Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
PLoS Biol ; 19(8): e3001339, 2021 08.
Article in English | MEDLINE | ID: mdl-34347789

ABSTRACT

How cytotoxic lymphocytes are protected against their own weapons during close combat with diseased target cells is an important and long-standing question in immunology. A study in this issue provides new insights into the mechanisms by which natural killer (NK) cells avoid self-destruction.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural
3.
Front Immunol ; 12: 619069, 2021.
Article in English | MEDLINE | ID: mdl-34108958

ABSTRACT

Natural killer (NK) cells are innate effector lymphocytes with strong antitumor effects against hematologic malignancies such as chronic lymphocytic leukemia (CLL). However, NK cells fail to control CLL progression on the long term. For effective lysis of their targets, NK cells use a specific cell-cell interface, known as the immunological synapse (IS), whose assembly and effector function critically rely on dynamic cytoskeletal changes in NK cells. Here we explored the role of CLL cell actin cytoskeleton during NK cell attack. We found that CLL cells can undergo fast actin cytoskeleton remodeling which is characterized by a NK cell contact-induced accumulation of actin filaments at the IS. Such polarization of the actin cytoskeleton was strongly associated with resistance against NK cell-mediated cytotoxicity and reduced amounts of the cell-death inducing molecule granzyme B in target CLL cells. Selective pharmacological targeting of the key actin regulator Cdc42 abrogated the capacity of CLL cells to reorganize their actin cytoskeleton during NK cell attack, increased levels of transferred granzyme B and restored CLL cell susceptibility to NK cell cytotoxicity. This resistance mechanism was confirmed in primary CLL cells from patients. In addition, pharmacological inhibition of actin dynamics in combination with blocking antibodies increased conjugation frequency and improved CLL cell elimination by NK cells. Together our results highlight the critical role of CLL cell actin cytoskeleton in driving resistance against NK cell cytotoxicity and provide new potential therapeutic point of intervention to target CLL immune escape.


Subject(s)
Actin Cytoskeleton/metabolism , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , cdc42 GTP-Binding Protein/antagonists & inhibitors , Actin Cytoskeleton/drug effects , Biomarkers , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Fluorescent Antibody Technique , HLA-G Antigens/immunology , Humans , Immunological Synapses/immunology , Immunological Synapses/metabolism , Immunophenotyping , Killer Cells, Natural/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
4.
Front Immunol ; 11: 581119, 2020.
Article in English | MEDLINE | ID: mdl-33240268

ABSTRACT

Congenital defects of the immune system called primary immunodeficiency disorders (PID) describe a group of diseases characterized by a decrease, an absence, or a malfunction of at least one part of the immune system. As a result, PID patients are more prone to develop life-threatening complications, including cancer. PID currently include over 400 different disorders, however, the variety of PID-related cancers is narrow. We discuss here reasons for this clinical phenotype. Namely, PID can lead to cell intrinsic failure to control cell transformation, failure to activate tumor surveillance by cytotoxic cells or both. As the most frequent tumors seen among PID patients stem from faulty lymphocyte development leading to leukemia and lymphoma, we focus on the extensive genomic alterations needed to create the vast diversity of B and T lymphocytes with potential to recognize any pathogen and why defects in these processes lead to malignancies in the immunodeficient environment of PID patients. In the second part of the review, we discuss PID affecting tumor surveillance and especially membrane trafficking defects caused by altered exocytosis and regulation of the actin cytoskeleton. As an impairment of these membrane trafficking pathways often results in dysfunctional effector immune cells, tumor cell immune evasion is elevated in PID. By considering new anti-cancer treatment concepts, such as transfer of genetically engineered immune cells, restoration of anti-tumor immunity in PID patients could be an approach to complement standard therapies.


Subject(s)
Leukemia, B-Cell/etiology , Lymphoma, B-Cell/etiology , Primary Immunodeficiency Diseases/complications , Actin Cytoskeleton/genetics , Actin Cytoskeleton/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , DNA Repair/genetics , DNA Repair/immunology , Exocytosis/genetics , Exocytosis/immunology , Genomic Instability , Humans , Immunological Synapses/genetics , Leukemia, B-Cell/genetics , Leukemia, B-Cell/immunology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Models, Immunological , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Risk Factors , Tumor Escape/genetics
6.
J Leukoc Biol ; 108(4): 1339-1360, 2020 10.
Article in English | MEDLINE | ID: mdl-32930468

