1
|
Geib K, Scharf S, Schäfer H, Hartmann S, Hansmann ML, Wurzel P. 3D examination reveals increased destruction of alpha-actin-positive structures in advanced follicular lymphoma stages. Acta Histochem 2025; 127:152232. [PMID: 39883976 DOI: 10.1016/j.acthis.2025.152232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 01/07/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Follicular lymphoma (FL) represents the most prevalent subtype of non-Hodgkin's-lymphoma in Western Europe and the United States. While the examination of two-dimensional histological slides remains the gold standard method for diagnosing FL stages, three-dimensional analysis provides additional insights, particularly regarding cellular morphology, spatial relationships and network connectivity. This investigation assessed the tumor-related morphological destruction of fibroreticular cell (FRC) networks bordering germinal centres in FL. A confocal laser scanning technology and a digital three-dimensional analysis system were used. Quantitive measurements included the length of fibroblastic reticular walls surrounding the germinal centres as well as the size of the gaps and holes within these structures. Three-dimensional analysis revealed progressive structural degradation and a reduction in mechanical barrier integrity, with differences observed between low- and high-grade FL. High-grade FL exhibited greater network destruction. Fibroblastic reticular cell networks' wall length demonstrated a consistent decline across all grades. The lengths of these walls and wall-like structures in FL grades 1 or 2 were similar to reactive germinal centres seen in lymphadenitis, as well as the gap size. The gaps are thought to be responsible for B- and T-cell exchange. This work demonstrated the massive destruction of neoplastic germinal centres in grades 3a and 3b FL. In grade 3b, this was accompanied by a likely dysfunctional mechanical border of the germinal centre and the near-complete loss of structural integrity. Under physiological conditions, gaps and holes regulate lymphoid traffic. Under reactive conditions, only a few specific T-cells can access the germinal centre. Under neoplastic conditions, the diameter of these gaps increases as grades increase, culminating in complete structural disruption in grade 3b. The mechanical destruction was found to begin at one pole of the germinal centre, as evidenced by localized decay and fragmentation of FRC walls on one side. Fibroblastic reticular cell networks are critical for maintaining chemokine gradients to ensure compartmentalisation of lymphoid structures. Their ongoing degradation in FL of the networks leads to a morphological loss of function. This is due to the blurring of various lymph node zones.
Collapse
Affiliation(s)
- Katharina Geib
- Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Sonja Scharf
- Department of Molecular Bioinformatics, Institute of Computer Science, Johann Wolfgang, Goethe-University, Frankfurt/Main, Hessen, Germany; Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany
| | - Hendrik Schäfer
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt/Main, Hessen, Germany; Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany
| | - Sylvia Hartmann
- Hospital of the Goethe University Frankfurt, Department of Pathology, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany
| | - Martin-Leo Hansmann
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt/Main, Hessen, Germany; Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany
| | - Patrick Wurzel
- Department of Molecular Bioinformatics, Institute of Computer Science, Johann Wolfgang, Goethe-University, Frankfurt/Main, Hessen, Germany; Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany
| |
Collapse
|
2
|
Moghe P, Belousov R, Ichikawa T, Iwatani C, Tsukiyama T, Erzberger A, Hiiragi T. Coupling of cell shape, matrix and tissue dynamics ensures embryonic patterning robustness. Nat Cell Biol 2025; 27:408-423. [PMID: 39966670 PMCID: PMC11906357 DOI: 10.1038/s41556-025-01618-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/20/2024] [Indexed: 02/20/2025]
Abstract
Tissue patterning coordinates morphogenesis, cell dynamics and fate specification. Understanding how precision in patterning is robustly achieved despite inherent developmental variability during mammalian embryogenesis remains a challenge. Here, based on cell dynamics quantification and simulation, we show how salt-and-pepper epiblast and primitive endoderm (PrE) cells pattern the inner cell mass of mouse blastocysts. Coupling cell fate and dynamics, PrE cells form apical polarity-dependent actin protrusions required for RAC1-dependent migration towards the surface of the fluid cavity, where PrE cells are trapped due to decreased tension. Concomitantly, PrE cells deposit an extracellular matrix gradient, presumably breaking the tissue-level symmetry and collectively guiding their own migration. Tissue size perturbations of mouse embryos and their comparison with monkey and human blastocysts further demonstrate that the fixed proportion of PrE/epiblast cells is optimal with respect to embryo size and tissue geometry and, despite variability, ensures patterning robustness during early mammalian development.
Collapse
Grants
- The Hiiragi laboratory was supported by the EMBL, and currently by the Hubrecht Institute, the European Research Council (ERC Advanced Grant “SelforganisingEmbryo” grant agreement 742732, ERC Advanced Grant “COORDINATION” grant agreement 101055287), Stichting LSH-TKI (LSHM21020), and Japan Society for the Promotion of Science (JSPS) KAKENHI grant numbers JP21H05038 and JP22H05166. The Erzberger laboratory is supported by the EMBL.
- European Molecular Biology Laboratory (EMBL Heidelberg)
- MEXT | Japan Society for the Promotion of Science (JSPS)
- T.I. was supported by the JSPS Overseas Research Fellowship
- The Erzberger laboratory is supported by the EMBL.
- The Hiiragi laboratory was supported by the EMBL, and currently by the Hubrecht Institute, the European Research Council (ERC Advanced Grant “SelforganisingEmbryo” grant agreement 742732, ERC Advanced Grant “COORDINATION” grant agreement 101055287), Stichting LSH-TKI (LSHM21020), and Japan Society for the Promotion of Science (JSPS) KAKENHI grant numbers JP21H05038 and JP22H05166.
Collapse
Affiliation(s)
- Prachiti Moghe
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Utrecht, Netherlands
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Roman Belousov
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Takafumi Ichikawa
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chizuru Iwatani
- Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan
| | - Tomoyuki Tsukiyama
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan
| | - Anna Erzberger
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Takashi Hiiragi
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Utrecht, Netherlands.
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan.
- Department of Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
3
|
Zhang-Zhou J, Movilla Meno N, Oñate Salafranca C, Gomez-Benito MJ, Guerrero PE, Pardo Jimeno J, García-Aznar JM. CAR-T cells are more affected than T lymphocytes by mechanical constraints: A microfluidic-based approach. Life Sci 2025; 363:123335. [PMID: 39732363 DOI: 10.1016/j.lfs.2024.123335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/31/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
AIMS CAR-T cell therapy has attracted considerable attention in recent years owing to its well-known efficacy against haematopoietic malignancies. Nevertheless, this immunotherapy fails against solid tumours due to hostile conditions found in the tumour microenvironment. In this context, many relevant biochemical factors have been thoroughly studied, but crucial mechanical cues have been underestimated. MAIN METHODS We developed an innovative approach using microfluidic devices, which recreate the biomechanical aspects of solid tumours. Using these platforms, we quantified immune cell migration (T and CAR-T cells) under different confinement conditions. KEY FINDINGS We found that both CAR-T cell and T cell velocities are affected by the biomechanical and chemical cues studied, which are closely related to those found in solid tumours. Under biochemical stimulus-free conditions, the velocity of T cells is independent of the width of the microchannel, whereas the velocity of CAR-T cells is greatly reduced in the highest confinement channels. When chemoattractants or tumour cells are added, immune cells display increased confined migration velocity. However, in the presence of immunosuppressive chemokines, T cells become slower, whereas CAR-T cells significantly increase their velocity via a chimeric cytokine receptor. SIGNIFICANCE Our approach contributes to a better understanding of immune cell migration and the influence of mechanical constraints, which will allow the testing of new ways to improve CAR-T cell trafficking into solid tumours. Therefore, our study revealed that the migratory behaviour of CAR-T cells differs from that of T cells under confined conditions and that biomechanical cues, such as cell deformability caused by confinement, can influence the correct infiltration of immune cells into solid tumours during the immune response.
Collapse
Affiliation(s)
- Jack Zhang-Zhou
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain.
| | - Nieves Movilla Meno
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain; Instituto de Investigación en Ingeniería de Aragón (I3A), Zaragoza, Spain.
| | | | - Maria Jose Gomez-Benito
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain; Instituto de Investigación en Ingeniería de Aragón (I3A), Zaragoza, Spain; Aragón Institute of Healthcare Research (IIS Aragón), Zaragoza, Spain.
| | - Pedro Enrique Guerrero
- Instituto de Investigación en Ingeniería de Aragón (I3A), Zaragoza, Spain; Department of Biochemistry and Molecular and Cellular Biology, University of Zaragoza, Zaragoza, Spain; Aragón Institute of Healthcare Research (IIS Aragón), Zaragoza, Spain.
| | - Julian Pardo Jimeno
- Faculty of Medicine, University of Zaragoza/IIS, Aragon, Spain; CIBER of Infectious diseases, IS Carlos III, Madrid, Spain; Aragón Institute of Healthcare Research (IIS Aragón), Zaragoza, Spain.
| | - Jose Manuel García-Aznar
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain; Instituto de Investigación en Ingeniería de Aragón (I3A), Zaragoza, Spain; Aragón Institute of Healthcare Research (IIS Aragón), Zaragoza, Spain.
| |
Collapse
|
4
|
Otterpohl KL, Busselman BW, Zimmerman JL, Mukherjee M, Evans C, Graber K, Thakkar VP, Johnston JG, Ilyas A, Gumz ML, Eaton DC, Sands JM, Surendran K, Chandrasekar I. Thick Ascending Limb Specific Inactivation of Myh9 and Myh10 Myosin Motors Results in Progressive Kidney Disease and Drives Sex-specific Cellular Adaptation in the Distal Nephron and Collecting Duct. FUNCTION 2025; 6:zqae048. [PMID: 39500539 PMCID: PMC11815580 DOI: 10.1093/function/zqae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
Our previous work established a role for myosin motor proteins MYH9 and MYH10 in trafficking of thick ascending limb (TAL) cargoes uromodulin and Na+-K+-2Cl- cotransporter NKCC2. We have generated a TAL-specific Myh9&10 conditional knockout (Myh9&10 TAL-cKO) mouse model to determine the cell autonomous roles for MYH9&10 in TAL cargo trafficking and to understand the consequence of TAL dysfunction in adult kidney. Myh9&10 TAL-cKO mice develop progressive kidney disease with pathological tubular injury confirmed by histological changes, tubular injury markers, upregulated endoplasmic reticulum (ER) stress/unfolded protein response, and higher blood urea nitrogen and serum creatinine. However, male mice survive twice as long as female mice. We have determined this sexual dimorphism in morbidity is due to adaptation of the distal nephron and collecting duct in response to TAL dysfunction and lower NKCC2 expression. We demonstrate that this triggers a compensatory mechanism involving sex-specific cellular adaptation within the distal nephron and collecting duct to boost sodium reabsorption. While both sexes overcompensate by activating epithelial sodium channel (ENaC) expression in medullary collecting ducts resulting in hypernatremia, this is initially subdued in male Myh9&10 TAL-cKO mice through higher sodium chloride cotransporter (NCC) expression within the distal nephron. Our results indicate that compromised TAL function ultimately results in maladaptation of medullary collecting duct cells which acquire cortical-like properties including ENaC expression. This work further confirms a cell autonomous role for MYH9&10 in maintenance of NKCC2 expression in the TAL and uncover distal nephron and collecting duct adaptive mechanisms which respond to TAL dysfunction.
Collapse
Affiliation(s)
- Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Brook W Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, SD 57069, USA
| | - Jenna L Zimmerman
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Malini Mukherjee
- Functional Genomics and Bioinformatics Core, Sanford Research, Sioux Falls, SD 57104, USA
| | - Claire Evans
- Histology and Imaging Core, Sanford Research, Sioux Falls, SD 57104, USA
| | - Kelly Graber
- Histology and Imaging Core, Sanford Research, Sioux Falls, SD 57104, USA
| | - Vedant P Thakkar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Jermaine G Johnston
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Arooba Ilyas
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, SD 57069, USA
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Douglas C Eaton
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA
| | - Jeff M Sands
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, SD 57103, USA
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, SD 57103, USA
| |
Collapse
|
5
|
Wilander BA, Harris TL, Mandarano AH, Guy CS, Prater MS, Pruett-Miller SM, Ogden SK, McGargill MA. DRAK2 regulates myosin light chain phosphorylation in T cells. J Cell Sci 2024; 137:jcs261813. [PMID: 39421891 DOI: 10.1242/jcs.261813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Death-associated protein kinase-related apoptosis-inducing kinase-2 (DRAK2; also known as STK17B) is a serine/threonine kinase expressed in T cells. Drak2-deficient (Drak2-/-) mice respond effectively to tumors and pathogens while displaying resistance to T cell-mediated autoimmune disease. However, the molecular mechanisms by which DRAK2 impacts T cell function remain unclear. Gaining further insight into the function of DRAK2 in T cells will shed light on differentially regulated pathways in autoreactive and pathogen-specific T cells, which is crucial for improving autoimmune therapies. Here, we demonstrate that DRAK2 contributes to activation of myosin light chain (MLC2, encoded by Myl2) in both murine and human T cells. In the absence of Drak2, the amount of polymerized actin was decreased, suggesting that DRAK2 modulates actomyosin dynamics. We further show that myosin-dependent T cell functions, such as migration, T cell receptor microcluster accumulation, and conjugation to antigen presenting cells are decreased in the absence of Drak2. These findings reveal that DRAK2 plays an important role in regulating MLC activation within T cells.
