bims-ecemfi Biomed News
on ECM and fibroblasts
Issue of 2025–06–08
eleven papers selected by
Badri Narayanan Narasimhan, University of California, San Diego



  1. Nat Commun. 2025 Jun 05. 16(1): 5213
      Neural progenitor cells (NPCs) hold immense potential as therapeutic candidates for neural regeneration, and materials-based strategies have emerged as attractive options for NPC expansion. However, maintaining NPC stemness has proven challenging in vitro, due to their propensity to form cell-dense neurospheres. While neurospheres promote cell-cell interactions required for NPC stem maintenance, they also restrict oxygen transport, leading to hypoxia and limited cell expansion. To overcome these limitations, we investigate two materials-based approaches to maintain NPC stemness: 1) physical matrix remodeling within a viscoelastic, stress-relaxing hydrogel and 2) matrix-induced N-cadherin-like signaling through a cell-instructive peptide. While viscoelasticity alone is sufficient to maintain NPC stemness compared to an elastic environment, NPCs still preferentially form neurospheres. The addition of N-cadherin-like peptides promotes a distributed culture of NPCs while maintaining their stemness through cadherin-mediated signaling, ultimately exhibiting improved long-term expansion and neural differentiation. Thus, our findings reveal matrix viscoelasticity and engineered N-cadherin-like interactions as having a synergistic effect on NPC expansion and differentiation within 3D matrices.
    DOI:  https://doi.org/10.1038/s41467-025-60540-8
  2. Acta Biomater. 2025 Jun 03. pii: S1742-7061(25)00411-8. [Epub ahead of print]
      Semiflexible polymer networks are ubiquitous in biological systems, including a scaffolding structure within cells called the actin cytoskeleton. The polymers in these networks are interconnected by transient bonds. For example, actin filaments in the cytoskeleton are physically connected via cross-linker proteins. The mechanical and kinetic properties of the cross-linkers significantly affect the rheological properties of the actin cytoskeleton. Here, we employed an agent-based model to elucidate how the force-dependent behaviors of the cross-linkers determine the material properties of passive networks without molecular motors and the force generation of active networks with molecular motors. The cross-linkers are assumed to behave either as a slip bond, whose dissociation rate increases with forces, or as a catch-slip bond, whose dissociation rate decreases with forces at low force level but increases with forces at high force level. We found that catch-slip-bond cross-linkers can simultaneously increase both the stress and the strain at the yield point. Through a systematic variation in the force dependence of the catch-slip bonds, we identified the specific parameter regimes that enable network reinforcement and enhanced extensibility simultaneously. Specifically, we found that a sufficiently large force threshold for the catch-slip transition is essential for maintaining dynamic force-bearing elements that turnover continuously-a mechanism not achievable with slip bonds. Additionally, we demonstrate that such force-dependent redistribution of the catch-slip bonds substantially enhances internal contractile forces generated by a motor in active networks. STATEMENT OF SIGNIFICANCE: Polymer networks are ubiquitous in industrial and biological systems. The polymers in these networks are often interconnected by transient bonds. The transient bonds behave as a slip bond whose dissociation rate is proportional to forces or as a catch-slip bond whose dissociation rate decreases with increased force (catch) at low force level but increases with increased force (slip) at high force level. In this study, we computationally tested different types of catch-slip bonds to define how the material properties of polymer networks are fine-tuned by each property of molecular bonds. We found that catch-slip bonds can increase both stress and strain at a yield point, which is impossible to achieve without the catch-slip bonds.
