code that can make systems harder to maintain and more expensive to fix.
    • The “AI stack” — the hardware and software used to build, train, and run AI models — is becoming tightly integrated and increasingly controlled by a handful of powerful companies. This concentration risks leaving most firms dependent on a few providers that are difficult to replace.
    • Platform strategies, long focused on digital services, are beginning to extend into the physical world by helping companies recover, resell, and reuse hardware as part of a broader circular economy.

    Digital platforms have already changed how value is created and exchanged. Their next wave — spanning physical assets, artificial intelligence, and automation — promises new efficiencies but also new risks.

    At the 2025 MIT Platform Strategy Summit, hosted by the MIT Initiative on the Digital Economy, experts highlighted four emerging trends that together offer a snapshot of where platforms are heading next. 

    Read the 2025 Platform Strategy Summit report 

    Platforms adapt for agentic AI 

    AI is now being programmed to act on its own. A new generation of autonomous “agents” can already buy, sell, and negotiate on behalf of their users — a shift that could transform how digital markets operate. 

    “You are going to launch a marketplace, you’re going to have to onboard and get critical mass for agents. You’re going to have to design interfaces for agents,” said Marshall Van Alstyne, SM ’91, PhD ’98. “You’re going to have to create value and take value with agents. You’re going to have to sell to agents.”

    Van Alstyne, a digital fellow at the MIT IDE and a Boston University professor, said these agents may soon make routine decisions without human involvement. This raises new questions about governance and oversight. 

    “This leads you to a learning-authority dilemma,” he said. “What happens when agent decision ability exceeds its formal authority?” 

    To prepare, Van Alstyne said, companies will need to adapt their platforms for AI interaction — creating interfaces agents can use, setting rules to manage agents’ behavior, and determining which decisions to automate and which to keep under human control.
     

    The hidden costs of AI become clearer 

    Generative AI is helping companies work faster and link data from different systems more easily, but it is also creating new kinds of technical problems.

    As firms experiment with AI tools that write code, for example, early studies are reporting big productivity gains. In reality, said Geoffrey Parker, SM ’93, PhD ’98, some of the world’s largest companies are discovering a hidden cost: piles of AI-generated code that does not work well in complex systems. Parker called this “turbocharged technical debt.”

    Parker, a Dartmouth College professor and digital fellow at the MIT IDE, explained that this buildup of flawed or poorly integrated code can make systems harder to maintain and more expensive to fix over time. 

    “The code assistance is certainly here,” he said, drawing on his recent research, which was published in MIT Sloan Management Review. “It’s valuable, but the tech debt is actually a strategic risk and could be really expensive, especially for incumbent organizations.”

    The AI stack becomes increasingly concentrated

    The “AI stack” — the hardware and software used to build, train, and run AI models — is becoming tightly integrated and increasingly controlled by a handful of powerful companies. This concentration risks leaving most firms dependent on a few providers that are difficult to replace, according to Lynn Wu, PhD ’11, an associate professor at the University of Pennsylvania.

    Wu noted that companies such as Google, Microsoft, and Amazon now dominate every layer of the AI ecosystem, from computing chips to cloud infrastructure to large language models. “When you have a vertically integrated stack, it’s great for efficiency,” she said. “But when they’re concentrated by a few players, you are also indebted to what they do.”

    She advised firms to be selective and deliberate when adopting AI tools, in addition to controlling their own data pipelines, focusing on areas with clear returns, and avoiding overreliance on third-party systems.

    Companies address their tech “circularity delta” 

    Platform strategies, long focused on digital services, are beginning to extend into the physical world by helping companies recover, resell, and reuse hardware as part of a broader circular economy. 

    According to Peter C. Evans, PhD ’05, senior adviser at Capital Growth Partners, the goal is to close what he calls the “circularity delta”: the gap between what firms spend on electronics and what they recover when those assets are retired. 

    Evans noted that most organizations still focus on acquiring new equipment while overlooking the value in what they replace — an oversight that leaves on the table an estimated $3 billion to $4 billion in recoverable value from laptops alone. 

    “There’s a lot of value in these assets,” he said, “but the companies aren’t actually capturing them.” 

    Evans argued that circularity works best when companies collaborate through platforms rather than acting alone. Platforms can make reuse and resale easier by lowering transaction costs, expanding participation, and creating network effects that help the circular economy grow.

    AI Executive Academy

    In person at MIT Sloan


    Peter Evans, Geoffrey Parker, and Marshall Van Alstyne are the cochairs of the MIT Platform Summit. 

    Peter Evans is the co-founder and managing partner of All Things Circular, a company based on circular economy transformation, and a senior adviser for enterprise strategy at investment firm Capitol Growth Partners. He is recognized for his leadership in circular platforms, reverse logistics, and digital transformation. 

    Geoffrey Parker is a professor of engineering at Dartmouth College, and a digital fellow and visiting scholar at the MIT IDE. His research areas include data analytics, platform economics and strategy, intellectual property, and product innovation. With Marshall Van Alstyne, he developed the theory of “two-sided markets.” 

    Marshall Van Alstyne is a professor in information systems at Boston University and a digital fellow at the MIT IDE. He conducts research on information economics, covering topics such as economics of speech markets, platform economics, intellectual property, and social effects of technology. 

    Lynn Wu is an associate professor at the Wharton School of the University of Pennsylvania. Her research focuses on the intersection of artificial intelligence, analytics, and innovation, exploring how these technologies reshape business strategy, productivity, and workforce dynamics. 

    Continue Reading

  • P300-Targeted Acetylome Reveals A Role For HMGB1 Translocation In Cent

    P300-Targeted Acetylome Reveals A Role For HMGB1 Translocation In Cent

    Introduction

    Cardiovascular diseases (CVD) are major global health burden worldwide,1,2 whereas myocardial infarction (MI) is the most severe form of CVD that accounts for the leading cause of death.2 Fatal ventricular arrhythmias (VAs) remain the primary causes of sudden cardiac death (SCD) among patients with acute MI.3 Sympathetic hyperactivation is one of the major causes of ventricular arrhythmogenesis.4–6 Investigation into possible interventions and the underlying mechanisms of VA post-MI has mainly focused on the cardiac sympathetic nerves, however, the brain-cardiac axes in the pathogenesis of sympathetic overactivation remain poorly understood. Accumulating evidence has suggested that central paraventricular nucleus (PVN) within the hypothalamus is the most important regulator of autonomic nervous tone and involves in cardiovascular homeostasis regulation. Recently, we found that microglia-mediated inflammation within the PVN exacerbated VAs by increasing sympathetic tone and ventricular arrhythmia susceptibility,7 yet the precise mechanism remains unclear.

    Protein post-translational modifications, including methylation, phosphorylation, and acetylation, are emerging as key mediators of cardiovascular pathogenesis following changes in immuno-microenvironment.8 Acetylation, the most common and conserved PTM, is important for protein function modification, including trans localization, binding partners interaction, conformation, catalytic activity, and stability. Lysine acetyltransferases (KAT), containing of MYST, GNAT and p300/CBP families, are responsible for histone and non-histone protein acetylation. It is evidenced that cardiovascular diseases change the acetylation status, participating various process, including cardiac remodeling and arrhythmia.9 Here, we explored the role of the acetyltransferases in the pathogenesis of sympathetic hyperactivity post-MI. In the present study, we found that among the acetyltransferases, p300 acetyltransferase (KAT3B) (HGNC: 3373) was specifically upregulated in PVN tissue post-MI. P300 is a large scaffold protein shuttling between the nucleus and cytoplasm following its N-terminal nuclear localization signals (NLS). Adeno-associated virus (AAV) vector carried short hairpin RNA (shRNA) was utilized for p300 silencing within PVN, to determine the role of p300 signaling in sympathoexcitation. Label-free proteomic technique combining enrichment identified for acetylated peptides downstream p300. We then conducted a series of bioinformatic analysis to classified p300 substrates into different pathways and identified the downstream target HMBG1. The current study will unveil the pathological process of central-cardiac sympathetic activation and provide new strategies for VAs therapy post-MI.

    Methods

    AMI Model

    Male Sprague-Dawley rats (~225 g; Beijing HFK Bioscience, China) were utilized in this study. All experimental procedures were performed in complied with the protocols approved by the Laboratory Animal Ethics Committee of the First Affiliated Hospital of Shandong First Medical University (QFSYYPZ2019022201). Open-chest surgery was conducted following 7 days adaption. Briefly, rats were anaesthetized by intraperitoneal injection of 3% pentobarbital sodium (30 mg/kg) and ventilated via tracheal intubations connected to a rodent ventilator as previously described.7 Following the heart exposure via incising along the left fourth intercostal space, a 6–0 polypropylene suture was carefully passed beneath the left anterior descending (LAD) coronary artery at the standard ligation site, approximately 2–3 mm distal to the junction between the conus arteriosus and left atrial appendage.10 The Sham group underwent all the surgery protocol without ligation on the heart. Then, the rats were allowed to recover on a heated pad to maintain a body temperature of 37°C after surgery. Finally, then they were returned to their individual cages. The rats were closely monitored at the first 24–72 h after surgery. Successful infarction was defined by observing the immediate ST elevation in electrocardiogram, regional wall motion abnormality and cyanosis were common criteria for infarction confirmation.11 The survival rate was 88% 3 h following the coronary ligation.