Collapse
Affiliation(s)
- Benjamin A Wilander
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Tarsha L Harris
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alexandra H Mandarano
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Cliff S Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mollie S Prater
- The Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- The Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Maureen A McGargill
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
6
|
Fabrèges D, Corominas-Murtra B, Moghe P, Kickuth A, Ichikawa T, Iwatani C, Tsukiyama T, Daniel N, Gering J, Stokkermans A, Wolny A, Kreshuk A, Duranthon V, Uhlmann V, Hannezo E, Hiiragi T. Temporal variability and cell mechanics control robustness in mammalian embryogenesis. Science 2024; 386:eadh1145. [PMID: 39388574 DOI: 10.1126/science.adh1145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 10/02/2023] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
How living systems achieve precision in form and function despite their intrinsic stochasticity is a fundamental yet ongoing question in biology. We generated morphomaps of preimplantation embryogenesis in mouse, rabbit, and monkey embryos, and these morphomaps revealed that although blastomere divisions desynchronized passively, 8-cell embryos converged toward robust three-dimensional shapes. Using topological analysis and genetic perturbations, we found that embryos progressively changed their cellular connectivity to a preferred topology, which could be predicted by a physical model in which actomyosin contractility and noise facilitate topological transitions, lowering surface energy. This mechanism favored regular embryo packing and promoted a higher number of inner cells in the 16-cell embryo. Synchronized division reduced embryo packing and generated substantially more misallocated cells and fewer inner-cell-mass cells. These findings suggest that stochasticity in division timing contributes to robust patterning.
Collapse
Affiliation(s)
- Dimitri Fabrèges
- Hubrecht Institute, Utrecht, Netherlands
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Prachiti Moghe
- Hubrecht Institute, Utrecht, Netherlands
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Alison Kickuth
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Takafumi Ichikawa
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chizuru Iwatani
- Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan
| | - Tomoyuki Tsukiyama
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan
| | - Nathalie Daniel
- UVSQ, INRAE, BREED, Paris-Saclay University, Jouy-en-Josas, France
| | | | | | - Adrian Wolny
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Anna Kreshuk
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Véronique Duranthon
- UVSQ, INRAE, BREED, Paris-Saclay University, Jouy-en-Josas, France
- École Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | | | - Edouard Hannezo
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Takashi Hiiragi
- Hubrecht Institute, Utrecht, Netherlands
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
7
|
Schliffka MF, Dumortier JG, Pelzer D, Mukherjee A, Maître JL. Inverse blebs operate as hydraulic pumps during mouse blastocyst formation. Nat Cell Biol 2024; 26:1669-1677. [PMID: 39261717 DOI: 10.1038/s41556-024-01501-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
During preimplantation development, mouse embryos form a fluid-filled lumen. Pressurized fluid fractures cell-cell contacts and accumulates into pockets, which coarsen into a single lumen. How the embryo controls intercellular fluid movement during coarsening is unknown. Here we report inverse blebs growing into cells at adhesive contacts. Throughout the embryo we observed hundreds of inverse blebs, each filling with intercellular fluid and retracting within a minute. Inverse blebs grow due to pressure build-up resulting from fluid accumulation and cell-cell adhesion, which locally confines fluid. Inverse blebs retract due to actomyosin contraction, practically pushing fluid within the intercellular space. Importantly, inverse blebs occur infrequently at contacts formed by multiple cells, which effectively serve as fluid sinks. Manipulation of the embryo topology reveals that without sinks inverse blebs pump fluid into one another in futile cycles. We propose that inverse blebs operate as hydraulic pumps to promote luminal coarsening, thereby constituting an instrument used by cells to control fluid movement.
Collapse
Affiliation(s)
- Markus F Schliffka
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University and Sorbonne Université, Paris, France
- Carl Zeiss SAS, Marly-le-Roy, France
| | - Julien G Dumortier
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University and Sorbonne Université, Paris, France
| | - Diane Pelzer
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University and Sorbonne Université, Paris, France
| | - Arghyadip Mukherjee
- Laboratoire de physique de l'École Normale Supérieure, CNRS UMR 8023, PSL Research University, Sorbonne Université and Université Paris Cité, Paris, France.
| | - Jean-Léon Maître
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University and Sorbonne Université, Paris, France.
| |
Collapse
|
8
|
Campos J, Osorio-Barrios F, Villanelo F, Gutierrez-Maldonado SE, Vargas P, Pérez-Acle T, Pacheco R. Chemokinergic and Dopaminergic Signalling Collaborates through the Heteromer Formed by CCR9 and Dopamine Receptor D5 Increasing the Migratory Speed of Effector CD4 + T-Cells to Infiltrate the Colonic Mucosa. Int J Mol Sci 2024; 25:10022. [PMID: 39337509 PMCID: PMC11432204 DOI: 10.3390/ijms251810022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Inflammatory bowel diseases (IBDs) involve chronic inflammation of the gastrointestinal tract, where effector CD4+ T-cells play a central role. Thereby, the recruitment of T-cells into the colonic mucosa represents a key process in IBD. We recently found that CCR9 and DRD5 might form a heteromeric complex on the T-cell surface. The increase in CCL25 production and the reduction in dopamine levels associated with colonic inflammation represent a dual signal stimulating the CCR9:DRD5 heteromer, which promotes the recruitment of CD4+ T-cells into the colonic lamina propria. Here, we aimed to analyse the molecular requirements involved in the heteromer assembly as well as to determine the underlying cellular mechanisms involved in the colonic tropism given by the stimulation of the CCR9:DRD5 complex. The results show that dual stimulation of the CCR9:DRD5 heteromer potentiates the phosphorylation of the myosin light chain 2 (MLC2) and the migration speed in confined microchannels. Accordingly, disrupting the CCR9:DRD5 assembly induced a sharp reduction in the pMLC2 in vitro, decreased the migratory speed in confined microchannels, and dampened the recruitment of CD4+ T-cells into the inflamed colonic mucosa. Furthermore, in silico analysis confirmed that the interface of interaction of CCR9:DRD5 is formed by the transmembrane segments 5 and 6 from each protomer. Our findings demonstrated that the CCR9:DRD5 heteromeric complex plays a fundamental role in the migration of CD4+ T-cells into the colonic mucosa upon inflammation. Thereby, the present study encourages the design of strategies for disassembling the formation of the CCR9:DRD5 as a therapeutic opportunity to treat IBD.
Collapse
Affiliation(s)
- Javier Campos
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
| | - Francisco Osorio-Barrios
- Gut Microbiology, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 25, 3001 Bern, Switzerland
| | - Felipe Villanelo
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
- Escuela de Ingeniería, Facultad de Ingeniería Arquitectura y Diseño, Universidad San Sebastián, Recoleta 8420524, Santiago, Chile
| | - Sebastian E. Gutierrez-Maldonado
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
- Escuela de Ingeniería, Facultad de Ingeniería Arquitectura y Diseño, Universidad San Sebastián, Recoleta 8420524, Santiago, Chile
| | - Pablo Vargas
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005 Paris, France;
- Université Paris Cité, CNRS, INSERM, Inserm, INEM, F-75015 Paris, France
| | - Tomás Pérez-Acle
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
- Escuela de Ingeniería, Facultad de Ingeniería Arquitectura y Diseño, Universidad San Sebastián, Recoleta 8420524, Santiago, Chile
| | - Rodrigo Pacheco
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510157, Santiago, Chile
| |
Collapse
|
9
|
Rezvan A, Romain G, Fathi M, Heeke D, Martinez-Paniagua M, An X, Bandey IN, Montalvo MJ, Adolacion JRT, Saeedi A, Sadeghi F, Fousek K, Puebla-Osorio N, Cooper LJN, Bernatchez C, Singh H, Ahmed N, Mattie M, Bot A, Neelapu S, Varadarajan N. Identification of a clinically efficacious CAR T cell subset in diffuse large B cell lymphoma by dynamic multidimensional single-cell profiling. NATURE CANCER 2024; 5:1010-1023. [PMID: 38750245 DOI: 10.1038/s43018-024-00768-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 04/10/2024] [Indexed: 05/24/2024]
Abstract
Chimeric antigen receptor (CAR) T cells used for the treatment of B cell malignancies can identify T cell subsets with superior clinical activity. Here, using infusion products of individuals with large B cell lymphoma, we integrated functional profiling using timelapse imaging microscopy in nanowell grids with subcellular profiling and single-cell RNA sequencing to identify a signature of multifunctional CD8+ T cells (CD8-fit T cells). CD8-fit T cells are capable of migration and serial killing and harbor balanced mitochondrial and lysosomal volumes. Using independent datasets, we validate that CD8-fit T cells (1) are present premanufacture and are associated with clinical responses in individuals treated with axicabtagene ciloleucel, (2) longitudinally persist in individuals after treatment with CAR T cells and (3) are tumor migrating cytolytic cells capable of intratumoral expansion in solid tumors. Our results demonstrate the power of multimodal integration of single-cell functional assessments for the discovery and application of CD8-fit T cells as a T cell subset with optimal fitness in cell therapy.
Collapse
Affiliation(s)
- Ali Rezvan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Gabrielle Romain
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | | | | | | | - Xingyue An
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Irfan N Bandey
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Melisa J Montalvo
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Jay R T Adolacion
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Arash Saeedi
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Fatemeh Sadeghi
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Kristen Fousek
- Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Nahum Puebla-Osorio
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Chantale Bernatchez
- Department of Biologics Development, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Harjeet Singh
- Divsion of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nabil Ahmed
- Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Mike Mattie
- Kite, a Gilead Company, Santa Monica, CA, USA
| | - Adrian Bot
- Kite, a Gilead Company, Santa Monica, CA, USA
| | - Sattva Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Navin Varadarajan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA.
| |
Collapse
|
10
|
Huycke TR, Häkkinen TJ, Miyazaki H, Srivastava V, Barruet E, McGinnis CS, Kalantari A, Cornwall-Scoones J, Vaka D, Zhu Q, Jo H, Oria R, Weaver VM, DeGrado WF, Thomson M, Garikipati K, Boffelli D, Klein OD, Gartner ZJ. Patterning and folding of intestinal villi by active mesenchymal dewetting. Cell 2024; 187:3072-3089.e20. [PMID: 38781967 PMCID: PMC11166531 DOI: 10.1016/j.cell.2024.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/30/2023] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Tissue folds are structural motifs critical to organ function. In the intestine, bending of a flat epithelium into a periodic pattern of folds gives rise to villi, finger-like protrusions that enable nutrient absorption. However, the molecular and mechanical processes driving villus morphogenesis remain unclear. Here, we identify an active mechanical mechanism that simultaneously patterns and folds the intestinal epithelium to initiate villus formation. At the cellular level, we find that PDGFRA+ subepithelial mesenchymal cells generate myosin II-dependent forces sufficient to produce patterned curvature in neighboring tissue interfaces. This symmetry-breaking process requires altered cell and extracellular matrix interactions that are enabled by matrix metalloproteinase-mediated tissue fluidization. Computational models, together with in vitro and in vivo experiments, revealed that these cellular features manifest at the tissue level as differences in interfacial tensions that promote mesenchymal aggregation and interface bending through a process analogous to the active dewetting of a thin liquid film.
Collapse
Affiliation(s)
- Tyler R Huycke
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Teemu J Häkkinen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Hikaru Miyazaki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Emilie Barruet
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Christopher S McGinnis
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Ali Kalantari
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jake Cornwall-Scoones
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Dedeepya Vaka
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Qin Zhu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Roger Oria
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Krishna Garikipati
- Departments of Mechanical Engineering, and Mathematics, University of Michigan, Ann Arbor, MI, USA
| | - Dario Boffelli
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA.
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
11
|
Du W, Verma A, Ye Q, Du W, Lin S, Yamanaka A, Klein OD, Hu JK. Myosin II mediates Shh signals to shape dental epithelia via control of cell adhesion and movement. PLoS Genet 2024; 20:e1011326. [PMID: 38857279 PMCID: PMC11192418 DOI: 10.1371/journal.pgen.1011326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 06/21/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024] Open
Abstract
The development of ectodermal organs begins with the formation of a stratified epithelial placode that progressively invaginates into the underlying mesenchyme as the organ takes its shape. Signaling by secreted molecules is critical for epithelial morphogenesis, but how that information leads to cell rearrangement and tissue shape changes remains an open question. Using the mouse dentition as a model, we first establish that non-muscle myosin II is essential for dental epithelial invagination and show that it functions by promoting cell-cell adhesion and persistent convergent cell movements in the suprabasal layer. Shh signaling controls these processes by inducing myosin II activation via AKT. Pharmacological induction of AKT and myosin II can also rescue defects caused by the inhibition of Shh. Together, our results support a model in which the Shh signal is transmitted through myosin II to power effective cellular rearrangement for proper dental epithelial invagination.
Collapse
Affiliation(s)
- Wei Du
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Adya Verma
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Qianlin Ye
- School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Wen Du
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Sandy Lin
- School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Atsushi Yamanaka
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Ophir D. Klein
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Jimmy K. Hu
- School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
12
|
Su T, Shen H, He M, Yang S, Gong X, Huang C, Guo L, Wang H, Feng S, Mi T, Zhao M, Liu Q, Huo F, Zhu JK, Zhu J, Li H, Liu H. Quercetin promotes the proportion and maturation of NK cells by binding to MYH9 and improves cognitive functions in aged mice. Immun Ageing 2024; 21:29. [PMID: 38730291 PMCID: PMC11084035 DOI: 10.1186/s12979-024-00436-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Quercetin is a flavonol compound widely distributed in plants that possesses diverse biological properties, including antioxidative, anti-inflammatory, anticancer, neuroprotective and senescent cell-clearing activities. It has been shown to effectively alleviate neurodegenerative diseases and enhance cognitive functions in various models. The immune system has been implicated in the regulation of brain function and cognitive abilities. However, it remains unclear whether quercetin enhances cognitive functions by interacting with the immune system. RESULTS In this study, middle-aged female mice were administered quercetin via tail vein injection. Quercetin increased the proportion of NK cells, without affecting T or B cells, and improved cognitive performance. Depletion of NK cells significantly reduces cognitive ability in mice. RNA-seq analysis revealed that quercetin modulated the RNA profile of hippocampal tissues in aging animals towards a more youthful state. In vitro, quercetin significantly inhibited the differentiation of Lin-CD117+ hematopoietic stem cells into NK cells. Furthermore, quercetin promoted the proportion and maturation of NK cells by binding to the MYH9 protein. CONCLUSIONS In summary, our findings suggest that quercetin promotes the proportion and maturation of NK cells by binding to the MYH9 protein, thereby improving cognitive performance in middle-aged mice.