    Keywords:  actin; agent-based model; cross-linker; cytoskeleton; slip bond
    DOI:  https://doi.org/10.1016/j.actbio.2025.06.004
  3. Adv Sci (Weinh). 2025 Jun 03. e2415029
      Nuclear translocation of the transcription regulatory proteins yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) is a critical readout of cellular mechanotransduction. Recent experiments have demonstrated that cells on substrates with well-defined nanotopographies demonstrate mechanoadaptation through a multitude of effects - increased integrin endocytosis as a function of nanopillar curvature, increased local actin assembly on nanopillars but decreased global cytoskeletal stiffness, and enhanced nuclear deformation. How do cells respond to local nanotopographical cues and integrate their responses across multiple length scales? This question is addressed using a biophysical model that incorporates plasma membrane (PM) curvature-dependent endocytosis, PM curvature-sensitive actin assembly, and stretch-induced opening of nuclear pore complexes (NPCs) in the nuclear envelope (NE). This model recapitulates lower levels of global cytoskeletal assembly on nanopillar substrates, which can be partially compensated for by local actin assembly and NE indentation, leading to enhanced YAP/TAZ transport through stretched NPCs. Using cell shapes informed by electron micrographs and fluorescence images, the model predicts lamin A and F-actin localization around nanopillars, in agreement with experimental measurements. Finally, simulations predict nuclear accumulation of YAP/TAZ following rupture of the NE and this is validated by experiments. Overall, this study indicates that nanotopography tunes mechanoadaptation through both positive and negative feedback on mechanotransduction.
    Keywords:  mechanoadaptation; mechanotransduction; nanotopography; nuclear transport; systems biophysics
    DOI:  https://doi.org/10.1002/advs.202415029
  4. Nanoscale. 2025 Jun 04.
      Nanoscale fabrication techniques have played an essential role in revealing the impact of extracellular matrix (ECM) nanotopography on cellular behavior. However, the mechanisms by which nanotopographical cues from the ECM influence cellular function remain unclear. To approach these questions, we have engineered a novel class of nanopatterned ECM constructs suitable for cryogenic electron tomography (cryo-ET), the highest resolution modality for imaging frozen hydrated cells in 3D. We electrospun aligned and randomly oriented ECM fibers directly onto transmission electron microscopy (TEM) supports to generate fibrous scaffolds that mimic physiological ECM in healthy (organized ECM) and diseased (disorganized ECM) states. We produced fibers from gelatin without toxic additives and cross-linked them to maintain structural stability in aqueous environments. The electrospun fibers had an average fiber diameter of hundreds of nanometers. We confirmed that the nanopatterned TEM supports can serve as viable cell culture substrates that can influence cell organization and demonstrated their compatibility with plunge freezing and cryo-ET. By enabling nanoscale structural analysis inside cells on substrates with programmable topographies, this platform can be used to study the physical cues necessary for healthy endothelial tissue formation and pathologies that are linked to endothelial dysfunction in diseases such as peripheral arterial disease.
    DOI:  https://doi.org/10.1039/d4nr05508j
  5. bioRxiv. 2025 May 22. pii: 2025.05.18.654456. [Epub ahead of print]
      The actin cortex, a thin layer of actomyosin network beneath the plasma membrane, regulates various cell functions by generating active forces and inducing membrane deformations, including blebs. Although upstream signaling is involved in regulating cell shape, the extent to which downstream actomyosin molecules can control the shape remains elusive. Here, using a minimal reconstituted system with a combination of agent-based computational model, we show that while actin-membrane coupling strength determines the magnitude of membrane deformation, its balance with actin network connectivity governs the bleb initiation mechanism, either by detachment of the cortex from the membrane or by rupture of the cortex. This balance also regulates whether single or multiple blebs form. Furthermore, both experiments and simulations suggest that not only the dense cortical network but also the sparse volume-spanning network actively contributes to regulating bleb number. These findings provide mechanical insights into how cells tune actin network organization to control their shape.