ABSTRACT

In recent years, NK cells, initially identified as potent cytotoxic effector cells, have revealed an unexpected complexity, both at phenotypic and functional levels. The discovery of different NK cell subsets, characterized by distinct gene expression and phenotypes, was combined with the characterization of the diverse functions NK cells can exert, not only as circulating cells, but also as cells localized or recruited in lymphoid organs and in multiple tissues. Besides the elimination of tumor and virus-infected cells, these functions include the production of cytokines and chemokines, the regulation of innate and adaptive immune cells, the influence on tissue homeostasis. In addition, NK cells display a remarkable functional plasticity, being able to adapt to the environment and to develop a kind of memory. Nevertheless, the powerful cytotoxic activity of NK cells remains one of their most relevant properties, particularly in the antitumor response. In this review, the process of tumor cell recognition and killing mediated by NK cells, starting from the generation of cytolytic granules and recognition of target cell, to the establishment of the NK cell immunological synapse, the release of cytotoxic molecules, and consequent tumor cell death is described. Next, the review focuses on the heterogeneous mechanisms, either intrinsic to tumors or induced by the tumor microenvironment, by which cancer cells can escape the NK cell-mediated attack.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Neoplasms/immunology , Tumor Escape , Tumor Microenvironment/immunology , Animals , Humans , Killer Cells, Natural/pathology , Neoplasms/pathology
8.
Cells ; 8(5)2019 05 16.
Article in English | MEDLINE | ID: mdl-31100864

ABSTRACT

The immune system is a fundamental part of the tumor microenvironment. In particular, cytotoxic lymphocytes, such as cytolytic T cells and natural killer cells, control tumor growth and disease progression by interacting and eliminating tumor cells. The actin cytoskeleton of cytotoxic lymphocytes engaged in an immunological synapse has received considerable research attention. It has been recognized as a central mediator of the formation and maturation of the immunological synapse, and its signaling and cytolytic activities. In comparison, fewer studies have explored the organization and function of actin filaments on the target cancer cell side of the immunological synapse. However, there is growing evidence that the actin cytoskeleton of cancer cells also undergoes extensive remodeling upon cytotoxic lymphocyte attack, and that such remodeling can alter physical and functional interactions at the immunological synapse. In this article, we review the current knowledge of actin organization and functions at both sides of the immunological synapse between cytotoxic lymphocytes and cancer cells, with particular focus on synapse formation, signaling and cytolytic activity, and immune evasion.


Subject(s)
Actin Cytoskeleton/metabolism , Actin Cytoskeleton/physiology , Immunological Synapses/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Animals , Cell Line , Humans , Immune Evasion , Killer Cells, Natural/cytology , Mice , T-Lymphocytes, Cytotoxic/cytology
9.
Oncoimmunology ; 7(9): e1468954, 2018.
Article in English | MEDLINE | ID: mdl-30393584

ABSTRACT

The Wiskott-Aldrich syndrome protein (WASp) is a key regulator of the actin cytoskeleton in hematopoietic cells and mutated in two severe immunodeficiency diseases with high incidence of cancer. Wiskott-Aldrich syndrome (WAS) is caused by loss-of-function mutations in WASp and most frequently associated with lymphoreticular tumors of poor prognosis. X-linked neuropenia (XLN) is caused by gain-of-function mutations in WASp and associated with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). To understand the role of WASp in tumorigenesis, we bred WASp+, WASp-, and WASp-XLN mice onto tumor susceptible p53+/- background and sub-lethally irradiated them to enhance tumor development. We followed the cohorts for 24 weeks and tumors were characterized by histology and flow cytometry to define the tumor incidence, onset, and cell origin. We found that p53+/-WASp+ mice developed malignancies, including solid tumors and T cell lymphomas with 71.4% of survival 24 weeks after irradiation. p53+/-WASp- mice showed lower survival rate and developed various early onset malignancies. Surprisingly, the p53+/-WASp-XLN mice developed malignancy mostly with late onset, which caused delayed mortality in this colony. This study provides evidence for that loss-of-function and gain-of-function mutations in WASp influence tumor incidence and onset.

10.
J Clin Invest ; 128(9): 4115-4131, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30124469

ABSTRACT

Congenital neutropenia is characterized by low absolute neutrophil numbers in blood, leading to recurrent bacterial infections, and patients often require life-long granulocyte CSF (G-CSF) support. X-linked neutropenia (XLN) is caused by gain-of-function mutations in the actin regulator Wiskott-Aldrich syndrome protein (WASp). To understand the pathophysiology in XLN and the role of WASp in neutrophils, we here examined XLN patients and 2 XLN mouse models. XLN patients had reduced myelopoiesis and extremely low blood neutrophil number. However, their neutrophils had a hyperactive phenotype and were present in normal numbers in XLN patient saliva. Murine XLN neutrophils were hyperactivated, with increased actin dynamics and migration into tissues. We provide molecular evidence that the hyperactivity of XLN neutrophils is caused by WASp in a constitutively open conformation due to contingent phosphorylation of the critical tyrosine-293 and plasma membrane localization. This renders WASp activity less dependent on regulation by PI3K. Our data show that the amplitude of WASp activity inside a cell could be enhanced by cell-surface receptor signaling even in the context in which WASp is already in an active conformation. Moreover, these data categorize XLN as an atypical congenital neutropenia in which constitutive activation of WASp in tissue neutrophils compensates for reduced myelopoiesis.


