Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Nature ; 626(8000): 864-873, 2024 Feb.
Article En | MEDLINE | ID: mdl-38326607

Macrophage activation is controlled by a balance between activating and inhibitory receptors1-7, which protect normal tissues from excessive damage during infection8,9 but promote tumour growth and metastasis in cancer7,10. Here we report that the Kupffer cell lineage-determining factor ID3 controls this balance and selectively endows Kupffer cells with the ability to phagocytose live tumour cells and orchestrate the recruitment, proliferation and activation of natural killer and CD8 T lymphoid effector cells in the liver to restrict the growth of a variety of tumours. ID3 shifts the macrophage inhibitory/activating receptor balance to promote the phagocytic and lymphoid response, at least in part by buffering the binding of the transcription factors ELK1 and E2A at the SIRPA locus. Furthermore, loss- and gain-of-function experiments demonstrate that ID3 is sufficient to confer this potent anti-tumour activity to mouse bone-marrow-derived macrophages and human induced pluripotent stem-cell-derived macrophages. Expression of ID3 is therefore necessary and sufficient to endow macrophages with the ability to form an efficient anti-tumour niche, which could be harnessed for cell therapy in cancer.


Inhibitor of Differentiation Proteins , Kupffer Cells , Neoplasms , Animals , Humans , Mice , Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage , Induced Pluripotent Stem Cells/cytology , Inhibitor of Differentiation Proteins/deficiency , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Kupffer Cells/cytology , Kupffer Cells/immunology , Kupffer Cells/metabolism , Liver/immunology , Liver/pathology , Macrophage Activation , Neoplasm Proteins , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phagocytosis
2.
bioRxiv ; 2024 Jan 25.
Article En | MEDLINE | ID: mdl-38328106

Somatic genetic heterogeneity resulting from post-zygotic DNA mutations is widespread in human tissues and can cause diseases, however few studies have investigated its role in neurodegenerative processes such as Alzheimer's Disease (AD). Here we report the selective enrichment of microglia clones carrying pathogenic variants, that are not present in neuronal, glia/stromal cells, or blood, from patients with AD in comparison to age-matched controls. Notably, microglia-specific AD-associated variants preferentially target the MAPK pathway, including recurrent CBL ring-domain mutations. These variants activate ERK and drive a microglia transcriptional program characterized by a strong neuro-inflammatory response, both in vitro and in patients. Although the natural history of AD-associated microglial clones is difficult to establish in human, microglial expression of a MAPK pathway activating variant was previously shown to cause neurodegeneration in mice, suggesting that AD-associated neuroinflammatory microglial clones may contribute to the neurodegenerative process in patients.

3.
J Cell Sci ; 128(21): 4014-23, 2015 Nov 01.
Article En | MEDLINE | ID: mdl-26359296

We show that the combination of an intracellular bi-partite calmodulin (CaM)-binding site and a distant assembly region affect how an ion channel is regulated by a membrane lipid. Our data reveal that regulation by phosphatidylinositol(4,5)bisphosphate (PIP2) and stabilization of assembled Kv7.2 subunits by intracellular coiled-coil regions far from the membrane are coupled molecular processes. Live-cell fluorescence energy transfer measurements and direct binding studies indicate that remote coiled-coil formation creates conditions for different CaM interaction modes, each conferring different PIP2 dependency to Kv7.2 channels. Disruption of coiled-coil formation by epilepsy-causing mutation decreases apparent CaM-binding affinity and interrupts CaM influence on PIP2 sensitivity.


Calmodulin/metabolism , KCNQ2 Potassium Channel/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Calmodulin/genetics , Cell Line , Humans , KCNQ2 Potassium Channel/genetics , Mutation/genetics , Protein Binding
4.
J Cell Sci ; 128(16): 3155-63, 2015 Aug 15.
Article En | MEDLINE | ID: mdl-26148514

Calmodulin (CaM) binding to the AB module is crucial for multiple mechanisms governing the function of Kv7.2 (also known as KCNQ2) K(+) channel subunits, which mediate one of the main components of the non-inactivating K(+) M-current, a key controller of neuronal excitability. Structural analysis indicates that the CaM N-lobe engages with helix B, whereas the C-lobe anchors to the IQ site within helix A. Here, we report the identification of a new site between helices A and B that assists in CaM binding whose sequence is reminiscent of the TW helix within the CaM C-lobe anchoring site of SK2 K(+) channels (also known as KCNN2). Mutations that disrupt CaM binding within the TW site, helix B or helix A yield functional channels, whereas no function is observed when the TW site and helix A, or the TW site and helix B are mutated simultaneously. Our data indicate that the TW site is dispensable for function, contributes to the stabilization of the CaM-Kv7.2 complex and becomes essential when docking to either helix A or when helix B is perturbed.


Calcium/metabolism , Calmodulin/chemistry , KCNQ2 Potassium Channel/chemistry , Structure-Activity Relationship , Amino Acid Sequence , Binding Sites , Calcium/chemistry , Calmodulin/genetics , Calmodulin/metabolism , HEK293 Cells , Humans , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , Molecular Docking Simulation , Mutation , Neurons/metabolism , Protein Binding , Protein Structure, Secondary
5.
Biochim Biophys Acta ; 1852(9): 1856-66, 2015 Sep.
Article En | MEDLINE | ID: mdl-26073431

Mutations in the KCNQ2 gene, encoding for voltage-gated Kv7.2K(+) channel subunits, are responsible for early-onset epileptic diseases with widely-diverging phenotypic presentation, ranging from Benign Familial Neonatal Seizures (BFNS) to epileptic encephalopathy. In the present study, Kv7.2 BFNS-causing mutations (W344R, L351F, L351V, Y362C, and R553Q) have been investigated for their ability to interfere with calmodulin (CaM) binding and CaM-induced channel regulation. To this aim, semi-quantitative (Far-Western blotting) and quantitative (Surface Plasmon Resonance and dansylated CaM fluorescence) biochemical assays have been performed to investigate the interaction of CaM with wild-type or mutant Kv7.2 C-terminal fragments encompassing the CaM-binding domain; in parallel, mutation-induced changes in CaM-dependent Kv7.2 or Kv7.2/Kv7.3 current regulation were investigated by patch-clamp recordings in Chinese Hamster Ovary (CHO) cells co-expressing Kv7.2 or Kv7.2/Kv7.3 channels and CaM or CaM1234 (a CaM isoform unable to bind Ca(2+)). The results obtained suggest that each BFNS-causing mutation prompts specific biochemical and/or functional consequences; these range from slight alterations in CaM affinity which did not translate into functional changes (L351V), to a significant reduction in the affinity and functional modulation by CaM (L351F, Y362C or R553Q), to a complete functional loss without significant alteration in CaM affinity (W344R). CaM overexpression increased Kv7.2 and Kv7.2/Kv7.3 current levels, and partially (R553Q) or fully (L351F) restored normal channel function, providing a rationale pathogenetic mechanism for mutation-induced channel dysfunction in BFNS, and highlighting the potentiation of CaM-dependent Kv7.2 modulation as a potential therapeutic approach for Kv7.2-related epilepsies.

6.
J Mol Biol ; 426(15): 2717-35, 2014 Jul 29.
Article En | MEDLINE | ID: mdl-24857860

The exceptional versatility of calmodulin (CaM) three-dimensional arrangement is reflected in the growing number of structural models of CaM/protein complexes currently available in the Protein Data Bank (PDB) database, revealing a great diversity of conformations, domain organization, and structural responses to Ca(2+). Understanding CaM binding is complicated by the diversity of target proteins sequences. Data mining of the structures shows that one face of each of the eight CaM helices can contribute to binding, with little overall difference between the Ca(2+) loaded N- and C-lobes and a clear prevalence of the C-lobe low Ca(2+) conditions. The structures reveal a remarkable variety of configurations where CaM binds its targets in a preferred orientation that can be reversed and where CaM rotates upon Ca(2+) binding, suggesting a highly dynamic metastable relation between CaM and its targets. Recent advances in structure-function studies and the discovery of CaM mutations being responsible for human diseases, besides expanding the role of CaM in human pathophysiology, are opening new exciting avenues for the understanding of the how CaM decodes Ca(2+)-dependent and Ca(2+)-independent signals.


Calcium Signaling , Calmodulin/chemistry , Calmodulin/physiology , Animals , Humans , Models, Molecular
7.
PLoS One ; 9(1): e86711, 2014.
Article En | MEDLINE | ID: mdl-24489773

Kv7.2 (KCNQ2) is the principal molecular component of the slow voltage gated M-channel, which strongly influences neuronal excitability. Calmodulin (CaM) binds to two intracellular C-terminal segments of Kv7.2 channels, helices A and B, and it is required for exit from the endoplasmic reticulum. However, the molecular mechanisms by which CaM controls channel trafficking are currently unknown. Here we used two complementary approaches to explore the molecular events underlying the association between CaM and Kv7.2 and their regulation by Ca(2+). First, we performed a fluorometric assay using dansylated calmodulin (D-CaM) to characterize the interaction of its individual lobes to the Kv7.2 CaM binding site (Q2AB). Second, we explored the association of Q2AB with CaM by NMR spectroscopy, using (15)N-labeled CaM as a reporter. The combined data highlight the interdependency of the N- and C-lobes of CaM in the interaction with Q2AB, suggesting that when CaM binds Ca(2+) the binding interface pivots between the N-lobe whose interactions are dominated by helix B and the C-lobe where the predominant interaction is with helix A. In addition, Ca(2+) makes CaM binding to Q2AB more difficult and, reciprocally, the channel weakens the association of CaM with Ca(2+).


Calcium/pharmacology , Calmodulin/chemistry , Calmodulin/metabolism , KCNQ2 Potassium Channel/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Humans , Ions , KCNQ2 Potassium Channel/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Protein Structure, Secondary , Rats , Spectrometry, Fluorescence
8.
J Cell Sci ; 126(Pt 1): 244-53, 2013 Jan 01.
Article En | MEDLINE | ID: mdl-23203804

Among the multiple roles assigned to calmodulin (CaM), controlling the surface expression of Kv7.2 channels by binding to two discontinuous sites is a unique property of this Ca(2+) binding protein. Mutations that interfere with CaM binding or the sequestering of CaM prevent this M-channel component from exiting the endoplasmic reticulum (ER), which reduces M-current density in hippocampal neurons, enhancing excitability and offering a rational mechanism to explain some forms of benign familial neonatal convulsions (BFNC). Previously, we identified a mutation (S511D) that impedes CaM binding while allowing the channel to exit the ER, hinting that CaM binding may not be strictly required for Kv7.2 channel trafficking to the plasma membrane. Alternatively, this interaction with CaM might escape detection and, indeed, we now show that the S511D mutant contains functional CaM-binding sites that are not detected by classical biochemical techniques. Surface expression and function is rescued by CaM, suggesting that free CaM in HEK293 cells is limiting and reinforcing the hypothesis that CaM binding is required for ER exit. Within the CaM-binding domain formed by two sites (helix A and helix B), we show that CaM binds to helix B with higher apparent affinity than helix A, both in the presence and absence of Ca(2+), and that the two sites cooperate. Hence, CaM can bridge two binding domains, anchoring helix A of one subunit to helix B of another subunit, in this way influencing the function of Kv7.2 channels.


Calmodulin/metabolism , KCNQ2 Potassium Channel/chemistry , KCNQ2 Potassium Channel/metabolism , Binding Sites , Calmodulin/genetics , Electrophysiology , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Immunoprecipitation , KCNQ2 Potassium Channel/genetics , Mutation , Protein Binding/genetics , Protein Binding/physiology , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
9.
PLoS One ; 7(10): e47263, 2012.
Article En | MEDLINE | ID: mdl-23115641

Kv7.2 and Kv7.3 are the main components of the neuronal voltage-dependent M-current, which is a subthreshold potassium conductance that exerts an important control on neuronal excitability. Despite their predominantly intracellular distribution, these channels must reach the plasma membrane in order to control neuronal activity. Thus, we analyzed the amino acid sequence of Kv7.2 to identify intrinsic signals that may control its surface expression. Removal of the interlinker connecting helix A and helix B of the intracellular C-terminus produces a large increase in the number of functional channels at the plasma membrane. Moreover, elimination of this linker increased the steady-state amount of protein, which was not associated with a decrease of protein degradation. The magnitude of this increase was inversely correlated with the number of helix A - helix B linkers present in the tetrameric channel assemblies. In contrast to the remarkable effect on the amount of Kv7.2 protein, removal of the Kv7.2 linker had no detectable impact on the steady-state levels of Kv7.3 protein.


KCNQ2 Potassium Channel/metabolism , Amino Acid Sequence , Animals , HEK293 Cells , Humans , KCNQ2 Potassium Channel/chemistry , KCNQ2 Potassium Channel/physiology , Microscopy, Confocal , Molecular Sequence Data , Patch-Clamp Techniques , Xenopus
10.
PLoS One ; 6(9): e25508, 2011.
Article En | MEDLINE | ID: mdl-21980481

M-channels are voltage-gated potassium channels composed of Kv7.2-7.5 subunits that serve as important regulators of neuronal excitability. Calmodulin binding is required for Kv7 channel function and mutations in Kv7.2 that disrupt calmodulin binding cause Benign Familial Neonatal Convulsions (BFNC), a dominantly inherited human epilepsy. On the basis that Kv7.2 mutants deficient in calmodulin binding are not functional, calmodulin has been defined as an auxiliary subunit of Kv7 channels. However, we have identified a presumably phosphomimetic mutation S511D that permits calmodulin-independent function. Thus, our data reveal that constitutive tethering of calmodulin is not required for Kv7 channel function.


Calmodulin/metabolism , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , KCNQ2 Potassium Channel/chemistry , KCNQ2 Potassium Channel/genetics , KCNQ3 Potassium Channel/chemistry , KCNQ3 Potassium Channel/genetics , Molecular Sequence Data , Mutation , Protein Structure, Secondary , Protein Transport
...