    Experimental Design

    Protocol 1

    Thirty of the 37 surviving rats were assigned to five groups (0, 1, 3, 5, and 7 days; n = 6/group). The expression of acetyltransferases was detected using real time-PCR), whereas the temporal expression pattern of p300 was detected by Western blotting.

    Protocol 2

    A total of 106 rats were randomly assigned to the following four groups to ensure an approximately equal number of survivors in each: Group A (sham + shCtrl; n = 20), Group B (sham + shp300; n = 20), Group C (MI + shCtrl; n = 21), and Group D (MI + shp300; n = 22). The virus was administered bilaterally to the PVN using a stereotaxic apparatus (RWD Life Science, China) following the three-dimensional coordinates (lateral: 0.4 mm, posterior: 1.8 mm, and 7.9 mm deep to the bregma).7 P300 knockdown was conducted using recombinant virus AAV1/2 vectors carrying p300 small hairpin RNAs (shp300) or scrambled shRNA (shCtrl) over-expressed under U6 promoter with co-expression of GFP as reporter under CMV promoter (Shanghai Genechem Co., China). To guarantee effective p300 silencing, three targeting sequences were selected: CCGGCACTGTGCATCTTCTCGACAATTCAAGAGATTGTCGAGAAGATGCACAGTGTTTTTG, CCGGAACAGTGGCACGAAGATATTATCAAGAGATAATATCTTCGTGCCACTGTTTTTTTG, and CCGGGAGCACGACTTACCAGATGAATTCAAGAGATTCATCTGGTAAGTCGTGCTCTTTTTG. Ultimately, the last one was selected. About 50 nl of either shp300 or a scrambled shCtrl (titer: 1.5 × 1012 genomic particles/mL) were separately administrated into PVN using microinjector.7 The microinjector was removed 15 min after injection. Four weeks later, the rats received MI surgery. On day 7 post-MI, the rats subject to Telazol intraperitoneally injection and isoflurane inhalation following the electrophysiological study.

    RNSA

    Renal sympathetic nerve activity (RSNA) serves as an indicator for peripheral sympathetic tone.12 Seven days after the MI surgery, the left renal nerve was dissected and carefully disassociated with surrounding tissue. Then, the left renal nerve was cut distally to eliminate the effect of afferent activity. The nerve was hooked using a pair of bipolar silver-wire recordings. A four-channel AC/DC differential amplifier (AD Instruments, Sydney, Australia) with a band-pass filter ranging from 100 to 3000 Hz was applied for signals amplification (time constant: 10ms; sampling frequency: 10 kHz).7 Background noise measurement was conducted at the end of each experiment.

    In vivo Electrophysiological Experiments

    Ventricular Arrhythmias (VAs) Susceptibility was conducted with Mapping lab, and ECG recording electrodes were placed on the left ear and right ventricle. Electrical stimulation (MappingLab, VCS-3001, 30 mV) was applied to the left ventricle of the heart. The stimulation protocol was applied by TTL generator software. In brief, eight S1 beats with a cycle length of 120 ms followed by one to three extrastimuli in shorter coupling intervals were applied. The programmed stimulation, VAs inducibility quotient qualified by VA scores were detailed described in published study.13

    Tissue Collection

    Following the electrophysiological study, the entire brain was then excised following decapitation. Brain and heart tissues were prepared for further analysis: i) The PVN was dissected from the brain tissue and transferred to −80°C storage immediately for subsequent molecular biology experiments. (2) For immunohistochemical study, the whole brain was fixed in 10% formalin and alternatively underwent paraffin section or continuing immersed in 15%, 30% sucrose for gradient dehydration until sinking completely to the bottom, and then embedded in Tissue-Tek® OCT compound and frozen on ice for 5 min before stored at –80°C for subsequent immunofluorescence analysis. (3) Additionally, the heart and peripheral blood were harvested. The middle section of hearts containing the infarcted myocardium was stored in 4% formalin and embedded in paraffin for Masson’s staining.

    Histological Test

    Following standard protocols, the infarct size was evaluated by Masson’s staining of heart cross section (Nanjing Jiancheng Technology, China).14 Planimetry was utilized to analyze digitized images, and the infarcted size was presented as the percentage of fibrosis area/total left ventricle area.

    Immunohistochemistry analysis was applied to assess central sympathetic activation via the Fos family. A mouse anti-c-Fos antibody (1:1000, Abcam, ab208942) was applied. The slides were incubated with goat anti-mouse (HRP) (1:200, Servicebio, G1214) and the DAB chromogenic kit (AIDACX Biotechnology, PNSJH-08). They were then counterstained with hematoxylin.

    Frozen tissue double/multiplex immunofluorescence staining was performed according to the protocol of a staining kit (Absin Bioscience Inc., abs50012). Briefly, following antigen retrieval with a rapid antigen retrieval solution and antigen blocking with QuickBlock™ Blocking Buffer (Beyotime, Nanjing, China), primary antibodies mouse anti-p300 (1:500, Abcam, ab275379), rabbit anti-Iba1 (1:500, Abcam, ab178846), rabbit anti-HMGB1 (1:250, Abcam, ab79823) and/or rabbit anti-NMDAR1 (1:200, ABclonal, A7677) was used. After incubated at room temperature for 2 h or 4°C overnight, the sections were washed and incubated with the secondary antibody in the kit for 40 min, then fluorescence amplification solution was used for 15 min each. Furthermore, DAPI solution (Abcam, ab104139) was used to identify the nuclei. Finally, fluorescence images were viewed and captured with a confocal microscope (Nikon, Japan). Ten random microscopic fields were selected and then analyzed using ImageJ software (version 1.38).

    RT-PCR

    Real time-PCR was performed to measure target mRNA levels.8 Briefly, total RNA was obtained by Trizol reagent and reverse transcribed into cDNA with cDNA Synthesis kit (Yeasen, China). RT-qPCR assay was performed using a SYBR qPCR Master Mix (Takara, Japan) in CFX96 Real-Time System. For quantification, GAPDH was utilized as a reference for sample normalization. Both the target gene and GAPDH were repeated in triplicate for each sample. A 2−ΔΔCT method was applied for relative quantification in comparison between groups. The list of PCR primers sequences is presented in Supplementary Table S1, which were designed by Primer-Blast tool in NCBI. We further confirmed primer specificity in by confirming that the amplification curve is a single exponential curve, and the dissolution curve is a single peak in the process of qPCR.

    Western Blot

    Cytoplasmic and nuclear proteins were separately extracted using the Nucleocytoplasmic Separation Kit (Beyotime, China; P0027). The following primary antibodies were utilized, with their respective dilutions: anti-HMGB1 rabbit pAb (1:10,000, Abcam, ab79823), anti-p300 rabbit pAb (1:1000, Abcam, ab275379), and acetylated lysine antibody (1:1000, Cell Signaling, #9441). β-tubulin and histone 3 served as internal references. The Ac-HMGB1 levels were referenced to total HMGB1 protein. Band intensity was quantified using scanning densitometry.

    Enzyme-Linked Immunosorbent Assay (ELISA)

    Myocardial tissue was minced by low temperature freezing grinder and suspended in perchloric acid (0.4 N) with reduced glutathione (5 mmol/L; pH 7.4). Then, the supernatants were collected following immediate centrifugation. Then, a commercial ELISA kit (USCN Life Science, LMC681Ra) was applied to measure cardiac norepinephrine concentration.

    Cell Preparation

    HCM3 and HEK293 cells were purchased from ATCC and cultured following the company’s instructions. In brief, RPMI-1640 medium and 10% FBS was used. For p300 siRNA transfection, the HMC3 cells were seeded in 6-well plates and nurtured to up to 30%–50% confluency. ShRNA directed against p300 (5’-GAGCACGACTTACCAGATGAA-3’) or scramble control (5’-TTCTCCGAACGTGTCACGT-3’) were transiently transfected into HMC3 cells, and INTERFERin®(Polyplus) transfection reagent was applied according to the instructed protocol. For plasmid transfection, the HEK293 cells (1×105 cells per well) were plated on 3.5 cm dish and cultivated to 70% confluence. Polyethylenimine (PEI, 25 kDa, Polysciences) 3 μg was added with 2 μg of plasmids into each well. Cellular analysis was conducted 48–72 h following siRNA treatment.