Collapse
Affiliation(s)
- Tingting Su
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, 832003, China
| | - Haitao Shen
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, 832003, China
| | - Mengyuan He
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, 832003, China
| | - Shanshan Yang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Xue Gong
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, 832003, China
| | - Ce Huang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Liuling Guo
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Hao Wang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Shengyu Feng
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Taotao Mi
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Meili Zhao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Qing Liu
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Fengjiao Huo
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Jian-Kang Zhu
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jianbo Zhu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, 832003, China.
| | - Hongbin Li
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, 832003, China.
| | - Hailiang Liu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, 832003, China.
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China.
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
13
|
Johnston AC, Alicea GM, Lee CC, Patel PV, Hanna EA, Vaz E, Forjaz A, Wan Z, Nair PR, Lim Y, Chen T, Du W, Kim D, Nichakawade TD, Rebecca VW, Bonifant CL, Fan R, Kiemen AL, Wu PH, Wirtz D. Engineering self-propelled tumor-infiltrating CAR T cells using synthetic velocity receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.13.571595. [PMID: 38168186 PMCID: PMC10760159 DOI: 10.1101/2023.12.13.571595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Chimeric antigen receptor (CAR) T cells express antigen-specific synthetic receptors, which upon binding to cancer cells, elicit T cell anti-tumor responses. CAR T cell therapy has enjoyed success in the clinic for hematological cancer indications, giving rise to decade-long remissions in some cases. However, CAR T therapy for patients with solid tumors has not seen similar success. Solid tumors constitute 90% of adult human cancers, representing an enormous unmet clinical need. Current approaches do not solve the central problem of limited ability of therapeutic cells to migrate through the stromal matrix. We discover that T cells at low and high density display low- and high-migration phenotypes, respectively. The highly migratory phenotype is mediated by a paracrine pathway from a group of self-produced cytokines that include IL5, TNFα, IFNγ, and IL8. We exploit this finding to "lock-in" a highly migratory phenotype by developing and expressing receptors, which we call velocity receptors (VRs). VRs target these cytokines and signal through these cytokines' cognate receptors to increase T cell motility and infiltrate lung, ovarian, and pancreatic tumors in large numbers and at doses for which control CAR T cells remain confined to the tumor periphery. In contrast to CAR therapy alone, VR-CAR T cells significantly attenuate tumor growth and extend overall survival. This work suggests that approaches to the design of immune cell receptors that focus on migration signaling will help current and future CAR cellular therapies to infiltrate deep into solid tumors.
Collapse
Affiliation(s)
- Adrian C Johnston
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | | | - Cameron C Lee
- Department of Biomedical Engineering, Johns Hopkins University
| | - Payal V Patel
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Eban A Hanna
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Eduarda Vaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - André Forjaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Zeqi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University
| | - Praful R Nair
- Institute for NanoBioTechnology, Johns Hopkins University
| | - Yeongseo Lim
- Department of Biomedical Engineering, Johns Hopkins University
| | - Tina Chen
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Wenxuan Du
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Dongjoo Kim
- Department of Biomedical Engineering, Yale University
| | - Tushar D Nichakawade
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University
| | - Vito W Rebecca
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University
| | - Challice L Bonifant
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University
| | - Rong Fan
- Department of Biomedical Engineering, Yale University
| | - Ashley L Kiemen
- Institute for NanoBioTechnology, Johns Hopkins University
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University
| | - Pei-Hsun Wu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
| | - Denis Wirtz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University
- Department of Oncology, Johns Hopkins School of Medicine, Johns Hopkins University
| |
Collapse
|
14
|
Ruef N, Martínez Magdaleno J, Ficht X, Purvanov V, Palayret M, Wissmann S, Pfenninger P, Stolp B, Thelen F, Barreto de Albuquerque J, Germann P, Sharpe J, Abe J, Legler DF, Stein JV. Exocrine gland-resident memory CD8 + T cells use mechanosensing for tissue surveillance. Sci Immunol 2023; 8:eadd5724. [PMID: 38134242 DOI: 10.1126/sciimmunol.add5724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/09/2023] [Indexed: 12/24/2023]
Abstract
Tissue-resident CD8+ T cells (TRM) continuously scan peptide-MHC (pMHC) complexes in their organ of residence to intercept microbial invaders. Recent data showed that TRM lodged in exocrine glands scan tissue in the absence of any chemoattractant or adhesion receptor signaling, thus bypassing the requirement for canonical migration-promoting factors. The signals eliciting this noncanonical motility and its relevance for organ surveillance have remained unknown. Using mouse models of viral infections, we report that exocrine gland TRM autonomously generated front-to-back F-actin flow for locomotion, accompanied by high cortical actomyosin contractility, and leading-edge bleb formation. The distinctive mode of exocrine gland TRM locomotion was triggered by sensing physical confinement and was closely correlated with nuclear deformation, which acts as a mechanosensor via an arachidonic acid and Ca2+ signaling pathway. By contrast, naïve CD8+ T cells or TRM surveilling microbe-exposed epithelial barriers did not show mechanosensing capacity. Inhibition of nuclear mechanosensing disrupted exocrine gland TRM scanning and impaired their ability to intercept target cells. These findings indicate that confinement is sufficient to elicit autonomous T cell surveillance in glands with restricted chemokine expression and constitutes a scanning strategy that complements chemosensing-dependent migration.
Collapse
Affiliation(s)
- Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Jose Martínez Magdaleno
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Xenia Ficht
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 22, 4058 Basel, Switzerland
| | - Vladimir Purvanov
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, 8280 Kreuzlingen, Switzerland
| | - Matthieu Palayret
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Stefanie Wissmann
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Petra Pfenninger
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Bettina Stolp
- Department for Infectious Diseases, Integrative Virology, Center for Integrative Infectious Disease Research, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Flavian Thelen
- Department of Medical Oncology and Hematology, University of Zürich and University Hospital Zürich, 8091 Zürich, Switzerland
| | | | - Philipp Germann
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08003 Barcelona, Spain
| | - James Sharpe
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08003 Barcelona, Spain
- European Molecular Biology Laboratory (EMBL) Barcelona, 08003 Barcelona, Spain
- Institucio' Catalana de Recerca i Estudis Avancats (ICREA), 08010 Barcelona, Spain
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, 8280 Kreuzlingen, Switzerland
- Faculty of Biology, University of Konstanz, 78464 Konstanz, Germany
- Theodor Kocher Institute, University of Bern, 3011 Bern, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
15
|
Barbazan J, Pérez-González C, Gómez-González M, Dedenon M, Richon S, Latorre E, Serra M, Mariani P, Descroix S, Sens P, Trepat X, Vignjevic DM. Cancer-associated fibroblasts actively compress cancer cells and modulate mechanotransduction. Nat Commun 2023; 14:6966. [PMID: 37907483 PMCID: PMC10618488 DOI: 10.1038/s41467-023-42382-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/09/2023] [Indexed: 11/02/2023] Open
Abstract
During tumor progression, cancer-associated fibroblasts (CAFs) accumulate in tumors and produce an excessive extracellular matrix (ECM), forming a capsule that enwraps cancer cells. This capsule acts as a barrier that restricts tumor growth leading to the buildup of intratumoral pressure. Combining genetic and physical manipulations in vivo with microfabrication and force measurements in vitro, we found that the CAFs capsule is not a passive barrier but instead actively compresses cancer cells using actomyosin contractility. Abrogation of CAFs contractility in vivo leads to the dissipation of compressive forces and impairment of capsule formation. By mapping CAF force patterns in 3D, we show that compression is a CAF-intrinsic property independent of cancer cell growth. Supracellular coordination of CAFs is achieved through fibronectin cables that serve as scaffolds allowing force transmission. Cancer cells mechanosense CAF compression, resulting in an altered localization of the transcriptional regulator YAP and a decrease in proliferation. Our study unveils that the contractile capsule actively compresses cancer cells, modulates their mechanical signaling, and reorganizes tumor morphology.
Collapse
Affiliation(s)
- Jorge Barbazan
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005, Paris, France
- Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), 15706, Santiago de Compostela, Spain
| | | | - Manuel Gómez-González
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028, Barcelona, Spain
| | - Mathieu Dedenon
- Institut Curie, PSL Research University, CNRS UMR 168, F-75005, Paris, France
| | - Sophie Richon
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005, Paris, France
| | - Ernest Latorre
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028, Barcelona, Spain
| | - Marco Serra
- Institut Curie, PSL Research University, CNRS UMR 168, F-75005, Paris, France
| | - Pascale Mariani
- Institut Curie, Department of surgical oncology, Curie Institute, F-75005, Paris, France
| | - Stéphanie Descroix
- Institut Curie, PSL Research University, CNRS UMR 168, F-75005, Paris, France
| | - Pierre Sens
- Institut Curie, PSL Research University, CNRS UMR 168, F-75005, Paris, France
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028, Barcelona, Spain.
- Facutltat de Medicina, Universitat de Barcelona, 08036, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08028, Barcelona, Spain.
| | | |
Collapse
|
16
|
Gajardo T, Bernard M, Lô M, Turck E, Leveau C, El-Daher MT, Deslys A, Panikulam P, Menche C, Kurowska M, Le Lay G, Barbier L, Moshous D, Neven B, Farin HF, Fischer A, Ménasché G, de Saint Basile G, Vargas P, Sepulveda FE. Actin dynamics regulation by TTC7A/PI4KIIIα limits DNA damage and cell death under confinement. J Allergy Clin Immunol 2023; 152:949-960. [PMID: 37390900 DOI: 10.1016/j.jaci.2023.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND The actin cytoskeleton has a crucial role in the maintenance of the immune homeostasis by controlling various cellular processes, including cell migration. Mutations in TTC7A have been described as the cause of a primary immunodeficiency associated to different degrees of gut involvement and alterations in the actin cytoskeleton dynamics. OBJECTIVES This study investigates the impact of TTC7A deficiency in immune homeostasis. In particular, the role of the TTC7A/phosphatidylinositol 4 kinase type III α pathway in the control of leukocyte migration and actin dynamics. METHODS Microfabricated devices were leveraged to study cell migration and actin dynamics of murine and patient-derived leukocytes under confinement at the single-cell level. RESULTS We show that TTC7A-deficient lymphocytes exhibit an altered cell migration and reduced capacity to deform through narrow gaps. Mechanistically, TTC7A-deficient phenotype resulted from impaired phosphoinositide signaling, leading to the downregulation of the phosphoinositide 3-kinase/AKT/RHOA regulatory axis and imbalanced actin cytoskeleton dynamics. TTC7A-associated phenotype resulted in impaired cell motility, accumulation of DNA damage, and increased cell death in dense 3-dimensional gels in the presence of chemokines. CONCLUSIONS These results highlight a novel role of TTC7A as a critical regulator of lymphocyte migration. Impairment of this cellular function is likely to contribute to the pathophysiology underlying progressive immunodeficiency in patients.
Collapse
Affiliation(s)
- Tania Gajardo
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France
| | - Mathilde Bernard
- UMR 144, Institut Curie, Paris, France; Institut Pierre-Gilles de Gennes, Paris Sciences and Letters Research University, Paris, France
| | - Marie Lô
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France
| | - Elisa Turck
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France
| | - Claire Leveau
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France
| | - Marie-Thérèse El-Daher
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France
| | - Alexandre Deslys
- Leukomotion Lab, Université de Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, F-75015 Paris, France
| | - Patricia Panikulam
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France
| | - Constantin Menche
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Main, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Mathieu Kurowska
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France
| | - Gregoire Le Lay
- UMR 144, Institut Curie, Paris, France; Institut Pierre-Gilles de Gennes, Paris Sciences and Letters Research University, Paris, France
| | - Lucie Barbier
- UMR 144, Institut Curie, Paris, France; Institut Pierre-Gilles de Gennes, Paris Sciences and Letters Research University, Paris, France
| | - Despina Moshous
- Imagine Institute, Université de Paris Cité, Paris, France; Pediatric Immunology Hematology and Rheumatology Department, Université Paris Cité, Paris, France
| | - Bénédicte Neven
- Imagine Institute, Université de Paris Cité, Paris, France; Pediatric Immunology Hematology and Rheumatology Department, Université Paris Cité, Paris, France
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Main, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Alain Fischer
- Imagine Institute, Université de Paris Cité, Paris, France; Pediatric Immunology Hematology and Rheumatology Department, Université Paris Cité, Paris, France; Collège de France, Paris, France
| | - Gaël Ménasché
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France
| | - Geneviève de Saint Basile
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France; Centre d'Etude des Déficits Immunitaires, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Pablo Vargas
- UMR 144, Institut Curie, Paris, France; Institut Pierre-Gilles de Gennes, Paris Sciences and Letters Research University, Paris, France; Leukomotion Lab, Université de Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, F-75015 Paris, France.
| | - Fernando E Sepulveda
- Molecular Basis of Altered Immune Homeostasis Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) Unite Mixte de Recherche (UMR) 1163, Paris, France; Imagine Institute, Université de Paris Cité, Paris, France; CNRS, Paris, France.
| |
Collapse
|
17
|
Saleh J, Fardin MA, Barai A, Soleilhac M, Frenoy O, Gaston C, Cui H, Dang T, Gaudin N, Vincent A, Minc N, Delacour D. Length limitation of astral microtubules orients cell divisions in murine intestinal crypts. Dev Cell 2023; 58:1519-1533.e6. [PMID: 37419117 DOI: 10.1016/j.devcel.2023.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 05/25/2023] [Accepted: 06/14/2023] [Indexed: 07/09/2023]
Abstract
Planar spindle orientation is critical for epithelial tissue organization and is generally instructed by the long cell-shape axis or cortical polarity domains. We introduced mouse intestinal organoids in order to study spindle orientation in a monolayered mammalian epithelium. Although spindles were planar, mitotic cells remained elongated along the apico-basal (A-B) axis, and polarity complexes were segregated to basal poles, so that spindles oriented in an unconventional manner, orthogonal to both polarity and geometric cues. Using high-resolution 3D imaging, simulations, and cell-shape and cytoskeleton manipulations, we show that planar divisions resulted from a length limitation in astral microtubules (MTs) which precludes them from interacting with basal polarity, and orient spindles from the local geometry of apical domains. Accordingly, lengthening MTs affected spindle planarity, cell positioning, and crypt arrangement. We conclude that MT length regulation may serve as a key mechanism for spindles to sense local cell shapes and tissue forces to preserve mammalian epithelial architecture.
