Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Eur J Immunol ; 54(6): e2350761, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38566526

ABSTRACT

In multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), early pathological features include immune cell infiltration into the central nervous system (CNS) and blood-brain barrier (BBB) disruption. We investigated the role of junctional adhesion molecule-A (JAM-A), a tight junction protein, in active EAE (aEAE) pathogenesis. Our study confirms JAM-A expression at the blood-brain barrier and its luminal redistribution during aEAE. JAM-A deficient (JAM-A-/-) C57BL/6J mice exhibited milder aEAE, unrelated to myelin oligodendrocyte glycoprotein-specific CD4+ T-cell priming. While JAM-A absence influenced macrophage behavior on primary mouse brain microvascular endothelial cells (pMBMECs) under flow in vitro, it did not impact T-cell extravasation across primary mouse brain microvascular endothelial cells. At aEAE onset, we observed reduced lymphocyte and CCR2+ macrophage infiltration into the spinal cord of JAM-A-/- mice compared to control littermates. This correlated with increased CD3+ T-cell accumulation in spinal cord perivascular spaces and brain leptomeninges, suggesting JAM-A absence leads to T-cell trapping in central nervous system border compartments. In summary, JAM-A plays a role in immune cell infiltration and clinical disease progression in aEAE.


Subject(s)
Blood-Brain Barrier , Encephalomyelitis, Autoimmune, Experimental , Endothelial Cells , Macrophages , Mice, Inbred C57BL , Mice, Knockout , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Macrophages/immunology , Macrophages/metabolism , Endothelial Cells/metabolism , Endothelial Cells/immunology , Spinal Cord/pathology , Spinal Cord/immunology , Spinal Cord/metabolism , CD4-Positive T-Lymphocytes/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal
2.
Cancers (Basel) ; 15(20)2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37894438

ABSTRACT

Melanoma frequently metastasises to the brain, and a detailed understanding of the molecular and cellular mechanisms underlying melanoma cell extravasation across the blood-brain barrier (BBB) is important for preventing brain metastasis formation. Making use of primary mouse brain microvascular endothelial cells (pMBMECs) as an in vitro BBB model, we imaged the interaction of melanoma cells into pMBMEC monolayers. We observed exclusive junctional intercalation of melanoma cells and confirmed that melanoma-induced pMBMEC barrier disruption can be rescued by protease inhibition. Interleukin (IL)-1ß stimulated pMBMECs or PECAM-1-knockout (-ko) pMBMECs were employed to model compromised BBB barrier properties in vitro and to determine increased melanoma cell intercalation compared to pMBMECs with intact junctions. The newly generated brain-homing melanoma cell line YUMM1.1-BrM4 was used to reveal increased in vivo extravasation of melanoma cells across the BBB of barrier-compromised PECAM-1-deficient mice compared to controls. Taken together, our data indicate that preserving BBB integrity is an important measure to limit the formation of melanoma-brain metastasis.

3.
Article in English | MEDLINE | ID: mdl-36617644

ABSTRACT

The endothelial blood-brain barrier (BBB) protects central nervous system (CNS) neurons from the changeable milieu of the bloodstream by strictly controlling the movement of molecules and immune cells between the blood and the CNS. Immune cell migration across the vascular wall is a multistep process regulated by the sequential interaction of different signaling and adhesion molecules on the endothelium and the immune cells. Accounting for its unique barrier properties and trafficking molecule expression profile, particular adaptions in immune cell migration across the BBB have been observed. Thus, in vitro models of the BBB are desirable to explore the precise cellular and molecular mechanisms involved in immune cell trafficking across the BBB. The challenge to overcome is that barrier properties of brain microvascular endothelial cells are not intrinsic and readily lost in culture. With a focus on human in vitro BBB models, we here discuss the suitability of available in vitro models for the BBB for exploring the specific mechanisms involved in immune cell trafficking across the BBB.