    Mass Spectrometry Analyses

    Acetylated Label-Free Method

    Protein samples were extracted and quantified using SDT Lysis Buffer and BCA Protein Assay Kit. Total proteins were reduced with DTT, and iodoacetamide was used to block the reduced cysteine residues. Subsequently, trypsin (Promega) was used for digestion and peptide mixture collection, followed by enrichment of Kac using the Acetyl-Lysine Motif Kit (Cell Signaling Technology, 13416S). Samples were separated using a Nano Elute (Bruker, Germany) system with a nanoliter flow rate connected to a Q Exactive HF-X mass spectrometer (Thermo Fisher Scientific) equipped with a CaptiveSpray ion source for mass spectrometry identification. The mass spectrometry data were searched using Maxquant software from the Uniprot_HomoSapiens_20386_20180905 database. A global false discovery rate cutoff of 0.01 was set for peptide and protein identification. Lysine acetylation sites with a fold change >2 or <0.5 and an uncorrected P-value (t-test) <0.05 were considered significantly different.

    Bioinformatics Analysis

    Gene Ontology (GO) Enrichment

    All protein sequences were aligned with the NCBI database, retaining sequences that ranked in the top 10 with an E-value ≤1e3. Subsequently, Blast2GO was used to complete the GO term annotation. InterProScan was utilized to conduct motif-based searches in the European Bioinformatics Institute database and augment functional information on the motif in proteins to improve annotation. Fisher’s exact test was used to compare the differentially expressed proteins and total proteins correlated using GO terms. Finally, the bubble plot in the enrichment plot package was used to show the GO enrichment results. The procedure above complied with the principles drafted in the Declaration of Helsinki.

    Screening Differentially Expressed Proteins

    Based on the protein expression profile, we conducted a cluster analysis of differentially expressed proteins using the heatmap package. The protein expression data were normalized for cluster analysis. The results are presented as a heat map.

    Expression Plasmids and shRNA

    The acetylation of NLS lysine is essential for protein cytoplasm translocation and release in immune cells.15 There are two NLSs identified, one located in the A box (aa 28–44) and the other in the B box (aa 179–185).16 To express these in HEK293 cells, we cloned complementary DNAs encoding wild-type HMGB1, HMGB1 A box, HMGB1 B box, point mutations of lysine K28/29/30/43/44 to arginine (R), and p300 into a plasmid cloning DNA vector.

    Coimmunoprecipitation

    HEK293 cells were harvested, washed with freezing phosphate buffered saline, and lysed on ice for 30 min with a lysis buffer (150 mM NaCl, 0.5% NaD OC, 50 mM Tris-HCl (pH 7.5), 1% NP40, 1 mM Na2EDTA, and 10% glycerol) containing protein inhibitors (cocktail, 1 mM Na3VO4, 10 mM NaF, and 1 mM PMSF). The supernatants were immunoprecipitated after centrifugation of the cell lysates at 4°C. This involves incubating with antibodies while rotating for 1 h at 4°C, followed by incubating with protein A/G-agarose beads for another 1 h, and subsequently washed the bead for 4 times with lysis buffer. Then, Western blotting and immunoprecipitation were conducted as described previously.17

    Patch Clamp Study

    Paraventricular Nucleus Slice Recordings

    Viable brain slices were obtained from animals that were transcardial perfused with CSF (glycerol based artificial cerebrospinal fluid). The CSF contained 252 mM glycerol, 1.2 mM NaH2PO4, 1.2 mM MgCl, 1.6 mM KCL, 2.4 mM CaCl2, 26 mM NaHCO3, 11 mM glucoses. Removed the brain carefully and sliced the brain into 300 μm thickness which contained paraventricular nucleus (PVN) neurons marked by the third ventricle. Transfer the brain slices to a solution comprising: 110 mM N-methyl D-glucamine, 1.2 mM NaH2PO4, 2.5 mM KCl, 25 mM NaHCO3, 110 mM HCl, 25 mM glucose, 10 mM MgSO4, and 0.5 mM CaCl2. Equilibrated the solution at 37°C for 15 minutes with 95% O2 and 5% CO2. Transfer the slices into a superfusion recording chamber with perfusion solution containing 125 mM NaCl, 2 mM CaCl2, 3 mM KCl, 5 mM glucose, 26 mM NaHCO3, and 5 mM HEPES. The perfusion solution was equilibrated with 5% CO2 and 95% O2 at 25°C. Equilibrate each slice in this perfusion solution for 30 minutes. Visualize the PVN Neurons with differential interference contrast optics and record the neurons with the prepared patch pipette. Patch pipettes, with the resistance of 5–7 MΩ, were filled with an intracellular solution (ICS) containing of 135 mM K-gluconic acid, 10 mM EGTA, 10 mM HEPES, 1 mM MgCl2, and 1 mM CaCl2. The PVN neurons were then patched and clamped in the voltage recordings mode with a holding potential of −70 mV using Axon 700B and pClamp 10.3 software (Axon Instruments). CNQX (50 mM) and APV (50 mM) were applied by the end of the experiment to block glutamatergic AMPA/kainate and NMDA receptors, respectively, which would confirm that the EPSCs were glutamatergic. The amplitudes and frequency spontaneous mEPSCs were analyzed to assess the differences of synaptic transmission in PVN fibers between groups.

    In vitro Neuron Recordings

    Disinfect the neonatal 24 h SD rats with the 75% alcohol spray. And soaked the rats in 75% alcohol for 3–5 min and then decapitated at the head/neck junction using the small surgical scissors. The paraventricular nucleus (PVN) was isolated following the previously described protocols.18 Briefly, separated the PVN, and placed the PVN in HBSS (Invitrogen, NY) enriched with glucose and sectioned into small pieces for enzymatic digestion. Collagenase II (2 mg/mL, 15–25 min) was used to digest PVN tissues at 37°C. After digestion, collect the isolated single PVN neurons by centrifugation at 1000 rpm/5 min. These cells were then seeded on coverslips and cultured for 7–10 days with 2% B27 serum in a neurobasal medium (Gibco).

    The patch recordings were applied on the PVN neurons. EPC10 patch-clamp amplifier was used in this experiment. Collected and analyzed the electrical signals using a Patchmaster system (HEKA Elektronik, Germany). Pull the recording pipettes of a resistance of 3–5 MΩ. The SF-77B fast perfusion (Warner) system was addressed to perfuse or deliver chemicals onto the cells with extracellular solution (ECS). The ECS consisted of: 130 mM NaCl, 2 mM MgCl2, 2 mM CaCl2, 5 mM KCl, 10 mM glucose, 10 mM HEPES, and 10 mM sucrose, pH = 7.4, and the osmolarity was 310. The pipettes solution consisted of: 115 mM Cs-MeSO3, 5 mM NaCl, 10 mM CsCl, 10 mM HEPES, 20 mM TEA, 0.6 mM EGTA, 0.3 mM Na3GTP, 4 mM Mg-ATP, and 10 QX-314 (pH = 7.2), and the osmolarity was 300. For NMDA receptor-mediated miniature EPSCs (NMDA-mEPSCs), MgCl2 was 0 and the ECS was supplemented with 10 μm CNQX, 50 μm picrotoxin, 1 μm strychnine, and 15 μm glycine. To record AMPA receptor-dependent miniature EPSCs (AMPA-mEPSCs), we supplemented the ECS with 2 mM MgCl2, 1 μM TTX, 50 μM picrotoxin, and 50 μM APV.19 The exogenous HMGB1 (40 nM) was added into the ECs when recording NMDA/AMPA-mEPSCs respectively. Hold the membrane potential at −70 mV unless otherwise specified. Filtered the recordings at 2 kHz and sampled at 5 kHz. Mini-analysis software was used to analyze the mEPSCs in this study.

    Statistical Analysis

    SPSS17.0 was used for statistical analysis. t test, analysis of variance (ANOVA) with post hoc tests were used to analyse the differences among groups. Quantitative results were shown as Mean ± SD. p < 0.05 was considered statistically different.

    Results

    P300 Was Especially Upregulated, Predominantly in Microglia in the PVN Following MI

    We examined lysine acetyltransferase (KAT) enzymes alterations using a real-time PCR array at day 3 post-MI to elucidate the acetylation regulation mechanism which may affect central sympathetic activation, since VAs events increased within 3 days of AMI onset as previously reported.20 Among the selected 33 KATs, only 22 were detected. P300 was the most significant upregulated acetyltransferase (Figure 1A). Western blot detection showed that p300 exhibited a slight upregulation after 1 day, was significantly overexpressed on day 3, and remained at a high level beyond day 7 (Figure 1B and C). Increased p300 immunoreactivity was noted within the PVN on AMI rats (Figure 1D and Figure S1). Interestingly, p300 was primarily co-stained with microglia marker Iba1 (Figure 1D–F). These data revealed the potential role of p300 in regulating microglia.

    Figure 1 Expression profile of the p300 in PVN post-MI. (A) A summary heatmap of quantitative RT-PCR analysis of acetyltransferase. n = 3 per group. (B and C) Western blot showing the expression profiles of p300 (300 kD) in PVN at 0 hr, 1-, 3-, 5-, and 7-days post‐MI. P300 was quantified relative to the β-tubulin (55 kD) levels. n = 4 per group and per time‐point. (D) Double‐immunostaining for Iba1 (green) and p300 (red) in the vehicle‐MI group shows a limited distribution of p300 on Iba1 in the PVN. Bar = 30 μm. (E and F) Qualification of Iba1 positive and p300 positive Iba1 cells in PVN, respectively n = 6 per group. T test and two-way analysis of variance (ANOVA) were applied for two groups and four groups analysis respectively. Each column with a bar represents the mean ± standard deviation. *P < 0.05 and **P < 0.01 vs 0 hr.