Collapse
Affiliation(s)
- Jad Saleh
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | | | - Amlan Barai
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Matis Soleilhac
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Olivia Frenoy
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Cécile Gaston
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Hongyue Cui
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Tien Dang
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Noémie Gaudin
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Audrey Vincent
- Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277, 59000 Lille, France; ORGALille Core Facility, CANTHER, Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277, 59000 Lille, France
| | - Nicolas Minc
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France; Equipe Labellisée La Ligue Contre le Cancer, France.
| | - Delphine Delacour
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France.
| |
Collapse
|
18
|
Fu L, Zou Y, Yu B, Hong D, Guan T, Hu J, Xu Y, Wu Y, Kou J, Lv Y. Background and roles: myosin in autoimmune diseases. Front Cell Dev Biol 2023; 11:1220672. [PMID: 37691828 PMCID: PMC10484797 DOI: 10.3389/fcell.2023.1220672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
The myosin superfamily is a group of molecular motors. Autoimmune diseases are characterized by dysregulation or deficiency of the immune tolerance mechanism, resulting in an immune response to the human body itself. The link between myosin and autoimmune diseases is much more complex than scientists had hoped. Myosin itself immunization can induce experimental autoimmune diseases of animals, and myosins were abnormally expressed in a number of autoimmune diseases. Additionally, myosin takes part in the pathological process of multiple sclerosis, Alzheimer's disease, Parkinson's disease, autoimmune myocarditis, myositis, hemopathy, inclusion body diseases, etc. However, research on myosin and its involvement in the occurrence and development of diseases is still in its infancy, and the underlying pathological mechanisms are not well understood. We can reasonably predict that myosin might play a role in new treatments of autoimmune diseases.
Collapse
Affiliation(s)
- Longsheng Fu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yonghui Zou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangxi, China
| | - Daojun Hong
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Teng Guan
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Jinfang Hu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi Xu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yaoqi Wu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangxi, China
| | - Yanni Lv
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Ueda Y, Higasa K, Kamioka Y, Kondo N, Horitani S, Ikeda Y, Bergmeier W, Fukui Y, Kinashi T. Rap1 organizes lymphocyte front-back polarity via RhoA signaling and talin1. iScience 2023; 26:107292. [PMID: 37520697 PMCID: PMC10374465 DOI: 10.1016/j.isci.2023.107292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/30/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Lymphocyte trafficking requires fine-tuning of chemokine-mediated cell migration. This process depends on cytoskeletal dynamics and polarity, but its regulation remains elusive. We quantitatively measured cell polarity and revealed critical roles performed by integrin activator Rap1 in this process, independent of substrate adhesion. Rap1-deficient naive T cells exhibited impaired abilities to reorganize the actin cytoskeleton into pseudopods and actomyosin-rich uropods. Rap1-GTPase activating proteins (GAPs), Rasa3 and Sipa1, maintained an unpolarized shape; deletion of these GAPs spontaneously induced cell polarization, indicative of the polarizing effect of Rap1. Rap1 activation required F-actin scaffolds, and stimulated RhoA activation and actomyosin contractility at the rear. Furthermore, talin1 acted on Rap1 downstream effectors to promote actomyosin contractility in the uropod, which occurred independently of substrate adhesion and talin1 binding to integrins. These findings indicate that Rap1 signaling to RhoA and talin1 regulates chemokine-stimulated lymphocyte polarization and chemotaxis in a manner independent of adhesion.
Collapse
Affiliation(s)
- Yoshihiro Ueda
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Koichiro Higasa
- The Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Yuji Kamioka
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Naoyuki Kondo
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Shunsuke Horitani
- Division of Gastroenterology and Hepatology, The Third Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Yoshiki Ikeda
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tatsuo Kinashi
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
20
|
Pruitt HC, Guan Y, Liu H, Carey AE, Brennen WN, Lu J, Joshu C, Weeraratna A, Lotan TL, Karin Eisinger-Mathason TS, Gerecht S. Collagen VI deposition mediates stromal T cell trapping through inhibition of T cell motility in the prostate tumor microenvironment. Matrix Biol 2023; 121:90-104. [PMID: 37331435 DOI: 10.1016/j.matbio.2023.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/11/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
The tumor extracellular matrix (ECM) is a barrier to anti-tumor immunity in solid tumors by disrupting T cell-tumor cell interaction underlying the need for elucidating mechanisms by which specific ECM proteins impact T cell motility and activity within the desmoplastic stroma of solid tumors. Here, we show that Collagen VI (Col VI) deposition correlates with stromal T cell density in human prostate cancer specimens. Furthermore, motility of CD4+ T cells is completely ablated on purified Col VI surfaces when compared with Fibronectin and Collagen I. Importantly, T cells adhered to Col VI surfaces displayed reduced cell spreading and fibrillar actin, indicating a reduction in traction force generation accompanied by a decrease in integrin β1 clustering. We found that CD4+ T cells largely lack expression of integrin α1 in the prostate tumor microenvironment and that blockade of α1β1 integrin heterodimers inhibited CD8+ T cell motility on prostate fibroblast-derived matrix, while re-expression of ITGA1 improved motility. Taken together, we show that the Col VI-rich microenvironment in prostate cancer reduces the motility of CD4+ T cells lacking integrin α1, leading to their accumulation in the stroma, thus putatively inhibiting anti-tumor T cell responses.
Collapse
Affiliation(s)
- Hawley C Pruitt
- Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ya Guan
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Hudson Liu
- Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alexis E Carey
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - W Nathaniel Brennen
- Department of Oncology, The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jiayun Lu
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Corrine Joshu
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ashani Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - T S Karin Eisinger-Mathason
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sharon Gerecht
- Institute for NanoBioTechnology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
21
|
D’Arcy BR, Lennox AL, Manso Musso C, Bracher A, Escobar-Tomlienovich C, Perez-Sanchez S, Silver DL. Non-muscle myosins control radial glial basal endfeet to mediate interneuron organization. PLoS Biol 2023; 21:e3001926. [PMID: 36854011 PMCID: PMC9974137 DOI: 10.1371/journal.pbio.3001926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/17/2023] [Indexed: 03/02/2023] Open
Abstract
Radial glial cells (RGCs) are essential for the generation and organization of neurons in the cerebral cortex. RGCs have an elongated bipolar morphology with basal and apical endfeet that reside in distinct niches. Yet, how this subcellular compartmentalization of RGCs controls cortical development is largely unknown. Here, we employ in vivo proximity labeling, in the mouse, using unfused BirA to generate the first subcellular proteome of RGCs and uncover new principles governing local control of cortical development. We discover a cohort of proteins that are significantly enriched in RGC basal endfeet, with MYH9 and MYH10 among the most abundant. Myh9 and Myh10 transcripts also localize to endfeet with distinct temporal dynamics. Although they each encode isoforms of non-muscle myosin II heavy chain, Myh9 and Myh10 have drastically different requirements for RGC integrity. Myh9 loss from RGCs decreases branching complexity and causes endfoot protrusion through the basement membrane. In contrast, Myh10 controls endfoot adhesion, as mutants have unattached apical and basal endfeet. Finally, we show that Myh9- and Myh10-mediated regulation of RGC complexity and endfoot position non-cell autonomously controls interneuron number and organization in the marginal zone. Our study demonstrates the utility of in vivo proximity labeling for dissecting local control of complex systems and reveals new mechanisms for dictating RGC integrity and cortical architecture.
Collapse
Affiliation(s)
- Brooke R. D’Arcy
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Ashley L. Lennox
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Camila Manso Musso
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Annalise Bracher
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Carla Escobar-Tomlienovich
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Stephany Perez-Sanchez
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Debra L. Silver
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Institute for Brain Sciences, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Regeneration Center, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
22
|
Čada Š, Vondálová Blanářová O, Gömoryová K, Mikulová A, Bačovská P, Zezula N, Kumari Jadaun A, Janovská P, Plešingerová H, Bryja V. Role of casein kinase 1 in the amoeboid migration of B-cell leukemic and lymphoma cells: A quantitative live imaging in the confined environment. Front Cell Dev Biol 2022; 10:911966. [PMID: 36561363 PMCID: PMC9763939 DOI: 10.3389/fcell.2022.911966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022] Open
Abstract
The migratory properties of leukemic cells are commonly associated with their pathological potential and can significantly affect the disease progression. While the research in immunopathology mostly employed powerful indirect methods such as flow cytometry, these cells were rarely observed directly using live imaging microscopy. This is especially true for the malignant cells of the B-cell lineage, such as those originating from chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL). In this study, we employed open-source image analysis tools to automatically and quantitatively describe the amoeboid migration of four B-cell leukemic and lymphoma cell lines and primary CLL cells. To avoid the effect of the shear stress of the medium on these usually non-adherent cells, we have confined the cells using a modified under-agarose assay. Surprisingly, the behavior of tested cell lines differed substantially in terms of basal motility or response to chemokines and VCAM1 stimulation. Since casein kinase 1 (CK1) was reported as a regulator of B-cell migration and a promoter of CLL, we looked at the effects of CK1 inhibition in more detail. Migration analysis revealed that CK1 inhibition induced rapid negative effects on the migratory polarity of these cells, which was quantitatively and morphologically distinct from the effect of ROCK inhibition. We have set up an assay that visualizes endocytic vesicles in the uropod and facilitates morphological analysis. This assay hints that the effect of CK1 inhibition might be connected to defects in polarized intracellular transport. In summary, 1) we introduce and validate a pipeline for the imaging and quantitative assessment of the amoeboid migration of CLL/MCL cells, 2) we provide evidence that the assay is sensitive enough to mechanistically study migration defects identified by the transwell assay, and 3) we describe the polarity defects induced by inhibition or deletion of CK1ε.
Collapse
Affiliation(s)
- Štěpán Čada
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | | | - Kristína Gömoryová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Antónia Mikulová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Petra Bačovská
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Nikodém Zezula
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Alka Kumari Jadaun
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Pavlína Janovská
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Hana Plešingerová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia,Department of Internal Medicine—Hematology and Oncology, University Hospital Brno, Brno, Czechia
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia,Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia,*Correspondence: Vítězslav Bryja,
| |
Collapse
|
23
|
Pentinmikko N, Lozano R, Scharaw S, Andersson S, Englund JI, Castillo-Azofeifa D, Gallagher A, Broberg M, Song KY, Sola Carvajal A, Speidel AT, Sundstrom M, Allbritton N, Stevens MM, Klein OD, Teixeira A, Katajisto P. Cellular shape reinforces niche to stem cell signaling in the small intestine. SCIENCE ADVANCES 2022; 8:eabm1847. [PMID: 36240269 PMCID: PMC9565803 DOI: 10.1126/sciadv.abm1847] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 08/30/2022] [Indexed: 06/06/2023]
Abstract
Niche-derived factors regulate tissue stem cells, but apart from the mechanosensory pathways, the effect of niche geometry is not well understood. We used organoids and bioengineered tissue culture platforms to demonstrate that the conical shape of Lgr5+ small intestinal stem cells (ISCs) facilitate their self-renewal and function. Inhibition of non-muscle myosin II (NM II)-driven apical constriction altered ISC shape and reduced niche curvature and stem cell capacity. Niche curvature is decreased in aged mice, suggesting that suboptimal interactions between old ISCs and their niche develop with age. We show that activation of NM IIC or physical restriction to young topology improves in vitro regeneration by old epithelium. We propose that the increase in lateral surface area of ISCs induced by apical constriction promotes interactions between neighboring cells, and the curved topology of the intestinal niche has evolved to maximize signaling between ISCs and neighboring cells.