    DOI:  https://doi.org/10.1101/2025.05.18.654456
  6. ACS Appl Mater Interfaces. 2025 Jun 05.
      To regulate the maintenance and differentiation of stem and progenitor cells, a variety of hydrogels have been developed and applied as two-dimensional (2D) cell culture substrates that can provide well-defined mechanical cues by adjusting the stiffness. Recently, cell-laden hydrogels have been drawing attention as the three-dimensional (3D) cellular environments that can be patterned or printed by extrusion of the cell-polymer mixtures. Hydrogels also serve as 3D microstructures that can stimulate cells both mechanically and geometrically. For flexible, modular functionalization, the coupling of different extracellular matrix (ECM) proteins to side walls and curved surfaces is necessary. However, widely used heterobifunctional photo-cross-linkers encounter a problem because the light cannot reach into the scaffolds uniformly. In this study, we overcame this problem by integrating monomers with N-hydroxysuccinimide (NHS) groups into the copolymer hydrogels with tunable stiffness via careful adjustment of solvent miscibility. This enabled one-step surface functionalization with extracellular matrix proteins such as fibronectin, laminin, and gelatin, replacing photoactivation or laborious multistep functionalization. On the 2D hydrogel substrates functionalized with fibronectin, we found that more than 80% of human mesenchymal stem cells (hMSCs) were viable, and about 60% of them maintained proliferation capacity. These data confirmed that the introduction of NHS monomers caused no cytotoxic effect. We further designed and fabricated 3D microstructures containing various wall and bottom architectures using 3D printed stamps. The uniform functionalization of side walls and bottom surfaces with ECM proteins enabled us to accommodate hMSCs inside the 3D scaffolds, which was in stark contrast to commonly used photo-cross-linkers. The 3D scaffolds showed reversible swelling and deswelling by the addition and removal of soluble guest molecules in the presence of hMSCs, suggesting that the one-step functionalization method established in this study can be applied for a variety of hydrogel-based 3D microstructures for various cell types.
    Keywords:  3D cellular scaffold; human mesenchymal stem cell; polyacrylamide; supramolecular hydrogel; surface functionalization
    DOI:  https://doi.org/10.1021/acsami.5c08146
  7. Exp Cell Res. 2025 Jun 02. pii: S0014-4827(25)00225-3. [Epub ahead of print]450(2): 114629
      The extracellular matrix (ECM) plays a crucial role in regulating intracellular signaling in tumor cells. Although the influence of ECM stiffness on tumor malignancy has been well studied, the adaptation of tumors to a soft extratumoral environment with an increasing proliferation rate remains poorly understood. In this study, we investigated the mechanism of tumor cell adaptation to a soft environment and its relationship with tumor malignancy. A soft extracellular environment was required for adaptation. Among the various cell lines, highly malignant cancer cells, such as MDA-MB-231, PC3 and KP4, demonstrated an adaptive response to soft environments, as evidenced by increased cell extension and proliferation. Furthermore, adaptation to a soft environment enhanced lung colonization in a mouse model of breast cancer, suggesting a potential link between adaptability to soft environments and accelerated tumor malignancy. The mechanical environment of the cells before being transferred to a softer environment affected their adaptation as a mechanical memory via histone modifications. Integrin β1-mediated adhesion to the ECM played a central role in this adaptive phenomenon. Interestingly, adaptation occurred through a mechanism with limited involvement of mechanotransduction and prominent enhancement of ribosomal activity, providing new insights into the proliferation mechanisms in a soft environment. This study revealed how tumor cells adapt to a soft environment and showed that the adaptation phenomenon may be related to malignant transformation. Thus, the adaptation mechanism may be a therapeutic target for the suppression of metastasis.
    Keywords:  Adaptation; Biophysics; Metastasis; Soft environment
    DOI:  https://doi.org/10.1016/j.yexcr.2025.114629
  8. Adv Sci (Weinh). 2025 Jun 05. e04351
      Extracellular matrix (ECM) remodeling plays critical roles in cancer progression and involves alterations in its composition and biophysical properties. Aggressiveness and malignancy of solid tumors are strongly correlated with tissue stiffening, mainly due to upregulated ECM production and cross-linking. However, nothing is known about the tensional alterations that occur at the single-fiber level during tumorigenesis in humans. The well-validated peptide tension probe (FnBPA5) now reveals that Fibronectin fibers lose their tension as invasive tumors progress while they are stretched in healthy human breast tissue stroma and in ductal carcinoma in situ (DCIS), the non-invasive precursor of breast cancer. In invasive carcinomas, cancer cells, cancer-associated fibroblasts (CAFs) and infiltrating immune cells (cytotoxic T cells and regulatory T cells), are predominantly located in proximity to untensed Fibronectin fibers. This is significant, as Fibronectin fiber stretching can mechano-regulate the reciprocal cell-ECM crosstalk and the bioavailability of ECM-bound molecules. Not only tissue stiffening, but also the accumulation of untensed Fibronectin fibers may serve as a mechanical biomarker that correlates with tumor grade. Loss of Fibronectin fiber tension may play a central role in regulating tumor invasiveness. This suggests that physically altered ECM fibers can be exploited for stroma-targeted drug delivery and immunotherapy.