    Abbreviations: MI, myocardial infarction; PVN, paraventricular nucleus, RT-PCR, reverse transcription polymerase chain reaction, HPF: high power field.

    P300 Participated in the Pathogenesis of Sympathetic Overactivition Post-MI

    To uncover the role of p300 in the pathogenesis of MI, we constructed a AAV1/2-U6-p300/scramble shRNA-CMV-GFP and microinjected it into the PVN following protocol 2 (Figure 2A). GFP expression was detected with microscopic to confirm the specific distribution of the administered virus in the PVN (Figure 2B). The efficacy of the p300 knockdown was verified through Western blotting (Figure 2C and D). Furthermore, the morphology changes were analyzed on day 7 post MI (Table 1). Masson staining ruled out those less than 30% of infarct size due to lack of clinical significance. Our data revealed a reduced infarcted area in the MI‐shp300 group than those in the MI‐shCtrl group (P < 0.05), coinciding with ameliorating cardiac dysfunction as indicated by the index of ejection fraction (Figure 2E–H).

    Table 1 Cardiac Morphology at the End of the Study

    Figure 2 Targeted p300 knockdown in PVN. (A) Sketch map of targeting virus injection into PVN using stereotaxic apparatus. (B) Image confirming PVN targeted virus transfection into microglia. (C) Representative expression of p300 at 7 days post‐MI as determined via Western blotting. (D) P300 expression was quantified relative to the β-tubulin (55 kD) levels. (E) Representative original color images of Masson’s trichrome staining of the infarcted area: (i) sham group, (ii) sham + shCtrl, (iii) MI + shp300 and (iv) MI + shCtrl. (F) Infarction size calculation by Image J. (G) Representative images of cardiac function recorded by M-mode echocardiography. Ejection Fraction (EF) calculation (H) and fractional shortening (FS) calculation (I). n = 6 per group. Two-way ANOVA was applied for statistical analysis. Each column with a bar represents the mean ± SD. *P < 0.05, **P < 0.01 vs sham + shCtrl group; # P < 0.05 vs MI + shCtrl group.

    Abbreviations: MI, myocardial infarction; PVN, paraventricular nucleus.

    Next, the central and peripheral sympathetic function was investigated. Immunohistochemical staining of Fos protein served as an indicator of central sympathetic activation. As a result, MI triggered widespread neuronal activation significantly throughout the PVN evidenced by upregulation of Fos protein level. In contrast, administration of p300 shRNA significantly down-regulated Fos expression (Figure 3A and D). RSNA levels represented peripheral sympathetic tone was then evaluated. As shown Figure 3B, the baseline RSNA post infarction was significantly higher than that underwent sham surgery (P < 0.05). Additionally, RSNA levels were significantly attenuated in the MI + shp300 group compared to those in the MI + shCtrl group (P < 0.05). Consequently, reduced ventricular arrhythmias vulnerability with p300 knockdown mirrored those of RSNA content (Figure 3C–F). The increasing levels of cardiac NE (Figure 3G) post-MI, which reflecting cardiac sympathoexcitation, was reversed by p300 shRNA administration.

    Figure 3 Effects of p300 on sympathetic nerve activity and programmed electrical stimulation at 7 days post-MI. (A) Representative immunohistochemical images of central sympathetic nerve activity marked using Fos family proteins, indicated by brown puncta. Nuclei (blue) were stained with hematoxylin: (i) sham + shCtrl, (ii) sham + shp300, (iii) MI + shCtrl, and (iv) MI + shp300 groups (magnification 200×). Bar = 50 μm. (B) Typical recordings from the left RSNA and integrated RSNA at 7 days post-MI in (i) sham + shCtrl, (ii) sham + shp300, (iii) MI + shCtrl, and (iv) MI + shp300 groups. Bar = 1s. (C) Typical inducible ventricular arrhythmias. (D) Quantitation of Fos‐positive cells. (E) Quantification of baseline of RSNA. (F) Quantification of ventricular arrhythmia scores. (F) Comparisons of the arrhythmia scores across four groups. The vertical bar = 1mV, abscissa bar = 0.5s. (G) Myocardial NE levels detected by ELISA. n = 6 per group. Two-way ANOVA was applied for statistical analysis. Data are expressed as mean ± S.D. **P < 0.01 and *P < 0.05 vs the sham group. #P < 0.05 vs the MI + shCtrl groups.

    Abbreviations: MI, myocardial infarction; NE, norepinephrine; RSNA, renal sympathetic nerve activity; SD, standard deviation.

    Characterization of the p300-Dependent Acetylome

    To identify the specific substrates downstream p300 acetylation, we performed an unbiased screen through MS-based proteomics. This involves comparing acetylated proteins in scramble RNA transfected cells with those in cells where p300 has been silenced. We utilized two stable HMC3 cell lines in which doxycycline was used to induce the expression of control shRNA (NC) or shRNA targeting p300 (KC). The efficiency of the p300 knockdown was verified using Western blot analysis. We identified 135 differential acetylated protein between control and p300 knockdown group. The heatmap showing the top 12 differentially acetylated proteins (Figure 4A). GO enrichment analysis showed that p300 is involved in biological processes, mainly included cellular processes, cellular component organization, biological regulation or developmental processes, biogenesis, protein transport, response to stimulus, and immune system processes (Figure 4A and B). Examples of proteins acetylated by p300 enriching in the same biological processes according to GO term analysis were presented in Figure 4C and D. According to previous study, protein transport and release from microglia played an important role in microglia-neuron crosstalk.21 Therefore, we hypothesized that p300-mediated protein transport serves as a bridge between microglia and neural activation. Then, we focused on the protein transport related genes. Among them, HMGB1 was most significant differential protein as indicated in heatmap in Figure 4A. HMGB1 has been demonstrated as the key bridge between microglia-neuron communication,22 therefore, we tested whether HMGB1 is the downstream target for p300 mediated sympathoexcitation.

    Figure 4 Identification of p300-dependent acetylome. (A) (upper panel); (lower panel) Ice logo shows analysis of the frequency of amino acids surrounding the acetylated lysines targeted by p300. Overall, we analyzed 159 distinct “Ks” derived from a list of 135 proteins, with 10 aa upstream (−10 on the x-axis) and 10 aa downstream (+10 on the x-axis). The overall height of the stack indicates the sequence conservation at that position, while the symbol height within the stack indicates the relative frequency of each amino acid at that position. (B) Go enrichment analysis of proteins acetylated by p300 in biological process, cellular component and molecular function. (C) Based on GO enrichment analysis, the cnetplot of proteins acetylated by p300 suggests HMGB1 is involved in the protein transport process. (D) Motif logo map depicting p300 binding protein in NC and KD groups.

    Abbreviations: GO, gene ontology; HMGB1, high-mobility group protein B1.

    P300 Directly Induces Nucleus-Cytoplasm Translocation and Release of HMGB1 in Microglia Post-MI

    The nucleocytoplasmic translocation of HMGB1 was markedly increased in the PVN among MI rats; in contrast, acetyltransferase p300 silencing significantly partially reversed MI‐induced HMGB1 cytoplasmic translocation as showed in immunofluorescence staining (Figure 5A and D, E). Similar tendency in nucleocytoplasmic translocation and enhanced HMGB1 acetylation were found, as revealed through Western blot analysis (Figure 5B and C, F, G). Our finding further suggests that nonacetylation states correlate with nuclear retention, whereas p300-mediated acetylation promotes cytosolic trans localization as indicated by immunofluorescence and Western blot.

    Figure 5 The effect of p300 shRNA intervention on HMGB1 levels, intracellular distribution, and acetylation in the PVN of MI rats. (A) Immunofluorescence costaining for HMGB1 (cyan) with Iba1 (green) and p300 (red) of PVN 7 days after MI. (B) Representative Western blot illustrating cytoplasm/nucleus/acetylation HMGB1 protein. (C) Qualification of acylation level of HMGB1. (D and E) Qualification of p300 positive cells and nucleus/cytoplasm HMGB1 percentage in PVN, respectively. (F and G) Qualification of cytoplasm and nucleus HMGB1 level, respectively. Bar = 30 μm. n = 6 per group. Two-way ANOVA was applied for statistical analysis. Values are expressed as the mean ± SD. *P < 0.05 and **P < 0.01 vs the sham group; # #P < 0.01, #P < 0.05 vs the MI + shCtrl groups.

    Abbreviations: HMGB1, high-mobility group protein B1; MI, myocardial infarction; PVN, paraventricular nucleus; HPF: high power field.