Collapse
Affiliation(s)
- Nalle Pentinmikko
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Rodrigo Lozano
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Sandra Scharaw
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
| | - Simon Andersson
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Johanna I. Englund
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - David Castillo-Azofeifa
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
- Immunology Discovery, Genentech Inc., South San Francisco, CA, USA
| | - Aaron Gallagher
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Martin Broberg
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ki-Young Song
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Agustín Sola Carvajal
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
| | - Alessondra T. Speidel
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Michael Sundstrom
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Nancy Allbritton
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Molly M. Stevens
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Materials and Department of Bioengineering, Imperial College London, UK
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ana Teixeira
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pekka Katajisto
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
- Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Delgado M, Lennon-Duménil AM. How cell migration helps immune sentinels. Front Cell Dev Biol 2022; 10:932472. [PMID: 36268510 PMCID: PMC9577558 DOI: 10.3389/fcell.2022.932472] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/13/2022] [Indexed: 12/01/2022] Open
Abstract
The immune system relies on the migratory capacity of its cellular components, which must be mobile in order to defend the host from invading micro-organisms or malignant cells. This applies in particular to immune sentinels from the myeloid lineage, i.e. macrophages and dendritic cells. Cell migration is already at work during mammalian early development, when myeloid cell precursors migrate from the yolk sac, an extra embryonic structure, to colonize tissues and form the pool of tissue-resident macrophages. Later, this is accompanied by a migration wave of precursors and monocytes from the bone marrow to secondary lymphoid organs and the peripheral tissues. They differentiate into DCs and monocyte-derived macrophages. During adult life, cell migration endows immune cells with the ability to patrol their environment as well as to circulate between peripheral tissues and lymphoid organs. Hence migration of immune cells is key to building an efficient defense system for an organism. In this review, we will describe how cell migratory capacity regulates the various stages in the life of myeloid cells from development to tissue patrolling, and migration to lymph nodes. We will focus on the role of the actin cytoskeletal machinery and its regulators, and how it contributes to the establishment and function of the immune system.
Collapse
|
25
|
Thompson SB, Waldman MM, Jacobelli J. Polymerization power: effectors of actin polymerization as regulators of T lymphocyte migration through complex environments. FEBS J 2022; 289:6154-6171. [PMID: 34273243 PMCID: PMC8761786 DOI: 10.1111/febs.16130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/24/2021] [Accepted: 07/16/2021] [Indexed: 11/26/2022]
Abstract
During their life span, T cells are tasked with patrolling the body for potential pathogens. To do so, T cells migrate through numerous distinct anatomical sites and tissue environments with different biophysical characteristics. To migrate through these different environments, T cells use various motility strategies that rely on actin network remodeling to generate shape changes and mechanical forces. In this review, we initially discuss the migratory journey of T cells and then cover the actin polymerization effectors at play in T cells, and finally, we focus on the function of these effectors of actin cytoskeleton remodeling in mediating T-cell migration through diverse tissue environments. Specifically, we will discuss the current state of the field pertaining to our understanding of the roles in T-cell migration played by members of the three main families of actin polymerization machinery: the Arp2/3 complex; formin proteins; and Ena/VASP proteins.
Collapse
Affiliation(s)
- Scott B. Thompson
- Department of Immunology and Microbiology, University of Colorado School of Medicine
| | - Monique M. Waldman
- Department of Immunology and Microbiology, University of Colorado School of Medicine
- Barbara Davis Research Center, University of Colorado School of Medicine
| | - Jordan Jacobelli
- Department of Immunology and Microbiology, University of Colorado School of Medicine
- Barbara Davis Research Center, University of Colorado School of Medicine
| |
Collapse
|
26
|
Waldman MM, Rahkola JT, Sigler AL, Chung JW, Willett BAS, Kedl RM, Friedman RS, Jacobelli J. Ena/VASP Protein-Mediated Actin Polymerization Contributes to Naïve CD8 + T Cell Activation and Expansion by Promoting T Cell-APC Interactions In Vivo. Front Immunol 2022; 13:856977. [PMID: 35757762 PMCID: PMC9222560 DOI: 10.3389/fimmu.2022.856977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Naïve T cell activation in secondary lymphoid organs such as lymph nodes (LNs) occurs upon recognition of cognate antigen presented by antigen presenting cells (APCs). T cell activation requires cytoskeleton rearrangement and sustained interactions with APCs. Enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) proteins are a family of cytoskeletal effector proteins responsible for actin polymerization and are frequently found at the leading edge of motile cells. Ena/VASP proteins have been implicated in motility and adhesion in various cell types, but their role in primary T cell interstitial motility and activation has not been explored. Our goal was to determine the contribution of Ena/VASP proteins to T cell–APC interactions, T cell activation, and T cell expansion in vivo. Our results showed that naïve T cells from Ena/VASP-deficient mice have a significant reduction in antigen-specific T cell accumulation following Listeria monocytogenes infection. The kinetics of T cell expansion impairment were further confirmed in Ena/VASP-deficient T cells stimulated via dendritic cell immunization. To investigate the cause of this T cell expansion defect, we analyzed T cell–APC interactions in vivo by two-photon microscopy and observed fewer Ena/VASP-deficient naïve T cells interacting with APCs in LNs during priming. We also determined that Ena/VASP-deficient T cells formed conjugates with significantly less actin polymerization at the T cell–APC synapse, and that these conjugates were less stable than their WT counterparts. Finally, we found that Ena/VASP-deficient T cells have less LFA-1 polarized to the T cell–APC synapse. Thus, we conclude that Ena/VASP proteins contribute to T cell actin remodeling during T cell–APC interactions, which promotes the initiation of stable T cell conjugates during APC scanning. Therefore, Ena/VASP proteins are required for efficient activation and expansion of T cells in vivo.
Collapse
Affiliation(s)
- Monique M Waldman
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeremy T Rahkola
- Rocky Mountain Regional Veterans Affairs (VA) Medical Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ashton L Sigler
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeffrey W Chung
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Benjamin A S Willett
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rachel S Friedman
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jordan Jacobelli
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Barbara Davis Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States
| |
Collapse
|
27
|
Nguyen DT, Ogando-Rivas E, Liu R, Wang T, Rubin J, Jin L, Tao H, Sawyer WW, Mendez-Gomez HR, Cascio M, Mitchell DA, Huang J, Sawyer WG, Sayour EJ, Castillo P. CAR T Cell Locomotion in Solid Tumor Microenvironment. Cells 2022; 11:1974. [PMID: 35741103 PMCID: PMC9221866 DOI: 10.3390/cells11121974] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 01/25/2023] Open
Abstract
The promising outcomes of chimeric antigen receptor (CAR) T cell therapy in hematologic malignancies potentiates its capability in the fight against many cancers. Nevertheless, this immunotherapy modality needs significant improvements for the treatment of solid tumors. Researchers have incrementally identified limitations and constantly pursued better CAR designs. However, even if CAR T cells are armed with optimal killer functions, they must overcome and survive suppressive barriers imposed by the tumor microenvironment (TME). In this review, we will discuss in detail the important role of TME in CAR T cell trafficking and how the intrinsic barriers contribute to an immunosuppressive phenotype and cancer progression. It is of critical importance that preclinical models can closely recapitulate the in vivo TME to better predict CAR T activity. Animal models have contributed immensely to our understanding of human diseases, but the intensive care for the animals and unreliable representation of human biology suggest in vivo models cannot be the sole approach to CAR T cell therapy. On the other hand, in vitro models for CAR T cytotoxic assessment offer valuable insights to mechanistic studies at the single cell level, but they often lack in vivo complexities, inter-individual heterogeneity, or physiologically relevant spatial dimension. Understanding the advantages and limitations of preclinical models and their applications would enable more reliable prediction of better clinical outcomes.
Collapse
Affiliation(s)
- Duy T. Nguyen
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
| | - Elizabeth Ogando-Rivas
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Ruixuan Liu
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Theodore Wang
- College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Jacob Rubin
- Warrington College of Business, University of Florida, Gainesville, FL 32610, USA;
| | - Linchun Jin
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Haipeng Tao
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - William W. Sawyer
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Hector R. Mendez-Gomez
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Matthew Cascio
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Duane A. Mitchell
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Jianping Huang
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - W. Gregory Sawyer
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
| | - Elias J. Sayour
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Paul Castillo
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| |
Collapse
|
28
|
Teng T, Teng CS, Kaartinen V, Bush JO. A unique form of collective epithelial migration is crucial for tissue fusion in the secondary palate and can overcome loss of epithelial apoptosis. Development 2022; 149:275520. [PMID: 35593401 PMCID: PMC9188751 DOI: 10.1242/dev.200181] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/11/2022] [Indexed: 11/20/2022]
Abstract
Tissue fusion frequently requires the removal of an epithelium that intervenes distinct primordia to form one continuous structure. In the mammalian secondary palate, a midline epithelial seam (MES) forms between two palatal shelves and must be removed to allow mesenchymal confluence. Abundant apoptosis and cell extrusion support their importance in MES removal. However, genetically disrupting the intrinsic apoptotic regulators BAX and BAK within the MES results in complete loss of cell death and cell extrusion, but successful removal of the MES. Novel static- and live-imaging approaches reveal that the MES is removed through streaming migration of epithelial trails and islands to reach the oral and nasal epithelial surfaces. Epithelial trail cells that express the basal epithelial marker ΔNp63 begin to express periderm markers, suggesting that migration is concomitant with differentiation. Live imaging reveals anisotropic actomyosin contractility within epithelial trails, and genetic ablation of actomyosin contractility results in dispersion of epithelial collectives and failure of normal MES migration. These findings demonstrate redundancy between cellular mechanisms of morphogenesis, and reveal a crucial and unique form of collective epithelial migration during tissue fusion. Summary: Multiple cellular processes mediate secondary palate fusion, including a unique form of streaming collective epithelial migration driven by pulsatile actomyosin contractility.
Collapse
Affiliation(s)
- Teng Teng
- University of California San Francisco 1 Department of Cell and Tissue Biology , , San Francisco, CA 94143 , USA
- University of California San Francisco 2 Program in Craniofacial Biology , , San Francisco, CA 94143 , USA
- Institute for Human Genetics, University of California San Francisco 3 , San Francisco, CA 94143 , USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco 4 , San Francisco, CA 94143 , USA
| | - Camilla S. Teng
- University of California San Francisco 1 Department of Cell and Tissue Biology , , San Francisco, CA 94143 , USA
- University of California San Francisco 2 Program in Craniofacial Biology , , San Francisco, CA 94143 , USA
- Institute for Human Genetics, University of California San Francisco 3 , San Francisco, CA 94143 , USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco 4 , San Francisco, CA 94143 , USA
| | - Vesa Kaartinen
- University of Michigan School of Dentistry 5 Department of Biologic and Materials Sciences , , Ann Arbor, MI 48109 , USA
| | - Jeffrey O. Bush
- University of California San Francisco 1 Department of Cell and Tissue Biology , , San Francisco, CA 94143 , USA
- University of California San Francisco 2 Program in Craniofacial Biology , , San Francisco, CA 94143 , USA
- Institute for Human Genetics, University of California San Francisco 3 , San Francisco, CA 94143 , USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco 4 , San Francisco, CA 94143 , USA
| |
Collapse
|
29
|
van Steijn L, Wortel IMN, Sire C, Dupré L, Theraulaz G, Merks RMH. Computational modelling of cell motility modes emerging from cell-matrix adhesion dynamics. PLoS Comput Biol 2022; 18:e1009156. [PMID: 35157694 PMCID: PMC8880896 DOI: 10.1371/journal.pcbi.1009156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 02/25/2022] [Accepted: 01/18/2022] [Indexed: 11/18/2022] Open
Abstract
Lymphocytes have been described to perform different motility patterns such as Brownian random walks, persistent random walks, and Lévy walks. Depending on the conditions, such as confinement or the distribution of target cells, either Brownian or Lévy walks lead to more efficient interaction with the targets. The diversity of these motility patterns may be explained by an adaptive response to the surrounding extracellular matrix (ECM). Indeed, depending on the ECM composition, lymphocytes either display a floating motility without attaching to the ECM, or sliding and stepping motility with respectively continuous or discontinuous attachment to the ECM, or pivoting behaviour with sustained attachment to the ECM. Moreover, on the long term, lymphocytes either perform a persistent random walk or a Brownian-like movement depending on the ECM composition. How the ECM affects cell motility is still incompletely understood. Here, we integrate essential mechanistic details of the lymphocyte-matrix adhesions and lymphocyte intrinsic cytoskeletal induced cell propulsion into a Cellular Potts model (CPM). We show that the combination of de novo cell-matrix adhesion formation, adhesion growth and shrinkage, adhesion rupture, and feedback of adhesions onto cell propulsion recapitulates multiple lymphocyte behaviours, for different lymphocyte subsets and various substrates. With an increasing attachment area and increased adhesion strength, the cells’ speed and persistence decreases. Additionally, the model predicts random walks with short-term persistent but long-term subdiffusive properties resulting in a pivoting type of motility. For small adhesion areas, the spatial distribution of adhesions emerges as a key factor influencing cell motility. Small adhesions at the front allow for more persistent motility than larger clusters at the back, despite a similar total adhesion area. In conclusion, we present an integrated framework to simulate the effects of ECM proteins on cell-matrix adhesion dynamics. The model reveals a sufficient set of principles explaining the plasticity of lymphocyte motility. During immunosurveillance, lymphocytes patrol through tissues to interact with cancer cells, other immune cells, and pathogens. The efficiency of this process depends on the kinds of trajectories taken, ranging from simple Brownian walks to Lévy walks. The composition of the extracellular matrix (ECM), a network of macromolecules, affects the formation of cell-matrix adhesions, thus strongly influencing the way lymphocytes move. Here, we present a model of lymphocyte motility driven by adhesions that grow, shrink and rupture in response to the ECM and cellular forces. Compared to other models, our model is computationally light making it suitable for generating long term cell track data, while still capturing actin dynamics and adhesion turnover. Our model suggests that cell motility is affected by the force required to break adhesions and the rate at which new adhesions form. Adhesions can promote cell protrusion by inhibiting retrograde actin flow. After introducing this effect into the model, we found that it reduces the cellular diffusivity and that it promotes stick-slip behaviour. Furthermore, location and size of adhesion clusters determined cell persistence. Overall, our model explains the plasticity of lymphocyte behaviour in response to the ECM.