Subject(s)
Brain , Endothelial Cells , Humans , Brain/metabolism , Blood-Brain Barrier/metabolism , Central Nervous System , Cell Movement
4.
Sci Transl Med ; 14(661): eaax8933, 2022 09 07.
Article in English | MEDLINE | ID: mdl-36070364

ABSTRACT

Brain metastasis is a complication of increasing incidence in patients with breast cancer at advanced disease stage. It is a severe condition characterized by a rapid decline in quality of life and poor prognosis. There is a critical clinical need to develop effective therapies to prevent and treat brain metastases. Here, we describe a unique and robust spontaneous preclinical model of breast cancer metastasis to the brain (4T1-BM2) in mice that has been instrumental in uncovering molecular mechanisms guiding metastatic dissemination and colonization of the brain. Key experimental findings were validated in the additional murine D2A1-BM2 model and in human MDA231-BrM2 model. Gene expression analyses and functional studies, coupled with clinical transcriptomic and histopathological investigations, identified connexins (Cxs) and focal adhesion kinase (FAK) as master molecules orchestrating breast cancer colonization of the brain. Cx31 promoted homotypic tumor cell adhesion, heterotypic tumor-astrocyte interaction, and FAK phosphorylation. FAK signaling prompted NF-κB activation inducing Lamc2 expression and laminin 332 (laminin 5) deposition, α6 integrin-mediated adhesion, and sustained survival and growth within brain parenchyma. In the MDA231-BrM2 model, the human homologous molecules CX43, LAMA4, and α3 integrin were involved. Systemic treatment with FAK inhibitors reduced brain metastasis progression. In conclusion, we report a spontaneous model of breast cancer metastasis to the brain and identified Cx-mediated FAK-NF-κB signaling as a mechanism promoting cell-autonomous and microenvironmentally controlled cell survival for brain colonization. Considering the limited therapeutic options for brain metastatic disease in cancer patients, we propose FAK as a therapeutic candidate to further pursue in the clinic.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Animals , Brain/metabolism , Breast Neoplasms/genetics , Connexins/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Melanoma , Mice , NF-kappa B/metabolism , Quality of Life , Skin Neoplasms , Melanoma, Cutaneous Malignant
5.
Eur J Immunol ; 52(1): 161-177, 2022 01.
Article in English | MEDLINE | ID: mdl-34524684

ABSTRACT

The migration of CD4+ effector/memory T cells across the blood-brain barrier (BBB) is a critical step in MS or its animal model, EAE. T-cell diapedesis across the BBB can occur paracellular, via the complex BBB tight junctions or transcellular via a pore through the brain endothelial cell body. Making use of primary mouse brain microvascular endothelial cells (pMBMECs) as in vitro model of the BBB, we here directly compared the transcriptome profile of pMBMECs favoring transcellular or paracellular T-cell diapedesis by RNA sequencing (RNA-seq). We identified the atypical chemokine receptor 1 (Ackr1) as one of the main candidate genes upregulated in pMBMECs favoring transcellular T-cell diapedesis. We confirmed upregulation of ACKR1 protein in pMBMECs promoting transcellular T-cell diapedesis and in venular endothelial cells in the CNS during EAE. Lack of endothelial ACKR1 reduced transcellular T-cell diapedesis across pMBMECs under physiological flow in vitro. Combining our previous observation that endothelial ACKR1 contributes to EAE pathogenesis by shuttling chemokines across the BBB, the present data support that ACKR1 mediated chemokine shuttling enhances transcellular T-cell diapedesis across the BBB during autoimmune neuroinflammation.


Subject(s)
Blood-Brain Barrier , CD4-Positive T-Lymphocytes , Duffy Blood-Group System , Encephalomyelitis, Autoimmune, Experimental , Memory T Cells , Multiple Sclerosis , Receptors, Cell Surface , Transendothelial and Transepithelial Migration , Animals , Mice , Blood-Brain Barrier/immunology , CD4-Positive T-Lymphocytes/immunology , Duffy Blood-Group System/genetics , Duffy Blood-Group System/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/genetics , Inflammation/immunology , Memory T Cells/immunology , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Transendothelial and Transepithelial Migration/genetics , Transendothelial and Transepithelial Migration/immunology
6.
J Invest Dermatol ; 142(4): 1171-1182.e10, 2022 04.
Article in English | MEDLINE | ID: mdl-34757069