    P300 Directly Interacts with HMGB1 with K43 the Key Acetylated Lysine Site

    Full-length HMGB1 contains two NLSs located in the HMGB1-A box (aa 28–44) and HMGB1-B box (aa 179–185), which guide the protein translocate into the nucleus. These lysine residues are conserved in HMGB1 across species, from Drosophila to humans (Figure S2), suggesting their functional relevance. To figure out the specific region of HMGB1 which undergoes acetylation by p300, we conducted co-transfection using truncation mutants Flag-tagged HMGB1 or wild-type HMGB1 plasmids with myc-tagged p300 into HEK293T cells. Subsequently, they were subjected to reciprocal coimmunoprecipitation (CO-IP) experiments, revealing that p300 acetylates HMGB1 with the A box (Figure 6A). Subsequently, we generated flag-tagged acetylation-mimicking non-acetylated (K28R, K29R, K30R, K43R, and K44R) mutants of HMGB1. CO-IP analysis revealed that the point mutation at lysine 43 to arginine significantly reduced the binding affinity between p300 and HMGB1 (Figure 6B). Following this, we examined the effects of p300 on HMGB1 translocation by immunofluorescence confocal imaging and found that HMGB1K43R mutants blocked HMGB1 cytoplasmic translocation and acetylation (Figure 6C–F). Eventually, ELISA analysis showed that K43R mutants consequently attenuating HMGB1 release level (Figure 6G).

    Figure 6 P300 acetylates HMGB1 at its K43 residue. (A) Co-transfection of Myc/Flag-tagged p300 into HEK293T cells with Flag-tagged HMGB1-A box or Flag-tagged HMGB1-B box. Immunoprecipitation was performed using an anti-Myc antibody, and Western blots were analyzed using the indicated antibodies. Cell lysates were immunoprecipitated with an anti-Myc antibody and subsequently analyzed using Western blotting. (B) Co-transfection of Myc-tagged p300 with HMGB1 truncation mutants or wild-type HMGB1 into HEK293T cells in the presence of HDAC inhibitors. Immunoprecipitation was performed using an anti-myc antibody and then analyzed using the specified antibodies. (C) Immunostaining for HMGB1 (green) and DAPI (blue) in the p300 /HMGB1 coinfection group (upper) and the p300/HMGB1 K43R mutant coinfection group (lower). Bar = 10 μm. (D) Representative Western blot illustrating cytoplasm/nucleus HMGB1 protein. (E and F) Qualification of cytoplasm and nucleus HMGB1 level, respectively. (G) ELISA for detecting HMGB1 content in cell culture supernatant in the p300 /HMGB1 coinfection group (upper panel) and p300/HMGB1 K43R mutant coinfection group. n = 4 per group. T test was applied for statistical analysis. **P < 0.01 vs the WT group.

    Abbreviations: ELISA, enzyme-linked immunosorbent assay; HMGB1, high-mobility group protein B1; WT, wild type.

    Increased NMDAR Activity in Presympathetic Neurons Through the p300-HMGB1 Axis

    Next, we sought to resolve whether p300 mediated HMGB1 traslocation participated in postsynaptic excitation. Enhanced glutamatergic input in PVN, especially the NMDAR activity, is a major source of the excitatory drive to sympathetic tone. Interestingly, despite of the functional variation of glutamate receptors, the protein level of NMDAR kept unchanged (Figure 7A–C). However, the amplitude and frequency of basal glutamatergic excitatory postsynaptic currents (mEPSCs) of PVN neurons was significantly greater in MI group than in sham group. Consistently, p300 knockdown significantly reversed the attenuated NMDAR-mEPSC (Figure 7D–G).

    Figure 7 P300 knockdown diminished the increase in NMDA-mEPSCs post-MI. (A) Western blot showing the expression profiles of GluN1 (105kD) and GLuN2B (166kD) in PVN at day-7 post‐MI. GluN1 and GluN2B expression were quantified relative to the β-tubulin (55 kD) levels. (B and C) Statistical bar graph of relative GluN1 and GLuN2B expression in Sham+shCtrl, Sham+shP300, MI+shCtrl, and MI+shP300 groups. (D) Representative single mEPSC currents of brain slices from Sham+shCtrl, Sham+shP300, MI+shCtrl, and MI+shP300 groups. (E) Statistical bar graph of normalized NMDAR-mEPSC current amplitude in the four groups. (F and G) Statistical bar graph of normalized NMDAR-mEPSC current frequency and interevent intervals in the four groups. n = 5 per group. *P < 0.05 vs the Sham+shCtrl group; # P <0.05 vs the MI+shCtrl group.

    Given the above evidence highlighting the critical role of macrophage-derived HMGB1 in central sympathetic activation, we assumed that HMGB1 may influence excitatory glutamate receptors function directly. Consistently, immunofluorescence experiments revealed colocalization of HMGB1 and NMDARs in primary PVN neurons post-MI (Figure 8A). Molecular docking using Discovery Studio 2016 software to predict the potential binding affinity between HMGB1 and NMDAR subunits. As a result, a strong binding affinity between HMGB1 and GluN1 and GluN2B subunits were found (highest Zdock score: 22.1 and 20.9, respectively; Figure 8B). Patch-clamping study further revealed that HMGB1 significantly enhanced both the amplitude and current numbers of mEPSCs in PVN neurons without affecting the half-width of mEPSCs (Figure 8C and D). To define the precise role of NMDARs in HMGB1-induced firing hyperactivity, we applied intracellular dialysis with an NMDAR channel specific blocker MK-801 to block NMDARs. When neurons were pretreated with the MK-801, the substantial increase in EPSCs was significantly decreased. Furthermore, HMGB1 induced a marked increase in NMDA puff-elicited currents, but it did not affect AMPA-mEPSCs in PVN neurons (Figure 8E–G). This indicates a direct role for HMGB1 in NMDAR-mediated neuronal activation.

    Figure 8 HMGB1 promotes NMDA-mEPSCs in PVN neurons. (A) Double‐immunostaining for HMGB1 (green) and NMDAR1 (red) in the vehicle‐MI group. (B) Quantification of HMGB1 positive NMDAR1 cells in the PVN. Bar = 50 μm. n = 6 per group. **P < 0.05 vs sham group. (C) The structural diagram illustrates the optimal docking between the HMGB1 ligand (purple) and NMDAR receptor molecule (yellow band:GluN1, the other is GluN2B) is as follows: (a) The enlarged docking site of the optimal combination; (b) Receptor residues bound in the optimal structural diagram (overall, 43 amino acid residues); (c) Surface interaction in the optimal docking structure diagram, with blue representing positively charged surface areas, red indicating negatively charged surface areas, and green representing hydrophobic surface areas. (D) Recordings of representative spontaneous mEPSCs from PVN neurons before and after HMGB1 protein application via voltage-clamp in whole-cell recording mode, with a membrane potential held at −70 mV. (E) The statistical bar graph shows that HMGB1 significantly increased the amplitude and current numbers of mEPSCs without affecting the half-width of the mEPSCs in PVN neurons. (F) Representative NMDA-mEPSCs and AMPA-mEPSCs in response to HMGB1 and NMDA/AMPA antagonist MK-801 (10 µM)/CNQX (10 µM). (G) The upper panel shows the representative distribution of mEPSCs amplitude and frequency before (gray color) and after (red color) HMGB1 trigger. The lower panel shows the normalized cumulative distribution analysis of NMDA-mEPSCs amplitude and frequency in the same neurons as in (D), shows that HMGB1 induced a significant shift toward higher amplitude (*P < 0.05) and decreased interevent interval (frequency) distribution (*P < 0.05). HMGB1 did not affect AMPA-mEPSCs in PVN neurons. n = 5 per group. T test was applied for statistical analysis.

    Abbreviations: AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor-dependent; HMGB1, high-mobility group protein B1; mEPSCs, miniature excitatory postsynaptic currents; NMDAR, N-methyl-D-aspartate receptor; PVN, paraventricular nucleus; SD, standard deviation.

    Discussion

    The conception of brain-spleen and gut-brain axes had been raised and studied in detail;23,24 however, the brain-cardiac axes remained poorly understood. The PVN plays a crucial role in regulating cardiovascular activity and sympathetic outflow, serving as an important interface between peripheral inflammation and the central nervous system mechanisms responsible for regulating cardiac sympathetic activity.7 In the current study, we identified the role of acetylase p300 on central-cardiac sympathetic hyperactivity post-MI. Specifically, p300 mediated immuno-neuron communication via promoting microglia HMGB1 nuclear-cytoplasm translocation; then translocated HMGB1 further combined with preganglionic NMDAR and facilitates sympathetic activation. This information has the potential to improve the current knowledge and update education surrounding brain-cardiac axes in the pathogenesis of post-MI arrhythmia, as well as microglia-neuron crosstalk in sympathoexcitation.

