Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 29.
Article in English | MEDLINE | ID: mdl-38854106

ABSTRACT

Chromosomal instability (CIN) is a hallmark of cancer that drives metastasis, immune evasion and treatment resistance. CIN results from chromosome mis-segregation events during anaphase, as excessive chromatin is packaged in micronuclei (MN), that can be enumerated to quantify CIN. Despite recent advancements in automation through computer vision and machine learning, the assessment of CIN remains a predominantly manual and time-consuming task, thus hampering important work in the field. Here, we present micronuclAI , a novel pipeline for automated and reliable quantification of MN of varying size, morphology and location from DNA-only stained images. In micronucleAI , single-cell crops are extracted from high-resolution microscopy images with the help of segmentation masks, which are then used to train a convolutional neural network (CNN) to output the number of MN associated with each cell. The pipeline was evaluated against manual single-cell level counts by experts and against routinely used MN ratio within the complete image. The classifier was able to achieve a weighted F1 score of 0.937 on the test dataset and the complete pipeline can achieve close to human-level performance on various datasets derived from multiple human and murine cancer cell lines. The pipeline achieved a root-mean-square deviation (RMSE) value of 0.0041, an R 2 of 0.87 and a Pearson's correlation of 0.938 on images obtained at 10X magnification. We tested the approach in otherwise isogenic cell lines in which we genetically dialed up or down CIN rates, and also on a publicly available image data set (obtained at 100X) and achieved an RMSE value of 0.0159, an R 2 of 0.90, and a Pearson's correlation of 0.951. Given the increasing interest in developing therapies for CIN-driven cancers, this method provides an important, scalable, and rapid approach to quantifying CIN on routinely obtained images. We release a GUI-implementation for easy access and utilization of the pipeline.

2.
Nat Biotechnol ; 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783148

ABSTRACT

Single-nucleotide variants (SNVs) in key T cell genes can drive clinical pathologies and could be repurposed to improve cellular cancer immunotherapies. Here, we perform massively parallel base-editing screens to generate thousands of variants at gene loci annotated with known or potential clinical relevance. We discover a broad landscape of putative gain-of-function (GOF) and loss-of-function (LOF) mutations, including in PIK3CD and the gene encoding its regulatory subunit, PIK3R1, LCK, SOS1, AKT1 and RHOA. Base editing of PIK3CD and PIK3R1 variants in T cells with an engineered T cell receptor specific to a melanoma epitope or in different generations of CD19 chimeric antigen receptor (CAR) T cells demonstrates that discovered GOF variants, but not LOF or silent mutation controls, enhanced signaling, cytokine production and lysis of cognate melanoma and leukemia cell models, respectively. Additionally, we show that generations of CD19 CAR T cells engineered with PIK3CD GOF mutations demonstrate enhanced antigen-specific signaling, cytokine production and leukemia cell killing, including when benchmarked against other recent strategies.

3.
Cancer Cell ; 41(7): 1207-1221.e12, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37327789

ABSTRACT

The cell-autonomous balance of immune-inhibitory and -stimulatory signals is a critical process in cancer immune evasion. Using patient-derived co-cultures, humanized mouse models, and single-cell RNA-sequencing of patient melanomas biopsied before and on immune checkpoint blockade, we find that intact cancer cell-intrinsic expression of CD58 and ligation to CD2 is required for anti-tumor immunity and is predictive of treatment response. Defects in this axis promote immune evasion through diminished T cell activation, impaired intratumoral T cell infiltration and proliferation, and concurrently increased PD-L1 protein stabilization. Through CRISPR-Cas9 and proteomics screens, we identify and validate CMTM6 as critical for CD58 stability and upregulation of PD-L1 upon CD58 loss. Competition between CD58 and PD-L1 for CMTM6 binding determines their rate of endosomal recycling over lysosomal degradation. Overall, we describe an underappreciated yet critical axis of cancer immunity and provide a molecular basis for how cancer cells balance immune inhibitory and stimulatory cues.


Subject(s)
B7-H1 Antigen , Melanoma , Mice , Animals , B7-H1 Antigen/genetics , T-Lymphocytes , CD58 Antigens/chemistry , CD58 Antigens/metabolism , Melanoma/genetics , Melanoma/metabolism , Lymphocyte Activation
4.
ACS Cent Sci ; 9(4): 787-804, 2023 Apr 26.
Article in English | MEDLINE | ID: mdl-37122450

ABSTRACT

Glycoengineered bacteria have emerged as a cost-effective platform for rapid and controllable biosynthesis of designer conjugate vaccines. However, little is known about the engagement of such conjugates with naïve B cells to induce the formation of germinal centers (GC), a subanatomical microenvironment that converts naïve B cells into antibody-secreting plasma cells. Using a three-dimensional biomaterials-based B-cell follicular organoid system, we demonstrate that conjugates triggered robust expression of hallmark GC markers, B cell receptor clustering, intracellular signaling, and somatic hypermutation. These responses depended on the relative immunogenicity of the conjugate and correlated with the humoral response in vivo. The occurrence of these mechanisms was exploited for the discovery of high-affinity antibodies against components of the conjugate on a time scale that was significantly shorter than for typical animal immunization-based workflows. Collectively, these findings highlight the potential of synthetic organoids for rapidly predicting conjugate vaccine efficacy as well as expediting antigen-specific antibody discovery.

5.
Nat Mater ; 22(4): 511-523, 2023 04.
Article in English | MEDLINE | ID: mdl-36928381

ABSTRACT

Activated B-cell-like diffuse large B-cell lymphomas (ABC-DLBCLs) are characterized by constitutive activation of nuclear factor κB driven by the B-cell receptor (BCR) and Toll-like receptor (TLR) pathways. However, BCR-pathway-targeted therapies have limited impact on DLBCLs. Here we used >1,100 DLBCL patient samples to determine immune and extracellular matrix cues in the lymphoid tumour microenvironment (Ly-TME) and built representative synthetic-hydrogel-based B-cell-lymphoma organoids accordingly. We demonstrate that Ly-TME cellular and biophysical factors amplify the BCR-MYD88-TLR9 multiprotein supercomplex and induce cooperative signalling pathways in ABC-DLBCL cells, which reduce the efficacy of compounds targeting the BCR pathway members Bruton tyrosine kinase and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1). Combinatorial inhibition of multiple aberrant signalling pathways induced higher antitumour efficacy in lymphoid organoids and implanted ABC-DLBCL patient tumours in vivo. Our studies define the complex crosstalk between malignant ABC-DLBCL cells and Ly-TME, and provide rational combinatorial therapies that rescue Ly-TME-mediated attenuation of treatment response to MALT1 inhibitors.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Tumor Microenvironment , Humans , Cell Line, Tumor , Signal Transduction , NF-kappa B/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism
6.
bioRxiv ; 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38168306

ABSTRACT

Base editing enables generation of single nucleotide variants, but large-scale screening in primary human T cells is limited due to low editing efficiency, among other challenges 1 . Here, we developed a high-throughput approach for high-efficiency and massively parallel adenine and cytosine base-editor screening in primary human T cells. We performed multiple large-scale screens editing 102 genes with central functions in T cells and full-length tiling mutagenesis of selected genes, and read out variant effects on hallmarks of T cell anti-tumor immunity, including activation, proliferation, and cytokine production. We discovered a broad landscape of gain- and loss-of-function mutations, including in PIK3CD and its regulatory subunit encoded by PIK3R1, LCK , AKT1, CTLA-4 and JAK1 . We identified variants that affected several (e.g., PIK3CD C416R) or only selected (e.g. LCK Y505C) hallmarks of T cell activity, and functionally validated several hits by probing downstream signaling nodes and testing their impact on T cell polyfunctionality and proliferation. Using primary human T cells in which we engineered a T cell receptor (TCR) specific to a commonly presented tumor testis antigen as a model for cellular immunotherapy, we demonstrate that base edits identified in our screens can tune specific or broad T cell functions and ultimately improve tumor elimination while exerting minimal off-target activity. In summary, we present the first large-scale base editing screen in primary human T cells and provide a framework for scalable and targeted base editing at high efficiency. Coupled with multi-modal phenotypic mapping, we accurately nominate variants that produce a desirable T cell state and leverage these synthetic proteins to improve models of cellular cancer immunotherapies.

7.
Cell ; 185(14): 2591-2608.e30, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35803246

ABSTRACT

Melanoma brain metastasis (MBM) frequently occurs in patients with advanced melanoma; yet, our understanding of the underlying salient biology is rudimentary. Here, we performed single-cell/nucleus RNA-seq in 22 treatment-naive MBMs and 10 extracranial melanoma metastases (ECMs) and matched spatial single-cell transcriptomics and T cell receptor (TCR)-seq. Cancer cells from MBM were more chromosomally unstable, adopted a neuronal-like cell state, and enriched for spatially variably expressed metabolic pathways. Key observations were validated in independent patient cohorts, patient-derived MBM/ECM xenograft models, RNA/ATAC-seq, proteomics, and multiplexed imaging. Integrated spatial analyses revealed distinct geography of putative cancer immune evasion and evidence for more abundant intra-tumoral B to plasma cell differentiation in lymphoid aggregates in MBM. MBM harbored larger fractions of monocyte-derived macrophages and dysfunctional TOX+CD8+ T cells with distinct expression of immune checkpoints. This work provides comprehensive insights into MBM biology and serves as a foundational resource for further discovery and therapeutic exploration.


Subject(s)
Brain Neoplasms , Melanoma , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , CD8-Positive T-Lymphocytes/pathology , Ecosystem , Humans , RNA-Seq
8.
Blood ; 137(6): 788-800, 2021 02 11.
Article in English | MEDLINE | ID: mdl-32785655

ABSTRACT

MALT1 inhibitors are promising therapeutic agents for B-cell lymphomas that are dependent on constitutive or aberrant signaling pathways. However, a potential limitation for signal transduction-targeted therapies is the occurrence of feedback mechanisms that enable escape from the full impact of such drugs. Here, we used a functional genomics screen in activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL) cells treated with a small molecule irreversible inhibitor of MALT1 to identify genes that might confer resistance or enhance the activity of MALT1 inhibition (MALT1i). We find that loss of B-cell receptor (BCR)- and phosphatidylinositol 3-kinase (PI3K)-activating proteins enhanced sensitivity, whereas loss of negative regulators of these pathways (eg, TRAF2, TNFAIP3) promoted resistance. These findings were validated by knockdown of individual genes and a combinatorial drug screen focused on BCR and PI3K pathway-targeting drugs. Among these, the most potent combinatorial effect was observed with PI3Kδ inhibitors against ABC-DLBCLs in vitro and in vivo, but that led to an adaptive increase in phosphorylated S6 and eventual disease progression. Along these lines, MALT1i promoted increased MTORC1 activity and phosphorylation of S6K1-T389 and S6-S235/6, an effect that was only partially blocked by PI3Kδ inhibition in vitro and in vivo. In contrast, simultaneous inhibition of MALT1 and MTORC1 prevented S6 phosphorylation, yielded potent activity against DLBCL cell lines and primary patient specimens, and resulted in more profound tumor regression and significantly improved survival of ABC-DLBCLs in vivo compared with PI3K inhibitors. These findings provide a basis for maximal therapeutic impact of MALT1 inhibitors in the clinic, by disrupting feedback mechanisms that might otherwise limit their efficacy.


Subject(s)
Antineoplastic Agents/therapeutic use , Feedback, Physiological/drug effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Receptors, Antigen, B-Cell/immunology , Toll-Like Receptors/immunology , Animals , Antineoplastic Agents/pharmacology , Drug Design , Drug Resistance, Neoplasm , Drug Synergism , Female , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred NOD , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/physiology , Neoplasm Proteins/physiology , Organoids/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Small Interfering/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
Blood Adv ; 4(20): 5226-5231, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33104794

ABSTRACT

Molecular alterations in the histone methyltransferase EZH2 and the antiapoptotic protein Bcl-2 frequently co-occur in diffuse large B-cell lymphoma (DLBCL). Because DLBCL tumors with these characteristics are likely dependent on both oncogenes, dual targeting of EZH2 and Bcl-2 is a rational therapeutic approach. We hypothesized that EZH2 and Bcl-2 inhibition would be synergistic in DLBCL. To test this, we evaluated the EZH2 inhibitor tazemetostat and the Bcl-2 inhibitor venetoclax in DLBCL cells, 3-dimensional lymphoma organoids, and patient-derived xenografts (PDXs). We found that tazemetostat and venetoclax are synergistic in DLBCL cells and 3-dimensional lymphoma organoids that harbor an EZH2 mutation and an IGH/BCL2 translocation but not in wild-type cells. Tazemetostat treatment results in upregulation of proapoptotic Bcl-2 family members and priming of mitochondria to BH3-mediated apoptosis, which may sensitize cells to venetoclax. The combination of tazemetostat and venetoclax was also synergistic in vivo. In DLBCL PDXs, short-course combination therapy resulted in complete remissions that were durable over time and associated with superior overall survival compared with either drug alone.


Subject(s)
Antineoplastic Agents , Lymphoma, Large B-Cell, Diffuse , Antineoplastic Agents/therapeutic use , Apoptosis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology
10.
Nat Rev Mater ; 4(6): 355-378, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31903226

ABSTRACT

Immunoengineering applies quantitative and materials-based approaches for the investigation of the immune system and for the development of therapeutic solutions for various diseases, such as infection, cancer, inflammatory diseases and age-related malfunctions. The design of immunomodulatory and cell therapies requires the precise understanding of immune cell formation and activation in primary, secondary and ectopic tertiary immune organs. However, the study of the immune system has long been limited to in vivo approaches, which often do not allow multidimensional control of intracellular and extracellular processes, and to 2D in vitro models, which lack physiological relevance. 3D models built with synthetic and natural materials enable the structural and functional recreation of immune tissues. These models are being explored for the investigation of immune function and dysfunction at the cell, tissue and organ levels. In this Review, we discuss 2D and 3D approaches for the engineering of primary, secondary and tertiary immune structures at multiple scales. We highlight important insights gained using these models and examine multiscale engineering strategies for the design and development of immunotherapies. Finally, dynamic 4D materials are investigated for their potential to provide stimuli-dependent and context-dependent scaffolds for the generation of immune organ models.

11.
Biomaterials ; 198: 27-36, 2019 04.
Article in English | MEDLINE | ID: mdl-30041943

ABSTRACT

Most antigen discovery and vaccine development aimed at driving functional B cell responses rely on mouse immunizations studies. To date, there is no 3D ex vivo immune tissues, which are capable of driving antigen-specific B cell responses to rapidly determine the humoral immunogenicity of antigens, understand the role of extracellular matrix in humoral immunity, and generate high affinity antibody responses. This can be attributed to the complexity of B cell differentiation and affinity maturation process in the germinal center (GC) reaction, which makes these highly specialized cells susceptible to rapid apoptosis ex vivo. We have previously reported immune tissues that show ex vivo GC-like response, however in a non-antigen specific manner. Here, we report a maleimide (MAL)-functionalized polyethylene glycol (PEG)-based designer immune tissues that modulate B cell differentiation and enriches antigen-specific GC B cells in the presence of T-cell like signals. With the 3D synthetic immune tissue platform, we assessed various hydrogel design parameters to control ex vivo GC reaction. Using an Ezh2fl/fl Cγ1-cre transgenic mouse model, we demonstrated ex vivo IgG1 antibody class switching. Using immune tissues developed from a B1-8hi mutant mouse that represents a recombined antibody variable region derived from a 4-hydroxy-3-nitrophenylacetyl (NP) hapten binding antibody (B1-8), we demonstrate antigen specificity and selective enrichment of antigen-specific B cells with high affinity at both cell surface and secreted levels in integrin ligand-dependent manner. The ex vivo antigen-specific platform technology offers use in scientific understanding of immunobiology, matrix immunology, and in biotechnology applications, ranging from the antigen testing, vaccine development, and generation of antibodies against diseases.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , T-Lymphocytes/immunology , Tissue Engineering/methods , Animals , Antigens/immunology , B-Lymphocytes/cytology , Biocompatible Materials/chemistry , Cell Differentiation , Cells, Cultured , Female , Germinal Center/cytology , Immunoglobulin G/immunology , Male , Maleimides/chemistry , Mice, Inbred C57BL , Polyethylene Glycols/chemistry , T-Lymphocytes/cytology
12.
Cell Rep ; 23(2): 499-511, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29642007

ABSTRACT

The role of microenvironment-mediated biophysical forces in human lymphomas remains elusive. Diffuse large B cell lymphomas (DLBCLs) are heterogeneous tumors, which originate from highly proliferative germinal center B cells. These tumors, their associated neo-vessels, and lymphatics presumably expose cells to particular fluid flow and survival signals. Here, we show that fluid flow enhances proliferation and modulates response of DLBCLs to specific therapeutic agents. Fluid flow upregulates surface expression of B cell receptors (BCRs) and integrin receptors in subsets of ABC-DLBCLs with either CD79A/B mutations or WT BCRs, similar to what is observed with xenografted human tumors in mice. Fluid flow differentially upregulates signaling targets, such as SYK and p70S6K, in ABC-DLBCLs. By selective knockdown of CD79B and inhibition of signaling targets, we provide mechanistic insights into how fluid flow mechanomodulates BCRs and integrins in ABC-DLBCLs. These findings redefine microenvironment factors that regulate lymphoma-drug interactions and will be critical for testing targeted therapies.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , Apoptosis/drug effects , CD79 Antigens/antagonists & inhibitors , CD79 Antigens/genetics , CD79 Antigens/metabolism , Cell Line, Tumor , Cytokines/metabolism , Doxorubicin/pharmacology , Humans , Integrins/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Inbred NOD , Microfluidics/instrumentation , Microfluidics/methods , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Antigen, B-Cell/genetics , Shear Strength , Signal Transduction , Tumor Microenvironment , Up-Regulation , src-Family Kinases/metabolism
13.
Curr Opin Hematol ; 24(4): 377-383, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28426555

ABSTRACT

PURPOSE OF REVIEW: The specialized microenvironments of lymphoid tissue affect immune cell function and progression of disease. However, current animal models are low throughput and a large number of human diseases are difficult to model in animals. Animal models are less amenable to manipulation of tissue niche components, signalling pathways, epigenetics, and genome editing than ex vivo models. On the other hand, conventional 2D cultures lack the physiological relevance to study precise microenvironmental interactions. Thus, artificial tissues are being developed to study these interactions in the context of immune development, function, and disease. RECENT FINDINGS: New bone marrow and lymph node models have been created to, respectively, study microenvironmental interactions in hematopoiesis and germinal center-like biology. These models have also been extended to understand the effect of these interactions on the progression and therapeutic response in leukemia, multiple myeloma, and lymphoma. SUMMARY: 3D in-vitro immune models have elucidated new cellular, biochemical, and biophysical interactions as potential regulatory mechanisms, therapeutic targets, or biomarkers that previously could not be studied in animal models and conventional 2D cultures. Incorporation of advanced biomaterials, microfluidics, genome editing, and single-cell analysis tools will enable further studies of function, driver mutations, and tumor heterogeneity. Continual refinement will help inform the development of antibody and cell-based immunotherapeutics and patient-specific treatment plans.


Subject(s)
Hematologic Neoplasms/immunology , Lymphoid Tissue/immunology , Tissue Engineering , Animals , Biomimetic Materials , Cellular Microenvironment , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Humans , In Vitro Techniques , Models, Animal , Tumor Microenvironment
14.
Acta Biomater ; 53: 29-45, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28159716

ABSTRACT

Organogenesis and morphogenesis have informed our understanding of physiology, pathophysiology, and avenues to create new curative and regenerative therapies. Thus far, this understanding has been hindered by the lack of a physiologically relevant yet accessible model that affords biological control. Recently, three-dimensional ex vivo cellular cultures created through cellular self-assembly under natural extracellular matrix cues or through biomaterial-based directed assembly have been shown to physically resemble and recapture some functionality of target organs. These "organoids" have garnered momentum for their applications in modeling human development and disease, drug screening, and future therapy design or even organ replacement. This review first discusses the self-organizing organoids as materials with emergent properties and their advantages and limitations. We subsequently describe biomaterials-based strategies used to afford more control of the organoid's microenvironment and ensuing cellular composition and organization. In this review, we also offer our perspective on how multifunctional biomaterials with precise spatial and temporal control could ultimately bridge the gap between in vitro organoid platforms and their in vivo counterparts. STATEMENT OF SIGNIFICANCE: Several notable reviews have highlighted PSC-derived organoids and 3D aggregates, including embryoid bodies, from a development and cellular assembly perspective. The focus of this review is to highlight the materials-based approaches that cells, including PSCs and others, adopt for self-assembly and the controlled development of complex tissues, such as that of the brain, gut, and immune system.


Subject(s)
Biocompatible Materials , Models, Biological , Organogenesis , Organoids/cytology , Organoids/growth & development , Animals , Brain/cytology , Brain/growth & development , Cell Aggregation , Cell Culture Techniques , Cellular Microenvironment , Humans , Intestines/cytology , Intestines/growth & development , Lymphoid Tissue/cytology , Lymphoid Tissue/growth & development , Materials Testing , Regeneration , Spheroids, Cellular/cytology , Tissue Engineering/methods
15.
Macromol Biosci ; 17(6)2017 06.
Article in English | MEDLINE | ID: mdl-28207187

ABSTRACT

In arterial tissue engineering, mimicking native structure and mechanical properties is essential because compliance mismatch can lead to graft failure and further disease. With bottom-up tissue engineering approaches, designing tissue components with proper microscale mechanical properties is crucial to achieve the necessary macroscale properties in the final implant. This study develops a thermoresponsive cell culture platform for growing aligned vascular smooth muscle cell (VSMC) sheets by photografting N-isopropylacrylamide (NIPAAm) onto micropatterned poly(dimethysiloxane) (PDMS). The grafting process is experimentally and computationally optimized to produce PNIPAAm-PDMS substrates optimal for VSMC attachment. To allow long-term VSMC sheet culture and increase the rate of VSMC sheet formation, PNIPAAm-PDMS surfaces were further modified with 3-aminopropyltriethoxysilane yielding a robust, thermoresponsive cell culture platform for culturing VSMC sheets. VSMC cell sheets cultured on patterned thermoresponsive substrates exhibit cellular and collagen alignment in the direction of the micropattern. Mechanical characterization of patterned, single-layer VSMC sheets reveals increased stiffness in the aligned direction compared to the perpendicular direction whereas nonpatterned cell sheets exhibit no directional dependence. Structural and mechanical anisotropy of aligned, single-layer VSMC sheets makes this platform an attractive microstructural building block for engineering a vascular graft to match the in vivo mechanical properties of native arterial tissue.


Subject(s)
Arteries/growth & development , Cell Culture Techniques , Muscle, Smooth, Vascular/growth & development , Tissue Engineering , Acrylamides/chemistry , Acrylamides/therapeutic use , Acrylic Resins/chemistry , Acrylic Resins/therapeutic use , Anisotropy , Arteries/drug effects , Arteries/physiopathology , Dimethylpolysiloxanes/chemistry , Dimethylpolysiloxanes/therapeutic use , Humans , Muscle, Smooth, Vascular/drug effects , Nylons/chemistry , Surface Properties , Temperature
16.
ACS Biomater Sci Eng ; 3(2): 214-225, 2017 Feb 13.
Article in English | MEDLINE | ID: mdl-33450794

ABSTRACT

Germinal centers are dynamic structures within lymphoid tissues, which develop once B cells receive activating signals from surrounding immune cells. Germinal center B cells are small in number, heterogeneous, and prone to rapid apoptosis unless selected by the body to form memory B cells. Despite extensive research in the B cell differentiation process, the role of the lymphoid niche, in particular integrin ligands, in the development of early germinal center-like phenotype remains unclear. Here, we report a biomaterials-based modular immune organoid that enables development of early germinal-center phenotype in an integrin ligand-specific manner. We demonstrate the differential role of integrin α4ß1- and αvß3-binding ligands in the induction of GL7+ (GC-like) and GL7- (non-GC-like) phenotype in differentiating B cells while in the presence of CD40 ligand and interleukin-4. We further demonstrate the role of integrin ligand specificities in clustering of ß3 integrin and B cell receptor on the surface of differentiated B cells in 3D organoids as compared to the classic 2D cocultures. The study demonstrates that biomaterials-based immune organoids represent an ex vivo platform technology, which recapitulates certain aspects of GC biology to understand the process of B cell differentiation and induction of immunological responses. This platform is particularly useful in understanding the role of selective biomolecular signals and the temporal dependency of immune responses to these signals.

SELECTION OF CITATIONS
SEARCH DETAIL
...