ABSTRACT

The STK11/LKB1 is a tumor suppressor involved in metabolism and cell motility. In BRAFV600E melanoma, STK11 is inactivated by extracellular signal‒regulated kinase and RSK, preventing it from binding and activating adenosine monophosphate-activated protein kinase and promoting melanoma cell proliferation. Although STK11 mutations occur in 5‒10% of cutaneous melanoma, few functional studies have been performed. By knocking out STK11 with CRISPR/Cas9 in two human BRAF-mutant melanoma cell lines, we found that STK11 loss reduced the sensitivity to a BRAF inhibitor. More strikingly, STK11 loss led to an increased invasive phenotype in both three-dimensional spheroids and in vivo zebrafish xenograft models. STK11 overexpression consistently reversed the invasive phenotype. Interestingly, STK11 knockout increased invasion also in an NRAS-mutant melanoma cell line. Furthermore, although STK11 was expressed in primary human melanoma tumors, its expression significantly decreased in melanoma metastases, especially in brain metastases. In the STK11-knockout cells, we observed increased activating phosphorylation of signal transducer and activator of transcription 3/5 and FAK. Using inhibitors of signal transducer and activator of transcription 3/5 and FAK, we reversed the invasive phenotype in both BRAF- and NRAS-mutated cells. Our findings confirm an increased invasive phenotype on STK11 inactivation in BRAF- and NRAS-mutant cutaneous melanoma that can be targeted by signal transducer and activator of transcription 3/5 and FAK inhibition.


Subject(s)
Melanoma , Skin Neoplasms , AMP-Activated Protein Kinase Kinases , Animals , Cell Line, Tumor , Focal Adhesion Kinase 1 , Humans , Melanoma/pathology , Mutation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , STAT3 Transcription Factor/metabolism , Skin Neoplasms/genetics , Zebrafish/metabolism , Melanoma, Cutaneous Malignant
7.
J Autoimmun ; 119: 102610, 2021 05.
Article in English | MEDLINE | ID: mdl-33621930

ABSTRACT

CD4+ T cell trafficking is a fundamental property of adaptive immunity. In this study, we uncover a novel role for histone deacetylase 1 (HDAC1) in controlling effector CD4+ T cell migration, thereby providing mechanistic insight into why a T cell-specific deletion of HDAC1 protects against experimental autoimmune encephalomyelitis (EAE). HDAC1-deficient CD4+ T cells downregulated genes associated with leukocyte extravasation. In vitro, HDAC1-deficient CD4+ T cells displayed aberrant morphology and migration on surfaces coated with integrin LFA-1 ligand ICAM-1 and showed an impaired ability to arrest on and to migrate across a monolayer of primary mouse brain microvascular endothelial cells under physiological flow. Moreover, HDAC1 deficiency reduced homing of CD4+ T cells into the intestinal epithelium and lamina propria preventing weight-loss, crypt damage and intestinal inflammation in adoptive CD4+ T cell transfer colitis. This correlated with reduced expression levels of LFA-1 integrin chains CD11a and CD18 as well as of selectin ligands CD43, CD44 and CD162 on transferred circulating HDAC1-deficient CD4+ T cells. Our data reveal that HDAC1 controls T cell-mediated autoimmunity via the regulation of CD4+ T cell trafficking into the CNS and intestinal tissues.


Subject(s)
Autoimmunity , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chemotaxis, Leukocyte/immunology , Histone Deacetylase 1/metabolism , Inflammation/etiology , Inflammation/metabolism , Animals , Biomarkers , Cell Adhesion , Chemotaxis, Leukocyte/genetics , Disease Models, Animal , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelial Cells , Gene Expression Profiling , Gene Expression Regulation , Histone Deacetylase 1/genetics , Immunohistochemistry , Inflammation/diagnosis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout
8.
Sci Rep ; 9(1): 10702, 2019 Jul 18.
Article in English | MEDLINE | ID: mdl-31320665