Collapse
Affiliation(s)
| | - Inge M. N. Wortel
- Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, The Netherlands
| | - Clément Sire
- Laboratoire de Physique Théorique, Centre National de la Recherche Scientifique (CNRS) & Université de Toulouse—Paul Sabatier, Toulouse, France
| | - Loïc Dupré
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Université de Toulouse, Toulouse, France
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Guy Theraulaz
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Centre National de la Recherche Scientifique (CNRS) & Université de Toulouse—Paul Sabatier, Toulouse, France
- Centre for Ecological Sciences, Indian Institute of Science, Bengaluru, India
| | - Roeland M. H. Merks
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
30
|
Rivera CA, Randrian V, Richer W, Gerber-Ferder Y, Delgado MG, Chikina AS, Frede A, Sorini C, Maurin M, Kammoun-Chaari H, Parigi SM, Goudot C, Cabeza-Cabrerizo M, Baulande S, Lameiras S, Guermonprez P, Reis e Sousa C, Lecuit M, Moreau HD, Helft J, Vignjevic DM, Villablanca EJ, Lennon-Duménil AM. Epithelial colonization by gut dendritic cells promotes their functional diversification. Immunity 2022; 55:129-144.e8. [PMID: 34910930 PMCID: PMC8751639 DOI: 10.1016/j.immuni.2021.11.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/19/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) patrol tissues and transport antigens to lymph nodes to initiate adaptive immune responses. Within tissues, DCs constitute a complex cell population composed of distinct subsets that can exhibit different activation states and functions. How tissue-specific cues orchestrate DC diversification remains elusive. Here, we show that the small intestine included two pools of cDC2s originating from common pre-DC precursors: (1) lamina propria (LP) CD103+CD11b+ cDC2s that were mature-like proinflammatory cells and (2) intraepithelial cDC2s that exhibited an immature-like phenotype as well as tolerogenic properties. These phenotypes resulted from the action of food-derived retinoic acid (ATRA), which enhanced actomyosin contractility and promoted LP cDC2 transmigration into the epithelium. There, cDC2s were imprinted by environmental cues, including ATRA itself and the mucus component Muc2. Hence, by reaching distinct subtissular niches, DCs can exist as immature and mature cells within the same tissue, revealing an additional mechanism of DC functional diversification.
Collapse
Affiliation(s)
- Claudia A Rivera
- Institut Curie, INSERM U932, PSL Research University, 75005 Paris, France
| | - Violaine Randrian
- Institut Curie, INSERM U932, PSL Research University, 75005 Paris, France
| | - Wilfrid Richer
- Institut Curie, INSERM U932, PSL Research University, 75005 Paris, France
| | | | | | - Aleksandra S Chikina
- Institut Curie, INSERM U932, PSL Research University, 75005 Paris, France; Institut Curie, CNRS UMR 144, PSL Research University, 75005 Paris, France
| | - Annika Frede
- Immunology and Allergy division, Department of Medicine, Solna, Karolinska Institutet and University Hospital, 17176 Stockholm, Sweden; Center of Molecular Medicine, 17176 Stockholm, Sweden
| | - Chiara Sorini
- Immunology and Allergy division, Department of Medicine, Solna, Karolinska Institutet and University Hospital, 17176 Stockholm, Sweden; Center of Molecular Medicine, 17176 Stockholm, Sweden
| | - Mathieu Maurin
- Institut Curie, INSERM U932, PSL Research University, 75005 Paris, France
| | - Hana Kammoun-Chaari
- Biology of Infection Unit, Institut Pasteur, INSERM U1117, 75015 Paris, France
| | - Sara M Parigi
- Immunology and Allergy division, Department of Medicine, Solna, Karolinska Institutet and University Hospital, 17176 Stockholm, Sweden; Center of Molecular Medicine, 17176 Stockholm, Sweden
| | - Christel Goudot
- Institut Curie, INSERM U932, PSL Research University, 75005 Paris, France
| | | | - Sylvain Baulande
- ICGex Next-Generation Sequencing Platform, Institut Curie, PSL Research University, 75005 Paris, France
| | - Sonia Lameiras
- ICGex Next-Generation Sequencing Platform, Institut Curie, PSL Research University, 75005 Paris, France
| | - Pierre Guermonprez
- Université de Paris, Centre for Inflammation Research, CNRS ERL8252, INSERM1149, Paris, France
| | | | - Marc Lecuit
- Biology of Infection Unit, Institut Pasteur, INSERM U1117, 75015 Paris, France; Université de Paris, Necker-Enfants Malades University Hospital, Department of Infectious Diseases and Tropical Medicine, APHP, Institut Imagine, Paris, France
| | - Hélène D Moreau
- Institut Curie, INSERM U932, PSL Research University, 75005 Paris, France
| | - Julie Helft
- Institut Curie, INSERM U932, PSL Research University, 75005 Paris, France
| | | | - Eduardo J Villablanca
- Immunology and Allergy division, Department of Medicine, Solna, Karolinska Institutet and University Hospital, 17176 Stockholm, Sweden; Center of Molecular Medicine, 17176 Stockholm, Sweden
| | | |
Collapse
|
31
|
Kamnev A, Lacouture C, Fusaro M, Dupré L. Molecular Tuning of Actin Dynamics in Leukocyte Migration as Revealed by Immune-Related Actinopathies. Front Immunol 2021; 12:750537. [PMID: 34867982 PMCID: PMC8634686 DOI: 10.3389/fimmu.2021.750537] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/12/2021] [Indexed: 01/13/2023] Open
Abstract
Motility is a crucial activity of immune cells allowing them to patrol tissues as they differentiate, sample or exchange information, and execute their effector functions. Although all immune cells are highly migratory, each subset is endowed with very distinct motility patterns in accordance with functional specification. Furthermore individual immune cell subsets adapt their motility behaviour to the surrounding tissue environment. This review focuses on how the generation and adaptation of diversified motility patterns in immune cells is sustained by actin cytoskeleton dynamics. In particular, we review the knowledge gained through the study of inborn errors of immunity (IEI) related to actin defects. Such pathologies are unique models that help us to uncover the contribution of individual actin regulators to the migration of immune cells in the context of their development and function.
Collapse
Affiliation(s)
- Anton Kamnev
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Claire Lacouture
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France.,Laboratoire De Physique Théorique, IRSAMC, Université De Toulouse (UPS), CNRS, Toulouse, France
| | - Mathieu Fusaro
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France
| | - Loïc Dupré
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France
| |
Collapse
|
32
|
Gupta S, Patteson AE, Schwarz JM. The role of vimentin-nuclear interactions in persistent cell motility through confined spaces. NEW JOURNAL OF PHYSICS 2021; 23:093042. [PMID: 35530563 PMCID: PMC9075336 DOI: 10.1088/1367-2630/ac2550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The ability of cells to move through small spaces depends on the mechanical properties of the cellular cytoskeleton and on nuclear deformability. In mammalian cells, the cytoskeleton is composed of three interacting, semi-flexible polymer networks: actin, microtubules, and intermediate filaments (IF). Recent experiments of mouse embryonic fibroblasts with and without vimentin have shown that the IF vimentin plays a role in confined cell motility. Here, we develop a minimal model of a cell moving through a microchannel that incorporates explicit effects of actin and vimentin and implicit effects of microtubules. Specifically, the model consists of a cell with an actomyosin cortex and a deformable cell nucleus and mechanical linkages between the two. By decreasing the amount of vimentin, we find that the cell speed increases for vimentin-null cells compared to cells with vimentin. The loss of vimentin increases nuclear deformation and alters nuclear positioning in the cell. Assuming nuclear positioning is a read-out for cell polarity, we propose a new polarity mechanism which couples cell directional motion with cytoskeletal strength and nuclear positioning and captures the abnormally persistent motion of vimentin-null cells, as observed in experiments. The enhanced persistence indicates that the vimentin-null cells are more controlled by the confinement and so less autonomous, relying more heavily on external cues than their wild-type counterparts. Our modeling results present a quantitative interpretation for recent experiments and have implications for understanding the role of vimentin in the epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Sarthak Gupta
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY USA
| | - Alison E Patteson
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY USA
| | - J M Schwarz
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY USA
- Indian Creek Farm, Ithaca, NY USA
| |
Collapse
|
33
|
Zhao R, Zhou X, Khan ES, Alansary D, Friedmann KS, Yang W, Schwarz EC, del Campo A, Hoth M, Qu B. Targeting the Microtubule-Network Rescues CTL Killing Efficiency in Dense 3D Matrices. Front Immunol 2021; 12:729820. [PMID: 34484240 PMCID: PMC8416057 DOI: 10.3389/fimmu.2021.729820] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/31/2021] [Indexed: 12/11/2022] Open
Abstract
Efficacy of cytotoxic T lymphocyte (CTL)-based immunotherapy is still unsatisfactory against solid tumors, which are frequently characterized by condensed extracellular matrix. Here, using a unique 3D killing assay, we identify that the killing efficiency of primary human CTLs is substantially impaired in dense collagen matrices. Although the expression of cytotoxic proteins in CTLs remained intact in dense collagen, CTL motility was largely compromised. Using light-sheet microscopy, we found that persistence and velocity of CTL migration was influenced by the stiffness and porosity of the 3D matrix. Notably, 3D CTL velocity was strongly correlated with their nuclear deformability, which was enhanced by disruption of the microtubule network especially in dense matrices. Concomitantly, CTL migration, search efficiency, and killing efficiency in dense collagen were significantly increased in microtubule-perturbed CTLs. In addition, the chemotherapeutically used microtubule inhibitor vinblastine drastically enhanced CTL killing efficiency in dense collagen. Together, our findings suggest targeting the microtubule network as a promising strategy to enhance efficacy of CTL-based immunotherapy against solid tumors, especially stiff solid tumors.
Collapse
Affiliation(s)
- Renping Zhao
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Xiangda Zhou
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Essak S. Khan
- INM-Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Dalia Alansary
- Molecular Biophysics, CIPMM, School of Medicine, Saarland University, Homburg, Germany
| | - Kim S. Friedmann
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Wenjuan Yang
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Eva C. Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | | | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- INM-Leibniz Institute for New Materials, Saarbrücken, Germany
| |
Collapse
|
34
|
Yang Q, Xue SL, Chan CJ, Rempfler M, Vischi D, Maurer-Gutierrez F, Hiiragi T, Hannezo E, Liberali P. Cell fate coordinates mechano-osmotic forces in intestinal crypt formation. Nat Cell Biol 2021; 23:733-744. [PMID: 34155381 PMCID: PMC7611267 DOI: 10.1038/s41556-021-00700-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 05/14/2021] [Indexed: 02/06/2023]
Abstract
Intestinal organoids derived from single cells undergo complex crypt-villus patterning and morphogenesis. However, the nature and coordination of the underlying forces remains poorly characterized. Here, using light-sheet microscopy and large-scale imaging quantification, we demonstrate that crypt formation coincides with a stark reduction in lumen volume. We develop a 3D biophysical model to computationally screen different mechanical scenarios of crypt morphogenesis. Combining this with live-imaging data and multiple mechanical perturbations, we show that actomyosin-driven crypt apical contraction and villus basal tension work synergistically with lumen volume reduction to drive crypt morphogenesis, and demonstrate the existence of a critical point in differential tensions above which crypt morphology becomes robust to volume changes. Finally, we identified a sodium/glucose cotransporter that is specific to differentiated enterocytes that modulates lumen volume reduction through cell swelling in the villus region. Together, our study uncovers the cellular basis of how cell fate modulates osmotic and actomyosin forces to coordinate robust morphogenesis.
Collapse
Affiliation(s)
- Qiutan Yang
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland.
| | - Shi-Lei Xue
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Chii Jou Chan
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Markus Rempfler
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | - Dario Vischi
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | | | | | - Edouard Hannezo
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
35
|
Kindberg AA, Srivastava V, Muncie JM, Weaver VM, Gartner ZJ, Bush JO. EPH/EPHRIN regulates cellular organization by actomyosin contractility effects on cell contacts. J Cell Biol 2021; 220:e202005216. [PMID: 33798261 PMCID: PMC8025214 DOI: 10.1083/jcb.202005216] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 02/02/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
EPH/EPHRIN signaling is essential to many aspects of tissue self-organization and morphogenesis, but little is known about how EPH/EPHRIN signaling regulates cell mechanics during these processes. Here, we use a series of approaches to examine how EPH/EPHRIN signaling drives cellular self-organization. Contact angle measurements reveal that EPH/EPHRIN signaling decreases the stability of heterotypic cell:cell contacts through increased cortical actomyosin contractility. We find that EPH/EPHRIN-driven cell segregation depends on actomyosin contractility but occurs independently of directed cell migration and without changes in cell adhesion. Atomic force microscopy and live cell imaging of myosin localization support that EPH/EPHRIN signaling results in increased cortical tension. Interestingly, actomyosin contractility also nonautonomously drives increased EPHB2:EPHB2 homotypic contacts. Finally, we demonstrate that changes in tissue organization are driven by minimization of heterotypic contacts through actomyosin contractility in cell aggregates and by mouse genetics experiments. These data elucidate the biomechanical mechanisms driving EPH/EPHRIN-based cell segregation wherein differences in interfacial tension, regulated by actomyosin contractility, govern cellular self-organization.
Collapse
Affiliation(s)
- Abigail A. Kindberg
- Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA
| | - Jonathon M. Muncie
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA
- Graduate Program in Bioengineering, University of California, San Francisco, and University of California, Berkeley, San Francisco, CA
| | - Valerie M. Weaver
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA
- UCSF Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
- Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA
- Center for Cellular Construction, University of California, San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| | - Jeffrey O. Bush
- Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
36
|
Moreau JM, Gouirand V, Rosenblum MD. T-Cell Adhesion in Healthy and Inflamed Skin. JID INNOVATIONS 2021; 1:100014. [PMID: 35024681 PMCID: PMC8669513 DOI: 10.1016/j.xjidi.2021.100014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
The diverse populations of tissue-resident and transitory T cells present in the skin share a common functional need to enter, traverse, and interact with their environment. These processes are largely dependent on the regulated expression of adhesion molecules, such as selectins and integrins, which mediate bidirectional interactions between immune cells and skin stroma. Dysregulation and engagement of adhesion pathways contribute to ectopic T-cell activity in tissues, leading to the initiation and/or exacerbation of chronic inflammation. In this paper, we review how the molecular interactions supported by adhesion pathways contribute to T-cell dynamics and function in the skin. A comprehensive understanding of the molecular mechanisms underpinning T-cell adhesion in inflammatory skin disorders will facilitate the development of novel tissue-specific therapeutic strategies.
Collapse
Key Words
- AD, atopic dermatitis
- BM, basement membrane
- DC, dendritic cell
- DETC, dendritic epidermal γδ T cell
- ECM, extracellular matrix
- HF, hair follicle
- JC, John Cunningham
- LAD, leukocyte adhesion deficiency
- PML, progressive multifocal leukoencephalopathy
- Th, T helper
- Treg, regulatory T cell
- Trm, tissue-resident memory
Collapse
Affiliation(s)
- Joshua M. Moreau
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Victoire Gouirand
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Michael D. Rosenblum
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
37
|
Engineering T cells to enhance 3D migration through structurally and mechanically complex tumor microenvironments. Nat Commun 2021; 12:2815. [PMID: 33990566 PMCID: PMC8121808 DOI: 10.1038/s41467-021-22985-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 04/07/2021] [Indexed: 12/18/2022] Open
Abstract
Defining the principles of T cell migration in structurally and mechanically complex tumor microenvironments is critical to understanding escape from antitumor immunity and optimizing T cell-related therapeutic strategies. Here, we engineered nanotextured elastic platforms to study and enhance T cell migration through complex microenvironments and define how the balance between contractility localization-dependent T cell phenotypes influences migration in response to tumor-mimetic structural and mechanical cues. Using these platforms, we characterize a mechanical optimum for migration that can be perturbed by manipulating an axis between microtubule stability and force generation. In 3D environments and live tumors, we demonstrate that microtubule instability, leading to increased Rho pathway-dependent cortical contractility, promotes migration whereas clinically used microtubule-stabilizing chemotherapies profoundly decrease effective migration. We show that rational manipulation of the microtubule-contractility axis, either pharmacologically or through genome engineering, results in engineered T cells that more effectively move through and interrogate 3D matrix and tumor volumes. Thus, engineering cells to better navigate through 3D microenvironments could be part of an effective strategy to enhance efficacy of immune therapeutics. The mechanics of the migration of T cells into tumours is an important aspect of tumour immunity. Here the authors engineer complex 3D environments to explore functions of microtubules and cell contractility as strategies to enhance T cell migration in tumour microenvironments.
Collapse
|
38
|
Zhou Z, Cui F, Wen Q, Susan Zhou H. Effect of vimentin on cell migration in collagen-coated microchannels: A mimetic physiological confined environment. BIOMICROFLUIDICS 2021; 15:034105. [PMID: 34025897 PMCID: PMC8133791 DOI: 10.1063/5.0045197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/02/2021] [Indexed: 06/12/2023]
Abstract
Cancer cell migration through tissue pores and tracks into the bloodstream is a critical biological step for cancer metastasis. Although in vivo studies have shown that expression of vimentin can induce invasive cell lines, its role in cell cytoskeleton reorganization and cell motility under in vitro physical confinement remains unknown. Here, a microfluidic device with cell culture chamber and collagen-coated microchannels was developed as an in vitro model for physiological confinement environments. Using this microchannel assay, we demonstrated that the knockdown of vimentin decreases 3T3 fibroblast cell directional migration speed in confined microchannels. Additionally, as cells form dynamic membranes that define the leading edge of motile cells, different leading edge morphologies of 3T3 fibroblast and 3T3 vimentin knockdown cells were observed. The leading edge morphology change under confinement can be explained by the effect of vimentin on cytoskeletal organization and focal adhesion. The microfluidic device integrated with a time-lapse microscope provided a new approach to study the effect of vimentin on cell adhesion, migration, and invasiveness.
Collapse
Affiliation(s)
- Zhiru Zhou
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, USA
| | - Feiyun Cui
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, USA
| | - Qi Wen
- Department of Physics, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
| | - H. Susan Zhou
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, USA
| |
Collapse
|
39
|
Lemaître F, Carmena Moratalla A, Farzam-Kia N, Carpentier Solorio Y, Tastet O, Cleret-Buhot A, Guimond JV, Haddad E, Arbour N. Capturing T Lymphocytes' Dynamic Interactions With Human Neural Cells Using Time-Lapse Microscopy. Front Immunol 2021; 12:668483. [PMID: 33968073 PMCID: PMC8100528 DOI: 10.3389/fimmu.2021.668483] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/29/2021] [Indexed: 11/17/2022] Open
Abstract
To fully perform their functions, T lymphocytes migrate within organs’ parenchyma and interact with local cells. Infiltration of T lymphocytes within the central nervous system (CNS) is associated with numerous neurodegenerative disorders. Nevertheless, how these immune cells communicate and respond to neural cells remains unresolved. To investigate the behavior of T lymphocytes that reach the CNS, we have established an in vitro co-culture model and analyzed the spatiotemporal interactions between human activated CD8+ T lymphocytes and primary human astrocytes and neurons using time-lapse microscopy. By combining multiple variables extracted from individual CD8+ T cell tracking, we show that CD8+ T lymphocytes adopt a more motile and exploratory behavior upon interacting with astrocytes than with neurons. Pretreatment of astrocytes or neurons with IL-1β to mimic in vivo inflammation significantly increases CD8+ T lymphocyte motility. Using visual interpretation and analysis of numerical variables extracted from CD8+ T cell tracking, we identified four distinct CD8+ T lymphocyte behaviors: scanning, dancing, poking and round. IL-1β-pretreatment significantly increases the proportion of scanning CD8+ T lymphocytes, which are characterized by active exploration, and reduces the proportion of round CD8+ T lymphocytes, which are less active. Blocking MHC class I on astrocytes significantly diminishes the proportion of poking CD8+ T lymphocytes, which exhibit synapse-like interactions. Lastly, our co-culture time-lapse model is easily adaptable and sufficiently sensitive and powerful to characterize and quantify spatiotemporal interactions between human T lymphocytes and primary human cells in different conditions while preserving viability of fragile cells such as neurons and astrocytes.
Collapse
Affiliation(s)
- Florent Lemaître
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Ana Carmena Moratalla
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Negar Farzam-Kia
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Yves Carpentier Solorio
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Olivier Tastet
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Aurélie Cleret-Buhot
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Jean Victor Guimond
- Centre Local de Services Communautaires des Faubourgs, Centre Intégré Universitaire en Santé et Services Sociaux du Centre-Sud-de-l'Ile-de-Montréal, Montréal, QC, Canada
| | - Elie Haddad
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Nathalie Arbour
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| |
Collapse
|
40
|
Schliffka MF, Tortorelli AF, Özgüç Ö, de Plater L, Polzer O, Pelzer D, Maître JL. Multiscale analysis of single and double maternal-zygotic Myh9 and Myh10 mutants during mouse preimplantation development. eLife 2021; 10:e68536. [PMID: 33871354 PMCID: PMC8096435 DOI: 10.7554/elife.68536] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 03/28/2021] [Indexed: 12/28/2022] Open
Abstract
During the first days of mammalian development, the embryo forms the blastocyst, the structure responsible for implanting the mammalian embryo. Consisting of an epithelium enveloping the pluripotent inner cell mass and a fluid-filled lumen, the blastocyst results from a series of cleavage divisions, morphogenetic movements, and lineage specification. Recent studies have identified the essential role of actomyosin contractility in driving cytokinesis, morphogenesis, and fate specification, leading to the formation of the blastocyst. However, the preimplantation development of contractility mutants has not been characterized. Here, we generated single and double maternal-zygotic mutants of non-muscle myosin II heavy chains (NMHCs) to characterize them with multiscale imaging. We found that Myh9 (NMHC II-A) is the major NMHC during preimplantation development as its maternal-zygotic loss causes failed cytokinesis, increased duration of the cell cycle, weaker embryo compaction, and reduced differentiation, whereas Myh10 (NMHC II-B) maternal-zygotic loss is much less severe. Double maternal-zygotic mutants for Myh9 and Myh10 show a much stronger phenotype, failing most of the attempts of cytokinesis. We found that morphogenesis and fate specification are affected but nevertheless carry on in a timely fashion, regardless of the impact of the mutations on cell number. Strikingly, even when all cell divisions fail, the resulting single-celled embryo can initiate trophectoderm differentiation and lumen formation by accumulating fluid in increasingly large vacuoles. Therefore, contractility mutants reveal that fluid accumulation is a cell-autonomous process and that the preimplantation program carries on independently of successful cell division.
Collapse
Affiliation(s)
- Markus Frederik Schliffka
- Institut Curie, PSL Research University, Sorbonne UniversitéParisFrance
- Carl Zeiss SASMarly-le-RoyFrance
| | | | - Özge Özgüç
- Institut Curie, PSL Research University, Sorbonne UniversitéParisFrance
| | | | - Oliver Polzer
- Institut Curie, PSL Research University, Sorbonne UniversitéParisFrance
| | - Diane Pelzer
- Institut Curie, PSL Research University, Sorbonne UniversitéParisFrance
| | - Jean-Léon Maître
- Institut Curie, PSL Research University, Sorbonne UniversitéParisFrance
| |
Collapse
|
41
|
Aoun L, Nègre P, Gonsales C, Seveau de Noray V, Brustlein S, Biarnes-Pelicot M, Valignat MP, Theodoly O. Leukocyte transmigration and longitudinal forward-thrusting force in a microfluidic Transwell device. Biophys J 2021; 120:2205-2221. [PMID: 33838136 DOI: 10.1016/j.bpj.2021.03.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/10/2021] [Accepted: 03/24/2021] [Indexed: 01/21/2023] Open
Abstract
Transmigration of leukocytes across blood vessels walls is a critical step of the immune response. Transwell assays examine transmigration properties in vitro by counting cells passages through a membrane; however, the difficulty of in situ imaging hampers a clear disentanglement of the roles of adhesion, chemokinesis, and chemotaxis. We used here microfluidic Transwells to image the cells' transition from 2D migration on a surface to 3D migration in a confining microchannel and measure cells longitudinal forward-thrusting force in microchannels. Primary human effector T lymphocytes adhering with integrins LFA-1 (αLβ2) had a marked propensity to transmigrate in Transwells without chemotactic cue. Both adhesion and contractility were important to overcome the critical step of nucleus penetration but were remarkably dispensable for 3D migration in smooth microchannels deprived of topographic features. Transmigration in smooth channels was qualitatively consistent with a propulsion by treadmilling of cell envelope and squeezing of cell trailing edge. Stalling conditions of 3D migration were then assessed by imposing pressure drops across microchannels. Without specific adhesion, the cells slid backward with subnanonewton forces, showing that 3D migration under stress is strongly limited by a lack of adhesion and friction with channels. With specific LFA-1 mediated adhesion, stalling occurred at around 3 and 6 nN in 2 × 4 and 4 × 4 μm2 channels, respectively, supporting that stalling of adherent cells was under pressure control rather than force control. The stall pressure of 4 mbar is consistent with the pressure of actin filament polymerization that mediates lamellipod growth. The arrest of adherent cells under stress therefore seems controlled by the compression of the cell leading edge, which perturbs cells front-rear polarization and triggers adhesion failure or polarization reversal. Although stalling assays in microfluidic Transwells do not mimic in vivo transmigration, they provide a powerful tool to scrutinize 2D and 3D migration, barotaxis, and chemotaxis.
Collapse
Affiliation(s)
- Laurene Aoun
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Paulin Nègre
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Cristina Gonsales
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | | | - Sophie Brustlein
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | | | - Marie-Pierre Valignat
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Olivier Theodoly
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
42
|
Vesperini D, Montalvo G, Qu B, Lautenschläger F. Characterization of immune cell migration using microfabrication. Biophys Rev 2021; 13:185-202. [PMID: 34290841 PMCID: PMC8285443 DOI: 10.1007/s12551-021-00787-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
The immune system provides our defense against pathogens and aberrant cells, including tumorigenic and infected cells. Motility is one of the fundamental characteristics that enable immune cells to find invading pathogens, control tissue damage, and eliminate primary developing tumors, even in the absence of external treatments. These processes are termed "immune surveillance." Migration disorders of immune cells are related to autoimmune diseases, chronic inflammation, and tumor evasion. It is therefore essential to characterize immune cell motility in different physiologically and pathologically relevant scenarios to understand the regulatory mechanisms of functionality of immune responses. This review is focused on immune cell migration, to define the underlying mechanisms and the corresponding investigative approaches. We highlight the challenges that immune cells encounter in vivo, and the microfabrication methods to mimic particular aspects of their microenvironment. We discuss the advantages and disadvantages of the proposed tools, and provide information on how to access them. Furthermore, we summarize the directional cues that regulate individual immune cell migration, and discuss the behavior of immune cells in a complex environment composed of multiple directional cues.
Collapse
Affiliation(s)
- Doriane Vesperini
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany
- Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| | - Galia Montalvo
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany
- Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, 66421 Homburg, Germany
- Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| | - Franziska Lautenschläger
- Department of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany
- Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
43
|
Cruz-Zárate D, López-Ortega O, Girón-Pérez DA, Gonzalez-Suarez AM, García-Cordero JL, Schnoor M, Santos-Argumedo L. Myo1g is required for efficient adhesion and migration of activated B lymphocytes to inguinal lymph nodes. Sci Rep 2021; 11:7197. [PMID: 33785780 PMCID: PMC8009870 DOI: 10.1038/s41598-021-85477-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/18/2021] [Indexed: 11/15/2022] Open
Abstract
Cell migration is a dynamic process that involves adhesion molecules and the deformation of the moving cell that depends on cytoskeletal remodeling and actin-modulating proteins such as myosins. In this work, we analyzed the role of the class I Myosin-1 g (Myo1g) in migratory processes of LPS + IL-4 activated B lymphocytes in vivo and in vitro. In vivo, the absence of Myo1g reduced homing of activated B lymphocytes into the inguinal lymph node. Using microchannel chambers and morphology analysis, we found that the lack of Myo1g caused adhesion and chemotaxis defects. Additionally, deficiency in Myo1g causes flaws in adopting a migratory morphology. Our results highlight the importance of Myo1g during B cell migration.
Collapse
Affiliation(s)
- D Cruz-Zárate
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, 07360, Mexico City, Mexico
- Departmento and Posgrado en Inmunologia, Escuela Nacional de Ciencias Biologicas del Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - O López-Ortega
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, 07360, Mexico City, Mexico
| | - D A Girón-Pérez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, 07360, Mexico City, Mexico
| | - A M Gonzalez-Suarez
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Monterrey, NL, Mexico
| | - J L García-Cordero
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Monterrey, NL, Mexico
| | - M Schnoor
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, 07360, Mexico City, Mexico
| | - L Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, 07360, Mexico City, Mexico.
| |
Collapse
|
44
|
Robertson TF, Chengappa P, Gomez Atria D, Wu CF, Avery L, Roy NH, Maillard I, Petrie RJ, Burkhardt JK. Lymphocyte egress signal sphingosine-1-phosphate promotes ERM-guided, bleb-based migration. J Cell Biol 2021; 220:211919. [PMID: 33764397 PMCID: PMC8006814 DOI: 10.1083/jcb.202007182] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/07/2021] [Accepted: 03/03/2021] [Indexed: 02/04/2023] Open
Abstract
Ezrin, radixin, and moesin (ERM) family proteins regulate cytoskeletal responses by tethering the plasma membrane to the underlying actin cortex. Mutations in ERM proteins lead to severe combined immunodeficiency, but the function of these proteins in T cells remains poorly defined. Using mice in which T cells lack all ERM proteins, we demonstrate a selective role for these proteins in facilitating S1P-dependent egress from lymphoid organs. ERM-deficient T cells display defective S1P-induced migration in vitro, despite normal responses to standard protein chemokines. Analysis of these defects revealed that S1P promotes a fundamentally different mode of migration than chemokines, characterized by intracellular pressurization and bleb-based motility. ERM proteins facilitate this process, controlling directional migration by limiting blebbing to the leading edge. We propose that the distinct modes of motility induced by S1P and chemokines are specialized to allow T cell migration across lymphatic barriers and through tissue stroma, respectively.
Collapse
Affiliation(s)
- Tanner F Robertson
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Daniela Gomez Atria
- Division of Hematology-Oncology, Department of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Christine F Wu
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Lyndsay Avery
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nathan H Roy
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Ryan J Petrie
- Department of Biology, Drexel University, Philadelphia, PA
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
45
|
Belotti Y, McGloin D, Weijer CJ. Effects of spatial confinement on migratory properties of Dictyostelium discoideum cells. Commun Integr Biol 2021; 14:5-14. [PMID: 33552382 PMCID: PMC7849737 DOI: 10.1080/19420889.2021.1872917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Migratory environments of various eukaryotic cells, such as amoeba, leukocytes and cancer cells, typically involve spatial confinement. Numerous studies have recently emerged, aimed to develop experimental platforms that better recapitulate the characteristics of the cellular microenvironment. Using microfluidic technologies, we show that increasing confinement of Dictyostelium discoideum cells into narrower micro-channels resulted in a significant change in the mode of migration and associated arrangement of the actomyosin cytoskeleton. We observed that cells tended to migrate at constant speed, the magnitude of which was dependent on the size of the channels, as was the locomotory strategy adopted by each cell. Two different migration modes were observed, pseudopod-based and bleb-based migration, with bleb based migration being more frequent with increasing confinement and leading to slower migration. Beside the migration mode, we found that the major determinants of cell speed are its protrusion rate, the amount of F-actin at its leading edge and the number of actin foci. Our results highlighted the impact of the microenvironments on cell behavior. Furthermore, we developed a novel quantitative movement analysis platform for mono-dimensional cell migration that allows for standardization and simplification of the experimental conditions and aids investigation of the complex and dynamic processes occurring at the single-cell level.
Collapse
Affiliation(s)
- Yuri Belotti
- School of Science and Engineering, University of Dundee, Dundee, Scotland, UK
| | - David McGloin
- School of Science and Engineering, University of Dundee, Dundee, Scotland, UK
| | | |
Collapse
|
46
|
Affiliation(s)
- Karl-Gösta Sundqvist
- Department of Laboratory Medicine, Division of Clinical Immunology, Karolinska Institute and Clinical Immunology and Transfusion Medicine Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
47
|
Sung DC, Ahmad M, Lerma Cervantes CB, Zhang Y, Adelstein RS, Ma X. Mutations in non-muscle myosin 2A disrupt the actomyosin cytoskeleton in Sertoli cells and cause male infertility. Dev Biol 2020; 470:49-61. [PMID: 33188738 DOI: 10.1016/j.ydbio.2020.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
Mutations in non-muscle myosin 2A (NM2A) encompass a wide spectrum of anomalies collectively known as MYH9-Related Disease (MYH9-RD) in humans that can include macrothrombocytopenia, glomerulosclerosis, deafness, and cataracts. We previously created mouse models of the three mutations most frequently found in humans: R702C, D1424N, and E1841K. While homozygous R702C and D1424N mutations are embryonic lethal, we found homozygous mutant E1841K mice to be viable. However the homozygous male, but not female, mice were infertile. Here, we report that these mice have reduced testis size and defects in actin-associated junctions in Sertoli cells, resulting in inability to form the blood-testis barrier and premature germ cell loss. Moreover, compound double heterozygous (R702C/E1841K and D1424/E1841K) males show the same abnormalities in testes as E1841K homozygous males. Conditional ablation of either NM2A or NM2B alone in Sertoli cells has no effect on fertility and testis size, however deletion of both NM2A and NM2B in Sertoli cells results in infertility. Isolation of mutant E1841K Sertoli cells reveals decreased NM2A and F-actin colocalization and thicker NM2A filaments. Furthermore, AE1841K/AE1841K and double knockout Sertoli cells demonstrate microtubule disorganization and increased tubulin acetylation, suggesting defects in the microtubule cytoskeleton. Together, these results demonstrate that NM2A and 2B paralogs play redundant roles in Sertoli cells and are essential for testes development and normal fertility.
Collapse
Affiliation(s)
- Derek C Sung
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Mohsin Ahmad
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Connie B Lerma Cervantes
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Yingfan Zhang
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Robert S Adelstein
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States
| | - Xuefei Ma
- Laboratory of Molecular Cardiology, Cell and Developmental Biology Center, Bethesda, MD, 20892-1583, United States; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892-1583, United States.
| |
Collapse
|
48
|
Thomos M, Wurzel P, Scharf S, Koch I, Hansmann ML. 3D investigation shows walls and wall-like structures around human germinal centres, probably regulating T- and B-cell entry and exit. PLoS One 2020; 15:e0242177. [PMID: 33170900 PMCID: PMC7654765 DOI: 10.1371/journal.pone.0242177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/27/2020] [Indexed: 11/19/2022] Open
Abstract
This study deals with 3D laser investigation on the border between the human lymph node T-zone and germinal centre. Only a few T-cells specific for antigen selected B-cells are allowed to enter germinal centres. This selection process is guided by sinus structures, chemokine gradients and inherent motility of the lymphoid cells. We measured gaps and wall-like structures manually, using IMARIS, a 3D image software for analysis and interpretation of microscopy datasets. In this paper, we describe alpha-actin positive and semipermeable walls and wall-like structures that may hinder T-cells and other cell types from entering germinal centres. Some clearly defined holes or gaps probably regulate lymphoid traffic between T- and B-cell areas. In lymphadenitis, the morphology of this border structure is clearly defined. However, in case of malignant lymphoma, the wall-like structure is disrupted. This has been demonstrated exemplarily in case of angioimmunoblastic T-cell lymphoma. We revealed significant differences of lengths of the wall-like structures in angioimmunoblastic T-cell lymphoma in comparison with wall-like structures in reactive tissue slices. The alterations of morphological structures lead to abnormal and less controlled T- and B-cell distributions probably preventing the immune defence against tumour cells and infectious agents by dysregulating immune homeostasis.
Collapse
Affiliation(s)
- Miguel Thomos
- Reference and Consultant Center of Lymph Node and Lymphoma Pathology at Dr. Senckenberg Institute for Pathology, Goethe-Universität Frankfurt am Main, Frankfurt/Main, Hessen, Germany
| | - Patrick Wurzel
- Department of Molecular Bioinformatics, Johann Wolfgang Goethe-University Frankfurt/Main, Frankfurt/Main, Hessen, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt/Main, Hessen, Germany
| | - Sonja Scharf
- Department of Molecular Bioinformatics, Johann Wolfgang Goethe-University Frankfurt/Main, Frankfurt/Main, Hessen, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt/Main, Hessen, Germany
| | - Ina Koch
- Department of Molecular Bioinformatics, Johann Wolfgang Goethe-University Frankfurt/Main, Frankfurt/Main, Hessen, Germany
| | - Martin-Leo Hansmann
- Reference and Consultant Center of Lymph Node and Lymphoma Pathology at Dr. Senckenberg Institute for Pathology, Goethe-Universität Frankfurt am Main, Frankfurt/Main, Hessen, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt/Main, Hessen, Germany
| |
Collapse
|
49
|
Otterpohl KL, Busselman BW, Ratnayake I, Hart RG, Hart KR, Evans CM, Phillips CL, Beach JR, Ahrenkiel P, Molitoris BA, Surendran K, Chandrasekar I. Conditional Myh9 and Myh10 inactivation in adult mouse renal epithelium results in progressive kidney disease. JCI Insight 2020; 5:138530. [PMID: 33001861 PMCID: PMC7710296 DOI: 10.1172/jci.insight.138530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/23/2020] [Indexed: 01/07/2023] Open
Abstract
Actin-associated nonmuscle myosin II (NM2) motor proteins play critical roles in a myriad of cellular functions, including endocytosis and organelle transport pathways. Cell type-specific expression and unique subcellular localization of the NM2 proteins, encoded by the Myh9 and Myh10 genes, in the mouse kidney tubules led us to hypothesize that these proteins have specialized functional roles within the renal epithelium. Inducible conditional knockout (cKO) of Myh9 and Myh10 in the renal tubules of adult mice resulted in progressive kidney disease. Prior to overt renal tubular injury, we observed intracellular accumulation of the glycosylphosphatidylinositol-anchored protein uromodulin (UMOD) and gradual loss of Na+ K+ 2Cl- cotransporter from the apical membrane of the thick ascending limb epithelia. The UMOD accumulation coincided with expansion of endoplasmic reticulum (ER) tubules and activation of ER stress and unfolded protein response pathways in Myh9&10-cKO kidneys. We conclude that NM2 proteins are required for localization and transport of UMOD and loss of function results in accumulation of UMOD and ER stress-mediated progressive renal tubulointerstitial disease. These observations establish cell type-specific role(s) for NM2 proteins in regulation of specialized renal epithelial transport pathways and reveal the possibility that human kidney disease associated with MYH9 mutations could be of renal epithelial origin.
Collapse
Affiliation(s)
- Karla L. Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Brook W. Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, USA
| | - Ishara Ratnayake
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota, USA
| | - Ryan G. Hart
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Kimberly R. Hart
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
| | - Claire M. Evans
- Histology and Imaging Core, Sanford Research, Sioux Falls, South Dakota, USA
| | - Carrie L. Phillips
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jordan R. Beach
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - Phil Ahrenkiel
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota, USA
| | - Bruce A. Molitoris
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kameswaran Surendran
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
| |
Collapse
|
50
|
Oliveira MMS, Westerberg LS. Cytoskeletal regulation of dendritic cells: An intricate balance between migration and presentation for tumor therapy. J Leukoc Biol 2020; 108:1051-1065. [PMID: 32557835 DOI: 10.1002/jlb.1mr0520-014rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/28/2022] Open
Abstract
Dendritic cells (DCs) are the main players in many approaches for cancer therapy. The idea with DC tumor therapy is to promote activation of tumor infiltrating cytotoxic T cells that kill tumor cells. This requires that DCs take up tumor Ag and present peptides on MHC class I molecules in a process called cross-presentation. For this process to be efficient, DCs have to migrate to the tumor draining lymph node and there activate the machinery for cross-presentation. In this review, we will discuss recent progress in understanding the role of actin regulators for control of DC migration and Ag presentation. The potential to target actin regulators for better DC-based tumor therapy will also be discussed.
Collapse
Affiliation(s)
- Mariana M S Oliveira
- Department of Microbiology Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Lisa S Westerberg
- Department of Microbiology Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|