Subject(s)
Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Neutropenia/genetics , Neutropenia/metabolism , Neutrophils/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism , Animals , Congenital Bone Marrow Failure Syndromes , Female , Gain of Function Mutation , Gene Knock-In Techniques , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neutropenia/congenital , Neutrophils/ultrastructure , Phagocytosis , Phosphorylation , Protein Conformation , Wiskott-Aldrich Syndrome Protein/chemistry
11.
Cancer Res ; 78(19): 5631-5643, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30104240

ABSTRACT

Elucidation of the underlying molecular mechanisms of immune evasion in cancer is critical for the development of immunotherapies aimed to restore and stimulate effective antitumor immunity. Here, we evaluate the role of the actin cytoskeleton in breast cancer cell resistance to cytotoxic natural killer (NK) cells. A significant fraction of breast cancer cells responded to NK-cell attack via a surprisingly rapid and massive accumulation of F-actin near the immunologic synapse, a process we termed "actin response." Live-cell imaging provided direct evidence that the actin response is associated with tumor cell resistance to NK-cell-mediated cell death. High-throughput imaging flow cytometry analyses showed that breast cancer cell lines highly resistant to NK cells were significantly enriched in actin response-competent cells as compared with susceptible cell lines. The actin response was not associated with a defect in NK-cell activation but correlated with reduced intracellular levels of the cytotoxic protease granzyme B and a lower rate of apoptosis in target cells. Inhibition of the actin response by knocking down CDC42 or N-WASP led to a significant increase in granzyme B levels in target cells and was sufficient to convert resistant breast cancer cell lines into a highly susceptible phenotype. The actin response and its protective effects were fully recapitulated using donor-derived primary NK cells as effector cells. Together, these findings establish the pivotal role of actin remodeling in breast cancer cell resistance to NK-cell-mediated killing.Significance: These findings establish the pivotal role of the actin cytoskeleton in driving breast cancer cell resistance to natural killer cells, a subset of cytotoxic lymphocytes with important roles in innate antitumor immunity. Cancer Res; 78(19); 5631-43. ©2018 AACR.


Subject(s)
Actin Cytoskeleton/metabolism , Breast Neoplasms/metabolism , Granzymes/metabolism , Killer Cells, Natural/metabolism , Apoptosis , Breast Neoplasms/pathology , Cell Line, Tumor , Cytotoxicity, Immunologic , Drug Resistance, Neoplasm , Female , Flow Cytometry , Humans , Leukocytes, Mononuclear/cytology , MCF-7 Cells , Peptide Hydrolases/metabolism , Signal Transduction , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , cdc42 GTP-Binding Protein/metabolism
12.
Sci Rep ; 8(1): 10191, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29976963

ABSTRACT

Hypoxia is a common feature of solid tumours that promotes invasion and metastatic dissemination. Invadopodia are actin-rich membrane protrusions that direct extracellular matrix proteolysis and facilitate tumour cell invasion. Here, we show that CSRP2, an invadopodial actin bundling protein, is upregulated by hypoxia in various breast cancer cell lines, as well as in pre-clinical and clinical breast tumour specimens. We functionally characterized two hypoxia responsive elements within the proximal promoter of CSRP2 gene which are targeted by hypoxia-inducible factor-1 (HIF-1) and required for promoter transactivation in response to hypoxia. Remarkably, CSRP2 knockdown significantly inhibits hypoxia-stimulated invadopodium formation, ECM degradation and invasion in MDA-MB-231 cells, while CSRP2 forced expression was sufficient to enhance the invasive capacity of HIF-1α-depleted cells under hypoxia. In MCF-7 cells, CSRP2 upregulation was required for hypoxia-induced formation of invadopodium precursors that were unable to promote ECM degradation. Collectively, our data support that CSRP2 is a novel and direct cytoskeletal target of HIF-1 which facilitates hypoxia-induced breast cancer cell invasion by promoting invadopodia formation.


Subject(s)
Breast Neoplasms/genetics , Extracellular Matrix/pathology , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , LIM Domain Proteins/genetics , Muscle Proteins/genetics , Nuclear Proteins/genetics , Adult , Aged , Animals , Breast/pathology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Female , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kaplan-Meier Estimate , LIM Domain Proteins/metabolism , Mice , Middle Aged , Muscle Proteins/metabolism , Neoplasm Invasiveness/pathology , Nuclear Proteins/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...