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

9.
Front Immunol ; 10: 711, 2019.
Article in English | MEDLINE | ID: mdl-31024547

ABSTRACT

Breakdown of the blood-brain barrier (BBB) and increased immune cell trafficking into the central nervous system (CNS) are hallmarks of the pathogenesis of multiple sclerosis (MS). Platelet endothelial cell adhesion molecule-1 (PECAM-1; CD31) is expressed on cells of the vascular compartment and regulates vascular integrity and immune cell trafficking. Involvement of PECAM-1 in MS pathogenesis has been suggested by the detection of increased levels of soluble PECAM-1 (sPECAM-1) in the serum and CSF of MS patients. Here, we report profound upregulation of cell-bound PECAM-1 in initial (pre-phagocytic) white matter as well as active cortical gray matter MS lesions. Using a human in vitro BBB model we observed that PECAM-1 is not essential for the transmigration of human CD4+ T-cell subsets (Th1, Th1*, Th2, and Th17) across the BBB. Employing an additional in vitro BBB model based on primary mouse brain microvascular endothelial cells (pMBMECs) we show that the lack of endothelial PECAM-1 impairs BBB properties as shown by reduced transendothelial electrical resistance (TEER) and increases permeability for small molecular tracers. Investigating T-cell migration across the BBB under physiological flow by in vitro live cell imaging revealed that absence of PECAM-1 in pMBMECs did not influence arrest, polarization, and crawling of effector/memory CD4+ T cells on the pMBMECs. Absence of endothelial PECAM-1 also did not affect the number of T cells able to cross the pMBMEC monolayer under flow, but surprisingly favored transcellular over paracellular T-cell diapedesis. Taken together, our data demonstrate that PECAM-1 is critically involved in regulating BBB permeability and although not required for T-cell diapedesis itself, its presence or absence influences the cellular route of T-cell diapedesis across the BBB. Upregulated expression of cell-bound PECAM-1 in human MS lesions may thus reflect vascular repair mechanisms aiming to restore BBB integrity and paracellular T-cell migration across the BBB as it occurs during CNS immune surveillance.


Subject(s)
Blood-Brain Barrier/immunology , Endothelium, Vascular/metabolism , Gray Matter/immunology , Multiple Sclerosis/immunology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , White Matter/immunology , Adult , Animals , Cells, Cultured , Endothelium, Vascular/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenic Inflammation , Paracrine Communication , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Transendothelial and Transepithelial Migration , Up-Regulation
10.
Sci Rep ; 9(1): 203, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30659216

ABSTRACT

The tight junction protein claudin-3 has been identified as a transcriptional target of the Wnt/ß-catenin signaling pathway regulating blood-brain barrier (BBB) maturation. In neurological disorders loss of claudin-3 immunostaining is observed at the compromised BBB and blood-cerebrospinal fluid barrier (BCSFB). Although these observations support a central role of claudin-3 in regulating brain barriers' tight junction integrity, expression of claudin-3 at the brain barriers has remained a matter of debate. This prompted us to establish claudin-3-/- C57BL/6J mice to study the role of claudin-3 in brain barrier integrity in health and neuroinflammation. Bulk and single cell RNA sequencing and direct comparative qRT-PCR analysis of brain microvascular samples from WT and claudin-3-/- mice show beyond doubt that brain endothelial cells do not express claudin-3 mRNA. Detection of claudin-3 protein at the BBB in vivo and in vitro is rather due to junctional reactivity of anti-claudin-3 antibodies to an unknown antigen still detected in claudin-3-/- brain endothelium. We confirm expression and junctional localization of claudin-3 at the BCSFB of the choroid plexus. Our study clarifies that claudin-3 is not expressed at the BBB and shows that absence of claudin-3 does not impair brain barrier function during health and neuroinflammation in C57BL/6J mice.


Subject(s)
Blood-Brain Barrier/metabolism , Claudin-3/metabolism , Tight Junctions/metabolism , Animals , Biological Transport , Brain/metabolism , Choroid Plexus/metabolism , Claudin-3/genetics , Endothelial Cells/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Tight Junctions/genetics , Wnt Signaling Pathway/physiology
11.
J Cereb Blood Flow Metab ; 39(10): 1995-2010, 2019 10.
Article in English | MEDLINE | ID: mdl-29762071

ABSTRACT

Melanoma is the most aggressive skin cancer in humans. One severe complication is the formation of brain metastasis, which requires extravasation of melanoma cells across the tight blood-brain barrier (BBB). Previously, VLA-4 has been assigned a role for the adhesive interaction of melanoma cells with non-BBB endothelial cells. However, the role of melanoma VLA-4 for breaching the BBB remained unknown. In this study, we used a mouse in vitro BBB model and imaged the shear resistant arrest of melanoma cells on the BBB. Similar to effector T cells, inflammatory conditions of the BBB increased the arrest of melanoma cells followed by a unique post-arrest behavior lacking immediate crawling. However, over time, melanoma cells intercalated into the BBB and compromised its barrier properties. Most importantly, antibody ablation of VLA-4 abrogated melanoma shear resistant arrest on and intercalation into the BBB and protected the BBB from barrier breakdown. A tissue microarray established from human brain metastasis revealed that indeed a majority of 92% of all human melanoma brain metastases stained VLA-4 positive. We propose VLA-4 as a target for the inhibition of brain metastasis formation in the context of personalized medicine identifying metastasizing VLA-4 positive melanoma.