    Cardiac autonomic imbalance by overactivation of central sympathetic outflow in PVN connects tightly with arrhythmogenesis post-MI.25,26 Inflammatory stress is acknowledged as the master regulator of numerous pathological processes that lead to sympathetic activation, with the underlying mechanism remained poorly understood.27 Accumulating evidence indicates that, alongside genetic factors, epigenetic mechanisms transducing the effects of gene-microenvironment interactions contribute to inflammation-mediated pathophysiological processes following injury, such as MI and stroke.28 We explored the epigenetic mechanisms underlying acetylation during central sympathetic nervous system activation.

    KATs are well-known for targeting Lys sites on N-terminal tails of histone, whereas acetyltransferase p300, the most widely studied KAT, has long been shown to acetylate many Lys residues in non-histone proteins and modifies the stability, localization, and DNA-binding capability of the latter.29,30 Specifically, p300 plays critical roles in regulating inflammation response by transporting of proteins involved in inflammation.17 Accordingly, we observed a significant increase in the expression of p300, specifically within the microglia of the PVN post-MI, as opposed to other acetyltransferases. Simultaneously, targeting delivery of p300 shRNA to PVN ameliorated sympathetic overactivation and ventricular arrhythmia. This supports the presence of a novel epigenetic mechanism underlying inflammation-related sympathetic activation. Next, we aimed to sought out the downstream target of p300 from HCM3 cell lines by label free acetylation omics mass spectrometry study. We found 135 differential acetylated protein involved in various biological process, such as biogenesis, cellular component organization or developmental processes, protein transport, immune system processes et al. Given that microglia influence sympathetic activation through cell-to-cell interactions, whether autocrine or paracrine, it is crucial to consider the potential connection between p300-mediated acetylation of histones and non-histone proteins within microglia at the regulatory site level. Eventually, we confirmed the top differential expressed protein-HMGB1 which enriched in protein transport process as the potential target. Interestingly, one of our previous studies conducted PVN targeting administration of anti-HMGB1 polyclonal antibody and attenuated central-cardiac sympathetic tone in rats after MI.31 Whether p300 mediated sympathetic hyperactivation through lysine acetylation of HMGB1?

    HMGB1 is considered as an important endogenous peptides participating in microglia-mediated central neuroinflammation by actively secreted from activated immune cells and functions as a proclamatory cytokine,32 acting as a mediator in the crosstalk between microglia/macrophages and neurons in an autocrine or paracrine manner.33 The mechanism of HMGB1 releasing into the extracellular space requires the transportation from the nucleus to cytoplasm. In macrophages, the equilibrium between acetylation and deacetylation of HMGB1 regulates the HMGB1 cytoplasm translocation. Nuclear transport of proteins larger than 45 kDa, such as HMGB1, requires NLS for nuclear import.34 Reports on the primary deacetylated lysine sites of HMGB1 vary between K28, K30, K90, and K177. We found that the lysine acetylation site of HMGB1 by p300 is K43 evidenced by the fact that K43R mutation efficiently deacetylated HMGB1 and reversed its cytoplasm translocation. Consistently, in vivo study further showed that p300 silencing reversed HMGB1 acetylation and translocation. Next, we explored how this reversal occurred as it blocked the translocation process, which serves as a messenger mediating cell-to-cell contact between microglia and neurons. The downstream mechanism of this reversal remains unclear.

    NMDARs and AMPARs in the hypothalamic PVN are critical in regulating sympathetic outflow.35 We found no significant changes in expression of glutamate receptor. Whereas the increase of NMDAR-mEPSCs was observed as the mainly driver of sympathetic activation. Inhibition of p300 profoundly decreases NMDAR-mediated mEPSCs in PVN post-MI without affecting the expression of glutamate receptors. Therefore, we tested whether HMGB1 directly or indirectly facilitates NMDAR current. Through conducting molecular docking to estimate the likely combination of HMGB1 and glutamate NMDAR using Discovery Studio software. We also found significant increase in co-staining of HMGB1 and NMDAR in PVN. Patch clamp study proofed he direct interaction of HMGB1 with NMDAR could further increase the NMDA current, thus facilitating postsynaptic sympathetic activation, which may explain the brain-cardiac axis of inflammation and sympathetic activation post-MI.

    In addition, non-HMGB1 acetylation targets (eg, histones, transcription factors) that may also involved in p300-mediated modulation of sympathetic signaling. NF-κB, a key mediator of neuroimmune inflammatory responses, was identified as playing a pivotal role in both central and peripheral sympathetic excitation post-MI.36 Specifically, p300 appears to be crucial in the regulation of NF-κB, either by interacting directly with NF-κB or by acetylation of H3K9 which facilitates NF-κB recruitment to the promoters of proinflammatory genes, such as IL-6, IL-8 and cyclooxygenase-2 (COX-2).37 These alternative mechanisms will be further investigated in our future studies.

    Clinical Implications

    The epigenetics modification in the PVN evidenced for targeting central sympathetic activation in the treatment of ventricular arrhythmias refractory to conventional therapies. Sympathetic activity modification targeting PVN focusing on p300 blockage or HMGB1 translocation may hold promise for improving outcomes and reducing the risk of SCD in this patient population. The early discoveries could be expanded to application for sympathetic storm associated arrhythmia in clinical work.

    Limitation

    First, extrapolating data from rats rather than humans has potential limitations. For example, the AMI rat model differs from that of natural myocardial ischemia. Second, we did not investigate dominant-negative mutations in specific HMGB1 acetylation sites in vivo owing to technical limitations. Third, since microglia tended to resist AAV transduction, lack of viral efficiency and specificity to microglia in vivo was observed. In future study, we planned to improve transduction efficiency by AAV vectors driven under Iba promoter or capsid-modified AAV variants. Last but not the least, we did not investigate whether p300 knockdown affected other functions of p300 in the inflammatory reaction. Nonetheless, the outcomes of the current study provided therapeutic potential of central p300 signaling in confronting life-threatening ventricular arrhythmias following acute MI.

    Conclusion

    Our study suggested that p300-mediated translocation of HMGB1 may be a fundamental epigenetic mechanism in NMDAR-mediated central sympathetic activation post-MI. This finding provides new insights into neuroimmune crosstalk and brain-cardiac communication. Based on current findings, we aim to further develop site-specific deacetylation techniques for HMGB1, thus enabling precise control over sympathetic activation following MI.

    Funding

    National Natural Science Foundation of China (81900296, 82200362, 82300354, 32400994), the Higher Education Youth Innovation Team Plan of Shandong Province (2022KJ191), the Natural Science Foundation of Shandong Province (ZR2020MH024), the cultivation Fund for the First Affiliated Hospital of Shandong First Medical University (QYPY2021NSFC0613) and Shandong Provincial Key Discipline Construction Project of Traditional Chinese Medicine.

    Disclosure

    The authors report no conflicts of interest in this work.

    References

    1. Li T, Mu N, Yin Y, et al. Targeting AMP-activated protein kinase in aging-related cardiovascular diseases. Aging Dis. 2020;11(4):967–977. doi:10.14336/AD.2019.0901

    2. Jiang S, Yang Y, Li T, et al. An overview of the mechanisms and novel roles of Nrf2 in cardiovascular diseases. Expert Opin Ther Targets. 2016;20(12):1413–1424. doi:10.1080/14728222.2016.1250887

    3. Yu JK, Liang JA, Franceschi WH, et al. Assessment of arrhythmia mechanism and burden of the infarcted ventricles following remuscularization with pluripotent stem cell-derived cardiomyocyte patches using patient-derived models. Cardiovasc Res. 2022;118(5):1247–1261. doi:10.1093/cvr/cvab140

    4. Shi Y, Li Y, Yin J, et al. A novel sympathetic neuronal GABAergic signalling system regulates NE release to prevent ventricular arrhythmias after acute myocardial infarction. Acta Physiologica. 2019;227(2):e13315. doi:10.1111/apha.13315

    5. Kluge N, Dacey M, Hadaya J, et al. Rapid measurement of cardiac neuropeptide dynamics by capacitive immunoprobe in the porcine heart. Am J Physiol Heart Circulatory Physiol. 2021;320(1):H66–h76. doi:10.1152/ajpheart.00674.2020

    6. Yin J, Wang Y, Hu H, et al. P2X(7) receptor inhibition attenuated sympathetic nerve sprouting after myocardial infarction via the NLRP3/IL-1β pathway. J Cell Mol Med. 2017;21(11):2695–2710. doi:10.1111/jcmm.13185

    7. Wang Y, Hu H, Yin J, et al. TLR4 participates in sympathetic hyperactivity Post-MI in the PVN by regulating NF-κB pathway and ROS production. Redox Biol. 2019;24:101186. doi:10.1016/j.redox.2019.101186

    8. Sapp RM, Chesney CA, Springer CB, et al. Race-specific changes in endothelial inflammation and microRNA in response to an acute inflammatory stimulus. Am J Physiol Heart Circ Physiol. 2021;320(6):H2371–H2384. doi:10.1152/ajpheart.00991.2020

    9. Yang M, Zhang Y, Ren J. Acetylation in cardiovascular diseases: molecular mechanisms and clinical implications. Biochim Biophys Acta Mol Basis Dis. 2020;1866(10):165836. doi:10.1016/j.bbadis.2020.165836

    10. Mohanraj R, Hu H, Xuan Y, et al. Targeted NGF siRNA delivery attenuates sympathetic nerve sprouting and deteriorates cardiac dysfunction in rats with myocardial infarction. PLoS One. 2014;9(4):e95106.

    11. Zhang Y, Cai Z, Shen Y, et al. Hydrogel-load exosomes derived from dendritic cells improve cardiac function via Treg cells and the polarization of macrophages following myocardial infarction. J Nanobiotechnol. 2021;19(1):271. doi:10.1186/s12951-021-01016-x

    12. Geng Z, Ye C, Tong Y, et al. Exacerbated pressor and sympathoexcitatory effects of central Elabela in spontaneously hypertensive rats. Am J Physiol Heart Circulatory Physiol. 2020;318(1):H124–h134. doi:10.1152/ajpheart.00449.2019

    13. Kim YG, Han S, Choi JI, et al. Impact of persistent left superior vena cava on radiofrequency catheter ablation in patients with atrial fibrillation. Europace. 2019;21(12):1824–1832. doi:10.1093/europace/euz254

    14. Yin J, Hu H, Li X, et al. Inhibition of Notch signaling pathway attenuates sympathetic hyperinnervation together with the augmentation of M2 macrophages in rats post-myocardial infarction. Am J Physiol Cell Physiol. 2016;310(1):C41–C53. doi:10.1152/ajpcell.00163.2015

    15. Venereau E, De Leo F, Mezzapelle R, et al. HMGB1 as biomarker and drug target. Pharmacol Res. 2016;111:534–544. doi:10.1016/j.phrs.2016.06.031

    16. Lu B, Antoine DJ, Kwan K, et al. JAK/STAT1 signaling promotes HMGB1 hyperacetylation and nuclear translocation. Proc Natl Acad Sci USA. 2014;111(8):3068–3073. doi:10.1073/pnas.1316925111

    17. Wang X, Yang Y, Ren D, et al. JQ1, a bromodomain inhibitor, suppresses Th17 effectors by blocking p300-mediated acetylation of RORγt. Br J Pharmacol. 2020;177(13):2959–2973. doi:10.1111/bph.15023

    18. Kuwahara S, Arima H, Banno R, et al. Regulation of vasopressin gene expression by cAMP and glucocorticoids in parvocellular neurons of the paraventricular nucleus in rat hypothalamic organotypic cultures. J Neurosci. 2003;23(32):10231–10237. doi:10.1523/JNEUROSCI.23-32-10231.2003

    19. Ramirez DMO, Crawford DC, Chanaday NL, et al. Loss of Doc2-dependent spontaneous neurotransmission augments glutamatergic synaptic strength. J Neurosci. 2017;37(26):6224–6230. doi:10.1523/JNEUROSCI.0418-17.2017

    20. Bhar-Amato J, Davies W, Agarwal S. Ventricular arrhythmia after acute myocardial infarction: ‘the perfect storm’. Arrhythm Electrophysiol Rev. 2017;6(3):134–139. doi:10.15420/aer.2017.24.1

    21. Marinelli S, Basilico B, Marrone MC, et al. Microglia-neuron crosstalk: signaling mechanism and control of synaptic transmission. Semin Cell Dev Biol. 2019;94:138–151. doi:10.1016/j.semcdb.2019.05.017

    22. Paudel YN, Angelopoulou E, Piperi C, et al. Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s disease (AD): from risk factors to therapeutic targeting. Cells. 2020;9(2):383. doi:10.3390/cells9020383

    23. Zhang X, Lei B, Yuan Y, et al. Brain control of humoral immune responses amenable to behavioural modulation. Nature. 2020;581(7807):204–208. doi:10.1038/s41586-020-2235-7

    24. Cryan JF, O’Riordan KJ, Cowan CSM, et al. The microbiota-gut-brain axis. Physiol Rev. 2019;99(4):1877–2013. doi:10.1152/physrev.00018.2018

    25. Hu H, Wu H, Zhu T, et al. Long-term transcranial ultrasound stimulation regulates neuroinflammation to ameliorate post-myocardial infarction cardiac arrhythmia and remodeling. Heart Rhythm. 2024.

    26. Chen J, Yin D, He X, et al. Modulation of activated astrocytes in the hypothalamus paraventricular nucleus to prevent ventricular arrhythmia complicating acute myocardial infarction. Int J Cardiol. 2020;308:33–41. doi:10.1016/j.ijcard.2020.01.035

    27. Zhu Y, Wang K, Ma Z, et al. SIRT1 activation by butein attenuates sepsis-induced brain injury in mice subjected to cecal ligation and puncture via alleviating inflammatory and oxidative stress. Toxicol Appl Pharmacol. 2019;363:34–46. doi:10.1016/j.taap.2018.10.013

    28. Placek K, Schultze JL, Aschenbrenner AC. Epigenetic reprogramming of immune cells in injury, repair, and resolution. J Clin Invest. 2019;129(8):2994–3005. doi:10.1172/JCI124619

    29. Ding HH, Zhang SB, Lv YY, et al. TNF-alpha/STAT3 pathway epigenetically upregulates Nav1.6 expression in DRG and contributes to neuropathic pain induced by L5-VRT. J Neuroinflammation. 2019;16(1):29. doi:10.1186/s12974-019-1421-8

    30. Whedon SD, Cole PA. KATs off: biomedical insights from lysine acetyltransferase inhibitors. Curr Opin Chem Biol. 2023;72:102255. doi:10.1016/j.cbpa.2022.102255

    31. Li P, Jie Y, YuGen S, et al. High mobility group box-1 in hypothalamic paraventricular nuclei attenuates sympathetic tone in rats at post-myocardial infarction. Cardiol J. 2019;26(5):555–563. doi:10.5603/CJ.a2018.0117

    32. Zhang S, Hu L, Jiang J, et al. HMGB1/RAGE axis mediates stress-induced RVLM neuroinflammation in mice via impairing mitophagy flux in microglia. J Neuroinflammation. 2020;17(1):15. doi:10.1186/s12974-019-1673-3

    33. Sekiguchi F, Domoto R, Nakashima K, et al. Paclitaxel-induced HMGB1 release from macrophages and its implication for peripheral neuropathy in mice: evidence for a neuroimmune crosstalk. Neuropharmacology. 2018;141:201–213. doi:10.1016/j.neuropharm.2018.08.040

    34. Wang M, Gauthier A, Daley L, et al. The role of HMGB1, a nuclear damage-associated molecular pattern molecule, in the pathogenesis of lung diseases. Antioxid Redox Signaling. 2019;31(13):954–993. doi:10.1089/ars.2019.7818

    35. Li Y, Lu YX, Chi HL, et al. Chronic blockade of NMDAR Subunit 2A in the hypothalamic paraventricular nucleus alleviates hypertension through suppression of MEK/ERK/CREB pathway. Am J Hypertens. 2021;34(8):840–850. doi:10.1093/ajh/hpab047

    36. Qi L, Wang Y, Hu H, et al. m6A methyltransferase METTL3 participated in sympathetic neural remodeling post-MI via the TRAF6/NF-?B pathway and ROS production. J Mol Cell Cardiol. 2022;170:87–99. doi:10.1016/j.yjmcc.2022.06.004

    37. Di Pietrantonio N, Di Tomo P, Mandatori D, et al. Diabetes and its cardiovascular complications: potential role of the acetyltransferase p300. Cells-Basel. 2023;12(3):431.

    Continue Reading

  • With Grokipedia, Top-Down Control of Knowledge Is New Again

    With Grokipedia, Top-Down Control of Knowledge Is New Again

    Grokipedia, the AI-generated encyclopedia owned by Elon Musk’s xAI, went live on October 27. It is positioned as, first and foremost, an ideological foil to Wikipedia, which for years has been the subject of escalating criticism by right-wing media in general and Musk in particular. With Grokipedia, Musk wants to produce something he sees as more neutral.

    Much has already been written about the character of Grokipedia’s content. This essay aims to explore the nature of the project and its version of neutrality, as compared to Wikipedia. Technologically, it is one of many experiments designed to replace human-generated writing with LLMs; conceptually, it is less a successor to Wikipedia than a return to an older model of producing officially sanctioned knowledge.

    Nearly every encyclopedia asserts some version of “neutrality.” Wikipedia’s definition is unusual: its “neutral point of view” policy aims not to pursue some Platonic ideal of balance or objectivity, but rather a faithful and proportional summary of what the best available sources say about a subject. Original ideas, reporting, and analysis on the part of its contributors are not allowed. Casting volunteers as “editors” and not “authors” is part of how “an encyclopedia that anyone can edit” is possible — by moving the locus of dispute from truth itself to which sources to use and how to incorporate them. As with the rest of Wikipedia, neutrality is less a perfect state than a continuously negotiated process wherein disputes are expected and common. While neutrality and sourcing discussions are often deeply fraught, with complicated histories that blur lines of reliability and result in lengthy discussions, they’re also constructive — a 2019 study in Nature found that articles with many such conflicts tended to be higher quality in general.