    Keywords:  breast cancer; ductal carcinoma in situ (DCIS); extracellular matrix; mechanobiology; tensional state of Fibronectin
    DOI:  https://doi.org/10.1002/advs.202404351
  9. Biomaterials. 2025 May 31. pii: S0142-9612(25)00379-5. [Epub ahead of print]324 123460
      Current treatments for autoimmune diseases like Type 1 Diabetes (T1D) carry significant risks because they lack tissue specificity. A promising strategy is to achieve persistent presentation of relevant antigens (Ags) in non-inflamed sites by tolerogenic Ag-presenting cells (APCs) like fibroblastic reticular cells (FRCs). FRCs build lymph node (LN) reticula and act as immunomodulatory non-professional APCs. However, their therapeutic potential for Ag-specific immunomodulation for T1D remains unexplored. We engineered 3D FRC-based reticula using freeze-dried macroporous gelatin scaffolds with customizable pore diameters (small: <50 μm, medium: <200 μm, large: <300 μm) to evaluate FRC phenotype and FRC-T cell interactions, leveraging FRCs' ability to build dynamic LN reticula that expand and contract during inflammation. Our scaffolds promoted FRC viability, reticular formation, FRC phenotypic marker expression, and extracellular matrix secretion compared to 2D culture. GFP-Luciferase fusion (GLF)-expressing FRCs subcutaneously implanted in our scaffolds survived for at least 21 days regardless of pore size. Implantation in the vascularized fat pad led to graft rejection by day 14 in pre-diabetic NOD mice but not in immunodeficient NODscid. Our scaffolds outperformed clinically-used biologic gels, demonstrated the limitations of the NOD mouse model in longitudinal imaging of GLF+ cell survival, and validated our scaffold-based FRC delivery approach for future therapeutic applications. Co-culture of T cells in scaffolds with FRCs presenting T1D Ags enabled Ag-specific T cell engagement with reduced cytotoxic and increased anergic and regulatory phenotypes. Our results validate the use of macroporous gelatin scaffolds to enhance FRC phenotypic markers, in vivo survival, and diabetogenic cell immunomodulation in vitro.
    Keywords:  3D culture; Antigen-specific therapies; Immune engineering; Lymphoid organs; Stromal cells; Tolerance; Type 1 diabetes
    DOI:  https://doi.org/10.1016/j.biomaterials.2025.123460
  10. Front Oncol. 2025 ;15 1585935
       Background: Cell-cell crosstalk in the tumor microenvironment (TME) is crucial for cancer development and strongly correlates with clinical outcomes. Interpatient variability in tumor microenvironment composition and function poses ongoing challenges for personalized therapy selection, remaining a significant clinical problem. Serpin family E member 2 (SERPINE2) released from the tumor microenvironment exhibits significant regulatory functions in cancer progression but the role of SERPINE2 in the tumor microenvironment remains unclear. In this study, we want to investigate the potential mechanism of SERPINE2 in tumor microenvironment of colon cancer.
    Methods: Bioinformatics analysis was used for exploring the mRNA expression level of SERPINE family in Pan-cancer, the prognostic significance of SERPINE family overexpression in four cancer types, the clinical relevance of SERPINE2 and the potential function of SERPINE2 in colorectal cancer. We conducted qRT-PCR, Western blot and ELISA to investigate the expression of SERPINE2. Additionally, Tissue chips, Transwell assays, Cell counting kit-8 assay, and co-culture system were used to evaluate the relationship between SERPINE2 and polarization of tumor-associated macrophages.
    Results: Based on public database screening, the SERPINE family genes were significantly upregulated in various cancers, and high expression of SERPINE family genes in colorectal cancer was closely associated with poor prognosis. Compared to other family members, SERPINE2 showed a high expression level and was closely related to clinical malignant progression of colon cancer patients. co-expression network analysis, KEGG and GO analysis revealed that SERPINE2 expression correlates with tumor immunoregulation, division and proliferation. Immune infiltration analysis indicated a significant positive correlation between SERPINE2 and M2 macrophage infiltration, and tissue chip confirmed the correlation between SERPINE2 expression in colon cancer tissues and macrophage infiltration. Cell co-culture experiments further demonstrated that SERPINE2 secreted by colon cancer cells can induce polarization of M2 macrophages. Next, the recombinant protein SERPINE2 was observed to stimulate macrophage polarization. We found macrophages induced by SERPINE2 in co-culture with cancer cells accelerated cancer cell proliferation and migration.
    Conclusion: Our study demonstrates that tumor-secreted SERPINE2 mediates a positive feedback loop between tumor cells and M2 macrophages to accelerate cancer progression, suggesting SERPINE2 may be as a promising therapeutic target for colon cancer treatment.
    Keywords:  M2 polarization; SerpinE2; colon cancer; immune infiltration; tumor-associated macrophages
    DOI:  https://doi.org/10.3389/fonc.2025.1585935
  11. Acta Biomater. 2025 May 29. pii: S1742-7061(25)00401-5. [Epub ahead of print]
      Cytokines are potent endogenous modulators of innate immunity, making them key mediators of macrophage plasticity for immunotherapy. However, the clinical translation of recombinant cytokines as therapeutics is limited by systemic side effects, caused by cytokines' pleiotropy, potency, and non-specific biodistribution following systemic dosing. We developed a cytokine delivery platform utilizing poly(acrylamide-co-methacrylic acid) synthetic nanogels as a biodegradable substrate for conjugated recombinant cytokines (i.e., IFNγ, IL4, or IL10), called Synthetic Nano-CytoKines or "SyNK". We evaluated the phenotypic response of macrophages to these conjugates following prophylactic or therapeutic dosing, in the presence or absence of soluble inflammatory signals. Our data confirmed that SyNK is highly cytocompatible with murine macrophages, preserves the activity of conjugated recombinant cytokines to both macrophages and dendritic cells, and minimizes systemic exposure to freely soluble recombinant cytokines. Intrinsic activity of the nanomaterial was modest, acting in combination with the conjugated cytokine, and resulted in unique phenotypes with IL4-SyNK and IL10-SyNK stimulation that could potentially be leveraged for therapeutic applications. We further demonstrated that RAW264.7 macrophages adopt distinct alternative phenotypes upon IL4 or IL10 stimulation in different classically polarizing microenvironments, as measured by spectral flow cytometry and secretome multiplex, which are similar for soluble recombinant cytokine and the corresponding SyNK. These findings offer a potential mechanism through which IL4 or IL10-SyNK can redirect the classically activated macrophage antigen presentation, T cell co-stimulation, or microenvironment regulatory functions for therapeutic purposes. STATEMENT OF SIGNIFICANCE: Cytokines have been extensively investigated as immune therapies, but their clinical translation is limited by their systemic toxicity and frequent dosing regimens. Existing approaches have improved cytokine stability and local delivery but still face challenges in systemic administration and controlling immune response. We developed a cytokine delivery platform using biodegradable poly(acrylamide-co-methacrylic acid) nanogels to conjugate cytokines (e.g. IFNγ, IL4, or IL10) aimed at systemic macrophage immunotherapy. We show that our platform preserves cytokine activity and eliminates the release of free cytokine. We further explore, for the first time, how different stimuli in the macrophage environment influence their response to the cytokine bioconjugates. Our work provides thorough insights into macrophage plasticity and addresses key limitations of current strategies.
    Keywords:  Bioconjugation; Cytokine; Immunotherapy; Macrophage; Nanogel
    DOI:  https://doi.org/10.1016/j.actbio.2025.05.066