Subject(s)
Blood-Brain Barrier/pathology , Brain Neoplasms/secondary , Endothelial Cells/pathology , Integrin alpha4beta1/metabolism , Melanoma/pathology , Animals , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Capillary Permeability , Cell Adhesion , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/metabolism , Humans , Integrin alpha4beta1/analysis , Melanoma/metabolism , Mice, Inbred C57BL , Transendothelial and Transepithelial Migration
12.
J Immunol ; 201(9): 2731-2743, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30257886

ABSTRACT

HIV-1 Nef is a multifunctional protein that optimizes virus spread and promotes immune evasion of infected cells to accelerate disease progression in AIDS patients. As one of its activities, Nef reduces the motility of infected CD4+ T lymphocytes in confined space. In vivo, Nef restricts T lymphocyte homing to lymph nodes as it reduces the ability for extravasation at the diapedesis step. Effects of Nef on T lymphocyte motility are typically mediated by its ability to reduce actin remodeling. However, interference with diapedesis does not depend on residues in Nef required for inhibition of host cell actin dynamics. In search for an alternative mechanism by which Nef could alter T lymphocyte extravasation, we noted that the viral protein interferes with the polarization of primary human CD4+ T lymphocytes upon infection with HIV-1. Expression of Nef alone is sufficient to disrupt T cell polarization, and this effect is conserved among lentiviral Nef proteins. Nef acts by arresting the oscillation of CD4+ T cells between polarized and nonpolarized morphologies. Mapping studies identified the binding site for the Nef-associated kinase complex (NAKC) as critical determinant of this Nef activity and a NAKC-binding-deficient Nef variant fails to impair CD4+ T lymphocyte extravasation and homing to lymph nodes. These results thus imply the disruption of T lymphocyte polarity via its NAKC binding site as a novel mechanism by which lentiviral Nef proteins alter T lymphocyte migration in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Polarity/immunology , Chemotaxis, Leukocyte/immunology , Transendothelial and Transepithelial Migration/immunology , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Binding Sites , CD4-Positive T-Lymphocytes/immunology , Humans , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
13.
J Exp Med ; 215(7): 1869-1890, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29875261

ABSTRACT

T cells are actively scanning pMHC-presenting cells in lymphoid organs and nonlymphoid tissues (NLTs) with divergent topologies and confinement. How the T cell actomyosin cytoskeleton facilitates this task in distinct environments is incompletely understood. Here, we show that lack of Myosin IXb (Myo9b), a negative regulator of the small GTPase Rho, led to increased Rho-GTP levels and cell surface stiffness in primary T cells. Nonetheless, intravital imaging revealed robust motility of Myo9b-/- CD8+ T cells in lymphoid tissue and similar expansion and differentiation during immune responses. In contrast, accumulation of Myo9b-/- CD8+ T cells in NLTs was strongly impaired. Specifically, Myo9b was required for T cell crossing of basement membranes, such as those which are present between dermis and epidermis. As consequence, Myo9b-/- CD8+ T cells showed impaired control of skin infections. In sum, we show that Myo9b is critical for the CD8+ T cell adaptation from lymphoid to NLT surveillance and the establishment of protective tissue-resident T cell populations.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Myosins/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Movement , Cell Polarity , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Epidermis/pathology , Epidermis/virology , Extracellular Matrix/metabolism , Immunity , Lymphocyte Activation/immunology , Lymphoid Tissue/metabolism , Mice, Inbred C57BL , Myosins/deficiency , Receptors, Lymphocyte Homing/metabolism , rho GTP-Binding Proteins/metabolism
14.
FASEB J ; 32(9): 4824-4835, 2018 09.
Article in English | MEDLINE | ID: mdl-29589978