    On which sources to use, Wikipedia’s guideline about identifying “reliable sources” details its priorities: a reputation for fact-checking, accuracy, issuing corrections, editorial oversight, separating facts and opinions, no compromising connection to the subject, and other traditional markers of information literacy that librarians have taught to students and researchers for more than a century. Secondary and tertiary sources are preferred, deferring to them for the task of vetting and interpreting primary sources. Independent subjects are also preferred for any non-trivial claim, as article subjects have a hard time writing about themselves objectively. Ideological orientation is not a factor except insofar as narrative drive affects this list of priorities. Both of the following statements can align with Wikipedia’s definition of a “reliable source,” even though they’re opposed: “unicorns aren’t real but I wish they were;” “unicorns aren’t real and I’m glad they aren’t.” Either source would take priority over a source that claims “unicorns are real,” regardless of the author’s pro- or anti-unicorn sentiment.

    However, sourcing is also at the center, implicitly or explicitly, of many allegations that Wikipedia is not actually neutral. Some of these claims focus on Wikipedia’s “perennial sources list,” which includes dozens of sources whose reliability is frequently discussed, highlighted according to the outcomes of those discussions. The idea is to be able to point to a central page where someone can find links and summaries of past discussions rather than have volunteers explain for the umpteenth time why e.g. InfoWars is not a reliable source.

    I agree with criticism of this page to the extent it has given rise to a genre of source classification discussion applied not just to extreme cases like InfoWars but to sources that require some nuance, indirectly short-circuiting debates that should take place on a case-by-case basis. But even if the list were to be deleted altogether, it wouldn’t turn unreliable sources (according to the guideline) into reliable ones; it would just require more of those debates to play out rather than let someone point to a line in a table. There’s an optics argument to be had, too: it’s not that there aren’t more unreliable right-wing sources than left-wing sources; it’s just that people try to use unreliable right-wing sources more frequently in Wikipedia articles.

    But in large part, allegations of bias are a straightforward extension of a decades-old argument: that academia, science, mainstream media, etc. are broadly biased towards the left and/or untrustworthy. Whether through Rush Limbaugh’s “four corners of deceit” (government was the fourth corner) or some other articulation, the frame is well established. The extent to which it is true is outside the scope of this essay, but anyone who holds this view will inevitably see that bias in Wikipedia, which summarizes academia, science, and media. Musk made this point earlier this year when he called Wikipedia “an extension of legacy media propaganda.”

    It should not be surprising, then, that the sourcing used by Grokipedia is often radically different from Wikipedia’s. It’s not clear how reliably Grok will explain its own internal processes, but it should at least communicate the way its developers want Grokipedia to be seen. So I asked it to explain the way it prioritizes sources for different kinds of content, and it provided a table that’s worth including here; see below.

    The most obvious trend is its preference on most topics for primary, self-published and official sources like verified X users’ social media posts and government documents. These are put on par with or at higher priority than peer-reviewed journal articles, depending on the category. The only examples it provides among high-priority sources, apart from X users, are ArXiv (itself contending with an influx of LLM content) and PubMed for scientific/technical topics and Kremlin.ru for historical events.

    Some of Wikipedia’s fiercest critics contend that its version of neutrality unfairly endorses “Establishment” views on issues like vaccines, climate change, or the results of the 2020 US Presidential election, omitting minority positions or describing them in unfavorable terms. If many people hold a view, the argument goes, it is worth presenting on its own terms rather than deciding one set of sources is better than another. Grokipedia appears to align with this perspective, as its low-priority source criteria explains that it is sensitized to “emotional bias,” labels like “pseudoscience,” and anything that doesn’t present alternative perspectives.

    There is another characteristic of the sourcing that will be immediately apparent to anyone who has tried to do a literature review on a subject using a chatbot: it relies on sources available on the open web (or sources widely described by sources available on the open web). Commercial sites with good search engine optimization, apparent content farms, and personal blogs appear alongside traditional media sources. Grok can find extant text on the web faster than Wikipedia’s human editors, but does it have access to the books and articles that aren’t internet-accessible?

    All of this is ultimately subordinate to Grokipedia’s unavoidable prime directive of neutrality: neutrality is whatever Elon Musk says is neutral.

    According to the New York Times, Musk has been directly involved with Grok’s development, nudging it to the right on several issues. Not only does Grokipedia extoll Musk’s personal worldviews, but, as pointed out by many of the news articles about the project, it “breathlessly” promotes him and his products. At the end of the day, it doesn’t really matter what the training data is, how it’s weighted, how it negotiates points of view, etc. when the last step is necessarily some sort of post-processing/output filtering/reranking intervention based on Musk’s final word.

    For much of Wikipedia’s history, journalists and academics have enjoyed comparing it to historical encyclopedias like the Natural History, the Encyclopédie, and of course Encyclopaedia Britannica. Sometimes, like with Giles’ influential 2005 Nature study, it’s to compare their factual accuracy, but usually it’s to look at their structural and conceptual differences: Wikipedia is larger; Wikipedia is online; Wikipedia is accessible for free by anyone with an internet connection; Wikipedia is editable by anyone. But the most important distinction frequently gets lost: unlike nearly all historic encyclopedias, Wikipedia doesn’t need anyone’s permission to publish. There is no ideological test for participation or publication. There is no emperor, bishop, investor, or CEO who must approve of ideas expressed within, and there is no owner.

    Whether due to the great expense of producing, copying, and distributing voluminous works or because of tight control that structures of governance have exerted on sources of knowledge, encyclopedists as far back as Pliny the Elder, in the first century AD, have always needed the support and consent of powerful people (Pliny had relationships with both Vespasian and Titus) in order for their work to be read. In this way, while Grokipedia is technologically new, with enthusiasm in some ways reminiscent of Wikipedia’s early days, its epistemic hierarchy is more old-fashioned.

    That brings me to my biggest question: who is Grokipedia for, other than its owner? How big is the market for corporate, for-profit general knowledge sources that promote their own products and strictly adhere to the views of a billionaire founder? I know that if any corporation/billionaire has that kind of caché, built-in audience, and resources for a sustained push, it’s X/Musk. But what happens when other CEOs decide they don’t like their article on Wikipedia or Grokipedia and get into the encyclopedia game? McDonaldspedia and BritishPetroloeumpedia vie with Grok for dominance?

    Beyond the corporate nature of Grokipedia, my impression is that most people are not excited to completely trade human-created knowledge sources for fully machine-generated ones. The format of Grokipedia obscures that it is fundamentally just structured LLM-generation, and thus succeeds and fails in similar ways as any other chatbot query, trading the limitations of human judgment for the limitations of LLMs. Given how much AI resentment has been bubbling up in various corners of the internet, I’m frankly surprised “Slopipedia” wasn’t trending from launch.

    For better or worse, and I increasingly think it’s for the better, Wikipedia has developed something of an allergy to AI in general and chatbots in particular. Don’t use them to write articles, don’t use them to illustrate articles, don’t use them to prepare arguments on talk pages, etc., or risk getting banned. There are a handful of non-LLM AI uses, but Wikipedia is human-centric to such an extent that it may miss opportunities to scale labor and improve user experience.

    Perhaps Wikipedians are a potential audience. Even if, as argued by 404 Media’s Jason Koebler, Grokipedia “is not a ‘Wikipedia competitor’ [but] a fully robotic regurgitation machine,” its experiments in LLM-based encyclopedism may be valuable as an example of what Wikipedia could do if it wanted to. Does Grokipedia shed any light on particular topics that are better suited to LLM-generation than others? Does it confirm Wikipedia’s status quo that LLMs have no business writing articles at all?

    The most instructive experiment may be the opening up of primary and self-published sources for use in articles. There is no shortage of companies, influencers, and politicians interested in having their own words used to craft an encyclopedia article about them. That doesn’t usually serve a general reader very well, but the downside is it omits a lot of potentially useful detail, too. Take a journalist, for example. There’s not a lot of writing about journalists, but a policy that welcomes primary and self-published sources could draw information about the person and their work from their own writing, and it would remain more up to date than articles that have to wait for a secondary source. What else is worth comparing?

    Wikipedia, for all its many flaws, has always aimed to “set knowledge free” — by giving volunteers the ability to create and apply principles from the bottom-up, using technology to create a knowledge resource as well as to give it away for free, based on the belief that free knowledge is empowering. Opinions will vary about how successful it has been and where its blind spots are, but it’s hard to dispute its idealism. In contrast, Grokipedia’s defining feature as an encyclopedic project is the use of technological power to re-exert top-down authority over information and knowledge.

    This piece is published with a CC BY-SA 4.0 license by agreement with the author. You may share, republish, or adapt it freely, as long as you credit the author, clearly indicate any changes, link to this original article at Tech Policy Press, and distribute any derivative works with the same license.

    Continue Reading