ABSTRACT

The ζ-associated protein of 70 kDa (ZAP70) is expressed in the aggressive form of B-cell chronic lymphocytic leukemia (CLL). Moreover, the integrin very late antigen (VLA)-1 is highly expressed on subtypes of CLL that are associated with high proliferation rates in the lymph node context. We herein identify a critical role for ZAP70 in chemokine-mediated, inside-out signaling to integrins in trisomy 12 carrying Ohio State University-CLL cell lines derived from a patient with previously treated CLL. We found that ZAP70-positive CLL cells migrated significantly better toward ligands of the lymph node homing chemokine receptors CCR7 and CXCR4 compared with ZAP70-negative cells. In addition, ZAP70-expressing CLL cells adhered more efficiently to integrin ligands under static conditions. We discovered that ZAP70 expression controls chemokine-driven clustering of the integrins VLA-4 and lymphocyte function-associated antigen-1. More precisely, chemokine stimulation resulted in a ZAP70-dependent integrin valency regulation on CLL cells, whereas high-affinity regulation of integrins was independent of ZAP70. Consequently, ZAP70-expressing CLL cells show increased chemokine-driven arrest on immobilized integrin ligands and on chemokine-presenting endothelial cells under physiologic flow conditions. Hence, we describe a novel mechanism showing how ZAP70 controls chemokine-driven valency regulation of integrins and arrest of CLL cells on endothelial cells, a process that might contribute to CLL disease progression.-Laufer, J. M., Lyck, R., Legler, D. F. ZAP70 expression enhances chemokine-driven chronic lymphocytic leukemia cell migration and arrest by valency regulation of integrins.


Subject(s)
Cell Movement/physiology , Integrins/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , B-Lymphocytes/metabolism , Chemokines/metabolism , Humans , Lymph Nodes/metabolism , Signal Transduction/physiology
15.
Elife ; 62017 10 04.
Article in English | MEDLINE | ID: mdl-28975893

ABSTRACT

Aberrant alternative pre-mRNA splicing (AS) events have been associated with several disorders. However, it is unclear whether deregulated AS directly contributes to disease. Here, we reveal a critical role of the AS regulator epithelial splicing regulator protein 1 (ESRP1) for intestinal homeostasis and pathogenesis. In mice, reduced ESRP1 function leads to impaired intestinal barrier integrity, increased susceptibility to colitis and altered colorectal cancer (CRC) development. Mechanistically, these defects are produced in part by modified expression of ESRP1-specific Gpr137 isoforms differently activating the Wnt pathway. In humans, ESRP1 is downregulated in inflamed biopsies from inflammatory bowel disease patients. ESRP1 loss is an adverse prognostic factor in CRC. Furthermore, generation of ESRP1-dependent GPR137 isoforms is altered in CRC and expression of a specific GPR137 isoform predicts CRC patient survival. These findings indicate a central role of ESRP1-regulated AS for intestinal barrier integrity. Alterations in ESRP1 function or expression contribute to intestinal pathology.


Subject(s)
Alternative Splicing , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/physiopathology , RNA-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Gene Expression Regulation , Humans , Mice
16.
ACS Appl Mater Interfaces ; 9(22): 18501-18511, 2017 Jun 07.
Article in English | MEDLINE | ID: mdl-28517937

ABSTRACT

A major hurdle in the development of biomedical nanoparticles (NP) is understanding how they interact with complex biological systems and navigate biological barriers to arrive at pathological targets. It is becoming increasingly evident that merely controlling particle physicochemical properties may not be sufficient to mediate particle biodistribution in dynamic environments. Thus, researchers are increasingly turning toward more complex but likewise more physiological in vitro systems to study particle--cell/particle-system interactions. An emerging paradigm is to utilize naturally migratory cells to act as so-called "Trojan horses" or cellular shuttles. We report here the use of monocytes/macrophages to transport NP across a confluent endothelial cell layer using a microfluidic in vitro model. With a custom-built flow chamber, we showed that physiological shear stress, when compared to low flow or static conditions, increased NP uptake by macrophages. We further provided a mathematical explanation for the effect of flow on NP uptake, namely that the physical exposure times of NP to cells is dictated by shear stress (i.e., flow rate) and results in increased particle uptake under flow. This study was extended to a multicellular, hydrodynamic in vitro model. Because monocytes are cells that naturally translocate across biological barriers, we utilized a monocyte/macrophage cell line as cellular NP transporters across an endothelial layer. In this exploratory study, we showed that monocyte/macrophage cells adhere to an endothelial layer and dynamically interact with the endothelial cells. The monocytes/macrophages took up NP and diapedesed across the endothelial layer with NP accumulating within the cellular uropod. These data illustrate that monocytes/macrophages may therefore act as active shuttles to deliver particles across endothelial barriers.


Subject(s)
Nanoparticles , Macrophages , Monocytes , Particle Size , Tissue Distribution
17.
J Cereb Blood Flow Metab ; 37(8): 2894-2909, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28273717

ABSTRACT

Activated leukocyte cell adhesion molecule (ALCAM) has been proposed to mediate leukocyte migration across the blood-brain barrier (BBB) in multiple sclerosis or experimental autoimmune encephalomyelitis (EAE). Here, we confirmed vascular ALCAM expression in human brain tissue samples in situ and on two different human in vitro BBB models. Antibody-mediated inhibition of ALCAM reduced diapedesis of human CD4+ Th1 but not of Th17 cells across the human BBB in vitro. In accordance to human Th1 cells, mouse Th1 cells showed reduced diapedesis across an ALCAM-/- in vitro BBB model under static but no longer under flow conditions. In contrast to the limited role of ALCAM in T cell extravasation across the BBB, we found a contribution of ALCAM to rolling, adhesion, and diapedesis of human CD14+ monocytes across the human BBB under flow and static conditions. Taken together, our study highlights the potential differences in the CNS expression of ALCAM in mouse and human and supports a prominent role for ALCAM in the multi-step extravasation of monocytes across the BBB.


Subject(s)
Antigens, CD/metabolism , Blood-Brain Barrier/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Fetal Proteins/metabolism , Monocytes/immunology , T-Lymphocytes/immunology , Transendothelial and Transepithelial Migration/immunology , Animals , Antigens, CD/genetics , Blood-Brain Barrier/immunology , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Fetal Proteins/genetics , Humans , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , T-Lymphocytes/metabolism , Transendothelial and Transepithelial Migration/physiology
19.
Proc Natl Acad Sci U S A ; 114(4): E524-E533, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28069965

ABSTRACT

Activated leukocyte cell adhesion molecule (ALCAM) is a cell adhesion molecule found on blood-brain barrier endothelial cells (BBB-ECs) that was previously shown to be involved in leukocyte transmigration across the endothelium. In the present study, we found that ALCAM knockout (KO) mice developed a more severe myelin oligodendrocyte glycoprotein (MOG)35-55-induced experimental autoimmune encephalomyelitis (EAE). The exacerbated disease was associated with a significant increase in the number of CNS-infiltrating proinflammatory leukocytes compared with WT controls. Passive EAE transfer experiments suggested that the pathophysiology observed in active EAE was linked to the absence of ALCAM on BBB-ECs. In addition, phenotypic characterization of unimmunized ALCAM KO mice revealed a reduced expression of BBB junctional proteins. Further in vivo, in vitro, and molecular analysis confirmed that ALCAM is associated with tight junction molecule assembly at the BBB, explaining the increased permeability of CNS blood vessels in ALCAM KO animals. Collectively, our data point to a biologically important function of ALCAM in maintaining BBB integrity.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelial Cells/metabolism , Activated-Leukocyte Cell Adhesion Molecule/genetics , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Homeostasis , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Severity of Illness Index , Spinal Cord/metabolism , Tight Junction Proteins/metabolism
20.
Nat Immunol ; 17(9): 1075-83, 2016 09.
Article in English | MEDLINE | ID: mdl-27400149

ABSTRACT

Adhesion and migration of T cells are controlled by chemokines and by adhesion molecules, especially integrins, and have critical roles in the normal physiological function of T lymphocytes. Using an RNA-mediated interference screen, we identified the WNK1 kinase as a regulator of both integrin-mediated adhesion and T cell migration. We found that WNK1 is a negative regulator of integrin-mediated adhesion, whereas it acts as a positive regulator of migration via the kinases OXSR1 and STK39 and the ion co-transporter SLC12A2. WNK1-deficient T cells home less efficiently to lymphoid organs and migrate more slowly through them. Our results reveal that a pathway previously known only to regulate salt homeostasis in the kidney functions to balance T cell adhesion and migration.


Subject(s)
Cell Adhesion/genetics , Cell Movement/genetics , Minor Histocompatibility Antigens/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/physiology , Animals , Cells, Cultured , Homeostasis , Ion Transport , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptors, Lymphocyte Homing/genetics , Solute Carrier Family 12, Member 2/metabolism , WNK Lysine-Deficient Protein Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL