The brain-associated LAMP-like molecule (BAD-LAMP) is a new member of the family of lysosome associated membrane proteins (LAMPs). In contrast to other LAMPs, which show a widespread expression, BAD-LAMP expression in mice is confined to the postnatal brain and therein to neuronal subpopulations in layers II/III and V of the neocortex. Onset of expression strictly parallels cortical synaptogenesis. In cortical neurons, the protein is found in defined clustered vesicles, which accumulate along neurites where it localizes with phosphorylated epitopes of neurofilament H. In primary neurons, BAD-LAMP is endocytosed, but is not found in classical lysosomal/endosomal compartments. Modification of BAD-LAMP by addition of GFP revealed a cryptic lysosomal retention motif, suggesting that the cytoplasmic tail of BAD-LAMP is actively interacting with, or modified by, molecules that promote its sorting away from lysosomes. Analysis of BAD-LAMP endocytosis in transfected HeLa cells provided evidence that the protein recycles to the plasma membrane through a dynamin/AP2-dependent mechanism. Thus, BAD-LAMP is an unconventional LAMP-like molecule and defines a new endocytic compartment in specific subtypes of cortical projection neurons. The striking correlation between the appearance of BAD-LAMP and cortical synatogenesis points towards a physiological role of this vesicular determinant for neuronal function.

Neurons are polarized cells specialized to carry out regulated secretion of storage vesicles when an appropriate stimulus is applied. Furthermore, synapse formation, stabilization and maintenance require the delivery of transport vesicles to the site of initial contact between axons and dendrites. These vesicles, containing the different proteins necessary for proper establishment and function of synapses, are the results of complex interplay between the secretion and the endocytic membrane transport pathways (Kennedy and Ehlers, 2006).

Another layer of complexity is introduced by the existence of ordered lipid domains in the plasma membrane (Maxfield and Tabas, 2005). In neurons, several types of microdomains have been shown to be distinguishable by the partitioning of different membrane-associated proteins such as thymus cell antigen 1 (THY1) or the prion protein (PrP) (Sunyach et al., 2003), which are found in different, albeit often closely adjacent domains (Madore et al., 1999). These differences in surface localization are reflected in the different trafficking and functions of these proteins. THY1 is slowly internalized and inhibits the activity of Src family kinases, whereas PrP is rapidly endocytosed and induces axonal outgrowth via the activation of fyn-related kinases (Santuccione et al., 2005). Vesicular transport and lipid microdomain organization, therefore, play key roles in neuronal development and function.

The LAMP family is composed of proteins bearing sequence and structural homology with the canonical LAMP1 and LAMP2 molecules. LAMP molecules harbor an endosomal and lysosomal addressing signal within their short cytoplasmic tail, and contain several conserved cysteine residues, which allow the formation of particular structural loops known as `LAMP folds'. Although the structure, subcellular localization and interaction partners of LAMP1 and LAMP2 have been extensively characterized, their physiological function is still elusive (Eskelinen et al., 2003). Lamp1-deficient mice are viable and show a mild astrogliosis in the brain (Andrejewski et al., 1999), whereas Lamp2 mutants show increased postnatal lethality and massive accumulation of autophagic vesicles in different tissues (Tanaka et al., 2000; Eskelinen et al., 2002). Interestingly, LAMP2 deficiency in humans induces Danon disease, a lysosomal glycogen storage disorder characterized by cardio- and skeletal myopathy and a variable degree of mental retardation (Saftig et al., 2001).

We identified a new member of the LAMP protein family in mice. Brain-associated LAMP-like molecule (BAD-LAMP) is expressed after birth in cortical neurons of particular layers, where it is enriched in defined zones along the neuronal projections. BAD-LAMP mainly accumulates in distinct intracellular vesicles, which do not contain any known markers of classical intracellular transport pathways. BAD-LAMP-containing vesicles have a remarkable clustered organization mirroring at the neuronal surface the presence of THY1-containing microdomains, but not of N-CAM and the ganglioside GM1-enriched microdomains. Interestingly the phosphoepitopes present on microtubule-associated protein 1B and neurofilament H also define BAD-LAMP-containing vesicle positioning in neurons. BAD-LAMP has the ability to be endocytosed, but is not targeted to the late endosomal/lysosome compartments (Gruenberg and Stenmark, 2004). The spatiotemporal specificity of BAD-LAMP expression and its distribution reveal therefore a new level of interplay involving unconventional endocytic compartments and membrane microdomains in specific cortical neurons.

BAD-LAMP is a new member of the LAMP family expressed specifically in the post-natal mouse brain

During a bioinformatics search to identify lysosomal-associated molecules, several overlapping nucleotide sequences were identified. After PCR cloning of the full-length cDNA from mouse cortex and extensive sequencing, we identified a potential open reading frame coding for a new putative member of the LAMP family. The new ORF codes for a protein of 280 aa (PI 6.42 and molecular mass of 31.7 kDa) predicted to contain a transmembrane domain (aa 236-256) and a cytoplasmic tail of 24 residues (Fig. 1A). This cytoplasmic domain contains a YKHM (aa 276) motif corresponding to a classical YxxΦ internalization and endosomal targeting signal. The sequence also contains four highly conserved cysteine residues separated by a fixed number of amino acids and is likely to form characteristic internal di-sulfide bonds required for a classical `LAMP fold'. The protein was also predicted to contain three consensus N-glycosylation sites. The nucleotide sequence shares 45% identity with LAMP1 and LAMP2, the founding members of the family, whereas alignments at the protein level displayed 25% similarity (19% identity) (see supplementary material Fig. S1). Thus, the protein was placed on an evolutionary classification tree between LAMP1 and DC-HIL sequences, clearly identifying it as a new member of the LAMP family (Fig. 1B). The tree indicates that DC-HIL (15.5% of similarity), a dendritic cell specific molecule functioning as an integrin ligand (Shikano et al., 2001), shared a common ancestor molecule after diverging away from the LAMP1/CD68 evolutionary axis. The molecule is extremely conserved, since it is found in worm, fly, fish, chicken, rodent and human (see supplementary material Fig. S1). The degree of identity at the amino acid level is close to 85% among mammals and 45% between mouse and fugu. This very high level of conservation across species suggests that the molecule performs a conserved cellular function, not accommodating many variations of its tertiary structure.

Northern blot analysis of the identified mRNA using different mouse tissues indicated that it is expressed almost exclusively in the adult brain, with a close to background signal in the E14 embryo (Fig. 1C). The detected mRNA corresponds to a unique transcript of around 1.8 kb with no apparent alternative spliced forms. Based on its relationship to the LAMP family and its restricted pattern of expression, the molecule was named BAD-LAMP, for brain-associated-LAMP.

Fig. 1.

Characterization of a new LAMP molecule. (A) BAD-LAMP protein sequence showing predicted glycosylation sites (bold and blue), putative di-sulfide bridges between two pairs of cysteins (red line), transmembrane domain (aa 236-256 in blue and underlined) and tyrosine endocytic sorting motif (YKHM, aa 276 in red). (B) Phylogram representation of all the LAMP family members in human and mouse. (C) Tissue distribution of BAD-LAMP by Northern blot. Among all mouse tissues tested BAD-LAMP appears to be expressed specifically in brain as a 2 kb mRNA transcript. Actin mRNA levels are shown as control.

Fig. 1.

Characterization of a new LAMP molecule. (A) BAD-LAMP protein sequence showing predicted glycosylation sites (bold and blue), putative di-sulfide bridges between two pairs of cysteins (red line), transmembrane domain (aa 236-256 in blue and underlined) and tyrosine endocytic sorting motif (YKHM, aa 276 in red). (B) Phylogram representation of all the LAMP family members in human and mouse. (C) Tissue distribution of BAD-LAMP by Northern blot. Among all mouse tissues tested BAD-LAMP appears to be expressed specifically in brain as a 2 kb mRNA transcript. Actin mRNA levels are shown as control.

Fig. 2.

BAD-LAMP is heavily glycosylated and is expressed after birth. (A) Immunoblot performed with a polyclonal antibody raised against the predicted peptide of the BAD-LAMP cytoplasmic tail. Lysates of mouse adult cortex and BAD-LAMP-transfected HeLa cells produced several bands migrating at above 35 kDa, whereas no reactivity is observed in control untransfected cells. The lowest form in the transfected cells is probably due to ER accumulation. (B) HeLa cells transfected with FLAG-BAD-LAMP cDNA were lysed and immunoprecipitated with anti-FLAG antibody. Immunoprecipitated material was treated with endoglycosidase H (endo H) or N-glycosidase F (N-gly F) prior to immunoblotting with anti-BAD-LAMP. In untransfected cells (NC), just the anti-FLAG IgG band (arrow) is observed, whereas several isoforms of BAD-LAMP were detected in transfected cells (Wt). Endo H treatment shows that the major band is endo H sensitive (gH) thus probably accumulating in the ER. The higher molecular mass bands were all N-gly F sensitive (gF). N-gly F treatment also demonstrated that all isoforms of BAD-LAMP are glycosylated and that the native molecular mass of the molecule is around 32 kDa (g0). The anti-FLAG IgG bands are also N-gly F sensitive, arrows. (C) After fractionation and isolation of cortical membranes, BAD-LAMP was found to be present exclusively in the membrane pellet (MbP) and not in the supernatant (Sup). Control syntaxin 6 and syntaxin 13 were also found in the membrane pellet, whereas RAB3, as expected, had a shared distribution due to its shuttling nature. (D) BAD-LAMP expression after birth. Mouse cortex lysates of different ages were immunoblotted with BAD-LAMP polyclonal antibody. BAD-LAMP expression levels are increased from birth to adulthood. (E) In situ hybridization for Bad-lamp on coronal post-natal brain sections from P2 to P12. Hemisections are presented. Bad-lamp expression appears at P2 in the cingulate cortex (arrowhead) and extends ventrally during the first post-natal weeks as a superficial and a deep band in the cortex. Subcortically, Bad-lamp is expressed transiently in the caudate putamen (cp). Bar, 500 μm.

Fig. 2.

BAD-LAMP is heavily glycosylated and is expressed after birth. (A) Immunoblot performed with a polyclonal antibody raised against the predicted peptide of the BAD-LAMP cytoplasmic tail. Lysates of mouse adult cortex and BAD-LAMP-transfected HeLa cells produced several bands migrating at above 35 kDa, whereas no reactivity is observed in control untransfected cells. The lowest form in the transfected cells is probably due to ER accumulation. (B) HeLa cells transfected with FLAG-BAD-LAMP cDNA were lysed and immunoprecipitated with anti-FLAG antibody. Immunoprecipitated material was treated with endoglycosidase H (endo H) or N-glycosidase F (N-gly F) prior to immunoblotting with anti-BAD-LAMP. In untransfected cells (NC), just the anti-FLAG IgG band (arrow) is observed, whereas several isoforms of BAD-LAMP were detected in transfected cells (Wt). Endo H treatment shows that the major band is endo H sensitive (gH) thus probably accumulating in the ER. The higher molecular mass bands were all N-gly F sensitive (gF). N-gly F treatment also demonstrated that all isoforms of BAD-LAMP are glycosylated and that the native molecular mass of the molecule is around 32 kDa (g0). The anti-FLAG IgG bands are also N-gly F sensitive, arrows. (C) After fractionation and isolation of cortical membranes, BAD-LAMP was found to be present exclusively in the membrane pellet (MbP) and not in the supernatant (Sup). Control syntaxin 6 and syntaxin 13 were also found in the membrane pellet, whereas RAB3, as expected, had a shared distribution due to its shuttling nature. (D) BAD-LAMP expression after birth. Mouse cortex lysates of different ages were immunoblotted with BAD-LAMP polyclonal antibody. BAD-LAMP expression levels are increased from birth to adulthood. (E) In situ hybridization for Bad-lamp on coronal post-natal brain sections from P2 to P12. Hemisections are presented. Bad-lamp expression appears at P2 in the cingulate cortex (arrowhead) and extends ventrally during the first post-natal weeks as a superficial and a deep band in the cortex. Subcortically, Bad-lamp is expressed transiently in the caudate putamen (cp). Bar, 500 μm.

BAD-LAMP is a glycosylated membrane-associated protein

To investigate further BAD-LAMP distribution and function, we raised antibodies against peptide epitopes present in its cytoplasmic tail. These antibodies were characterized by immunoblot of HeLa cells transfected with the cDNA coding for mouse BAD-LAMP (Fig. 2A). Several bands were detected in extracts from transfected cells, whereas control extracts remained non-reactive. Brain extracts also displayed several bands, mostly corresponding to those observed in transfected cells, confirming the existence of several isoforms of BAD-LAMP. The detected proteins had a significantly higher molecular mass than the one predicted from the primary sequence of BAD-LAMP (31.7 kDa). In order to define the nature of these post-translational modifications and in absence of immunoprecipitating antibodies, we transfected an N-terminally tagged form of BAD-LAMP (FLAG-BAD-LAMP), allowing efficient immunoprecipitation and treatment with endoglycosidase H (Endo H) and N-glycosidase F (N-gly F). Immunoprecipitated FLAG-BAD-LAMP was shown to be heavily glycosylated (Fig. 2B). The major form of the protein (38 kDa, gH) remained Endo H sensitive, thus reflecting endoplasmic reticulum (ER) retention due to over-expression. The two higher additional bands (47 and 53 kDa, gF) were Endo H resistant but remained N-gly F sensitive, as indicated by the accumulation of a fully trimmed 31 kDa protein (g0) after treatment. Transfected BAD-LAMP is therefore heavily glycosylated on at least two of its three acceptor sites, a situation likely to be shared with endogenous BAD-LAMP detected in brain.

Fig. 3.

BAD-LAMP is specifically expressed in neurons of the cortical layers II, III and V. (A) In situ hybridization for Bad-lamp (top panel), Cux2 (middle) and ER81 (bottom) on adult mouse brain coronal hemisections. (B) High magnification views of in situ hybridization on wild-type cortex shown in A. Bad-lamp is expressed in layers II, II and V, but excluded from layers IV and VI. In the Scrambler cortex, the entire region appears disorganized. However, the typical inversion of cortical layers is reflected by the altered BAD-LAMP staining, demonstrating that projection neurons express the protein. (C) Combined in situ hybridization for Bad-lamp (in blue) with immunohistochemistry for the specific neuronal marker NeuN (in brown). The left panel is a higher magnification of the boxed area. Bad-lamp is co-expressed with this pan-neuronal marker in many, although not all, neurons. (D) Immunohistochemistry of BAD-LAMP in the indicated cortex layers. (E) Immunofluorescence staining on adult cortex using anti-MAP2 (green) and anti-BAD-LAMP (red) antibodies; the merged image is on the left. Whereas MAP2 is present along the entire dendrites, BAD-LAMP accumulates in defined domains (arrows). Bars, 500 μm in A; 200 μm in B; 100 μm (left panel) and 10 μm (right panel) in C; 10 μm in D.

Fig. 3.

BAD-LAMP is specifically expressed in neurons of the cortical layers II, III and V. (A) In situ hybridization for Bad-lamp (top panel), Cux2 (middle) and ER81 (bottom) on adult mouse brain coronal hemisections. (B) High magnification views of in situ hybridization on wild-type cortex shown in A. Bad-lamp is expressed in layers II, II and V, but excluded from layers IV and VI. In the Scrambler cortex, the entire region appears disorganized. However, the typical inversion of cortical layers is reflected by the altered BAD-LAMP staining, demonstrating that projection neurons express the protein. (C) Combined in situ hybridization for Bad-lamp (in blue) with immunohistochemistry for the specific neuronal marker NeuN (in brown). The left panel is a higher magnification of the boxed area. Bad-lamp is co-expressed with this pan-neuronal marker in many, although not all, neurons. (D) Immunohistochemistry of BAD-LAMP in the indicated cortex layers. (E) Immunofluorescence staining on adult cortex using anti-MAP2 (green) and anti-BAD-LAMP (red) antibodies; the merged image is on the left. Whereas MAP2 is present along the entire dendrites, BAD-LAMP accumulates in defined domains (arrows). Bars, 500 μm in A; 200 μm in B; 100 μm (left panel) and 10 μm (right panel) in C; 10 μm in D.

Although glycosylation was in support of BAD-LAMP membrane association, we demonstrated the membrane-bound nature of BAD-LAMP by submitting mouse cortex post-nuclear supernatants to high speed ultracentrifugation, in which BAD-LAMP was found associated with the membranes pellets similar to other membrane-associated molecules such as RAB3a, syntaxin 6 and syntaxin 13 (Fig. 2C). Thus BAD-LAMP, is a glycosylated LAMP-like molecule associated with cortical membranes.

BAD-LAMP is expressed in neurons of specific cortical layers after birth

Analysis of mouse brain extracts by immunoblotting revealed that levels of BAD-LAMP increased strongly after birth (P0) reaching its maximum level at adulthood, but being already strongly expressed at P10-P12 (Fig. 2D). We used in situ hybridization to investigate in detail the expression pattern of BAD-LAMP in the developing mouse forebrain. The first expression of BAD-LAMP was found at P2 in the cingulate cortex, in a thin band of intermediate cells (Fig. 2E). At P5, expression extended ventrally into the cortical plate. Furthermore, the caudate putamen showed a punctuate expression of Bad-lamp transcripts. This expression pattern was maintained at P7, when an additional broad band of large and strongly Bad-lamp-positive cells appeared in superficial parts of the cortical plate. Although the cortical expression intensified until P9, no major regional changes in Bad-lamp expression were obvious during this period. At P12, expression of Bad-lamp in the striatum ceased, while expression further intensified in the cortex. This expression pattern was stable until adulthood. Altogether, this expression pattern indicates that BAD-LAMP does not function in the early steps of brain development, such as neurogenesis and cell migration, but potentially during terminal steps of neuronal differentiation and neuronal function.

Within the adult cortex, the homogenous staining in outer regions of the cortical plate, as well as in a more restricted band of cells localized centrally, was suggestive of an expression in neurons of specific cortical layers. We used well-known markers for cortical layers to further characterize the respective populations. Comparison of the expression of Bad-lamp to that of Cux2, a marker for layers II-IV, showed that the BAD-LAMP domain is included in the CUX2 domain and confined to its outer part (Fig. 3A,B). Thus, BAD-LAMP is expressed in the upper layers II and III of the neocortex, but is excluded from layer IV. Furthermore, there was a perfect overlap with the layer V marker ER81 (Fig. 3A,B) demonstrating that the deeply positioned Bad-lamp-positive population is located in layer V.

The size of the Bad-lamp-positive cells in the respective cortical layers was suggestive of neuronal cells. To confirm this observation we investigated the expression of Bad-lamp in Scrambler mice. These animals show a well described inversion of the layers of cortical projection neurons, with upper layer neurons (layers II-IV) positioned deeply whereas deep layer neurons (layers V and VI) are positioned superficially (Rice and Curran, 1999). The organization of B Bad-lamp-positive cells in the Scrambler cortex was strikingly altered (Fig. 3B). Small, lighter stained cell bodies were displaced towards the ventricular side, whereas larger and more strongly labelled cells were merely found at the pial side of the cortex. This pattern was in agreement with an inversion of the position of Bad-lamp-positive cells and suggestive of a projection neurone identity. Furthermore, all Bad-lamp-positive cells in the cortex were co-expressing the neuronal marker NeuN, again confirming the neuronal identity of labelled cells (Fig. 3C), which were found also to express BAD-LAMP protein (Fig. 3D).

Fig. 4.

BAD-LAMP is present in vesicles clustering in specific areas of the neurons. Immunofluorescence confocal microscopy of cortical neurons. (A) Staining for BAD-LAMP (red) and cholera toxin (GM1, green). (B) Staining for BAD-LAMP (red) and PrP (green). (C) Staining for BAD-LAMP (red) and N-CAM (green). BAD-LAMP is expressed in small vesicles clustered in neurites and accumulates in areas lacking surface semi-ordered lipid microdomain resident proteins (arrowheads). (D) THY1 labelling (green) defines the zones in which BAD-LAMP vesicles accumulate (red). However, THY1 (red) is present at the cell surface and does not co-localize with BAD-LAMP as seen at higher magnification (bottom panels, arrowheads). (E) Cholesterol depletion disrupts cluster organization and induces BAD-LAMP (red) and cholera toxin (GM1, green) co-localization. Bars, 20 μm; 10 μm for THY1 high magnification.

Fig. 4.

BAD-LAMP is present in vesicles clustering in specific areas of the neurons. Immunofluorescence confocal microscopy of cortical neurons. (A) Staining for BAD-LAMP (red) and cholera toxin (GM1, green). (B) Staining for BAD-LAMP (red) and PrP (green). (C) Staining for BAD-LAMP (red) and N-CAM (green). BAD-LAMP is expressed in small vesicles clustered in neurites and accumulates in areas lacking surface semi-ordered lipid microdomain resident proteins (arrowheads). (D) THY1 labelling (green) defines the zones in which BAD-LAMP vesicles accumulate (red). However, THY1 (red) is present at the cell surface and does not co-localize with BAD-LAMP as seen at higher magnification (bottom panels, arrowheads). (E) Cholesterol depletion disrupts cluster organization and induces BAD-LAMP (red) and cholera toxin (GM1, green) co-localization. Bars, 20 μm; 10 μm for THY1 high magnification.

BAD-LAMP is distributed in specific domains of cortical neurons

BAD-LAMP distribution in cortical brain sections was monitored by confocal microscopy (Fig. 3E). BAD-LAMP was found in vesicles mostly located in cell bodies as delineated by the MAP2 staining. In addition, BAD-LAMP accumulated in defined domains along cellular projections (Fig. 3E arrowheads). It could be detected at the plasma membrane and in vesicles present in the cell bodies, but was also enriched in vesicles clustered in defined domains of dendrites. BAD-LAMP mostly accumulated within the boundaries of specific neuronal areas in vivo.

In order to confirm the relevance of these observations, embryonic cortical neurons were explanted and BAD-LAMP sub-cellular distribution was investigated after 3 days in culture. Owing to the particular clustered distribution of BAD-LAMP, we also investigated the distribution of proteins known to partition in different cellular domains, such as lipid microdomain-associated proteins (Madore et al., 1999). Using confocal microscopy we found that semi-ordered lipid microdomain residents such as PrP, N-CAM, as well as the ganglioside GM1 (stained with cholera toxin, CT) were enriched in zones excluding BAD-LAMP vesicles (Fig. 4A-C). This observation was particularly striking with CT staining and N-CAM, which accumulated almost exclusively in areas negative for BAD-LAMP (Fig. 4A,C). By contrast, at this low magnification, THY1, a molecule representing ordered lipid microdomain-associated proteins, displayed an overlapping distribution with BAD-LAMP (Fig. 4D). However, at higher magnification, no direct co-localization of THY1 and BAD-LAMP molecules could be observed. Instead, accumulation of BAD-LAMP-containing vesicles was revealed directly underneath THY1-enriched areas at the plasma membrane (Fig. 4D, arrowheads). BAD-LAMP-containing vesicles therefore accumulate in cellular zones, defined by the presence of THY1 at the plasma membrane, whereas they are segregated from the detergent-resistant microdomains containing most of the PrP, GM1 and N-CAM (Madore et al., 1999).

Fig. 5.

BAD-LAMP organization is defined by microtubules. Immunofluorescence confocal microscopy of cortical neurons. (A) BAD-LAMP vesicles (red) accumulate in areas of the cell that are strongly enriched in the phospho-epitope detected by the Smi31 antibody (blue), whereas the L1 molecule (green) is distributed throughout the neuronal plasma membrane. (B) (top) BAD-LAMP vesicle accumulation (white) coincides with microtubule bundling (β-tubulin, red) and weak GM1 staining (green). Higher magnification (Z1) shows that BAD-LAMP vesicles align along microtubules. (Bottom) Nocodazole treatment induces microtubule destabilization and disorganization of BAD-LAMP vesicle clusters. Bars, 20 μm; 10 μm for Z1.

Fig. 5.

BAD-LAMP organization is defined by microtubules. Immunofluorescence confocal microscopy of cortical neurons. (A) BAD-LAMP vesicles (red) accumulate in areas of the cell that are strongly enriched in the phospho-epitope detected by the Smi31 antibody (blue), whereas the L1 molecule (green) is distributed throughout the neuronal plasma membrane. (B) (top) BAD-LAMP vesicle accumulation (white) coincides with microtubule bundling (β-tubulin, red) and weak GM1 staining (green). Higher magnification (Z1) shows that BAD-LAMP vesicles align along microtubules. (Bottom) Nocodazole treatment induces microtubule destabilization and disorganization of BAD-LAMP vesicle clusters. Bars, 20 μm; 10 μm for Z1.

BAD-LAMP distribution and microdomain organization seemed to be closely linked. Cholesterol depletion efficiently affects lipid microdomains and was therefore tested for its ability to influence BAD-LAMP distribution. Cortical neurons were treated for 2 hours with cholesterol-esterase prior to immunostaining and confocal microscopy visualization (Fig. 4E). As expected, cholesterol depletion had a potent effect on GM1 distribution at the plasma membrane. Moreover, BAD-LAMP vesicular staining was also deeply affected, displaying an extensive co-localization with GM1, which was never observed in normal conditions. Thus, microdomain organization at the membrane and clustering of BAD-LAMP-positive vesicles appeared to be directly linked.

The cytoskeleton controls the distribution of BAD-LAMP vesicles

This particular organization was likely to be maintained with the active participation of the cytoskeleton and/or associated proteins. In order to test this hypothesis, several candidate molecules were followed by confocal microscopy in cortical neurons. Surprisingly, BAD-LAMP containing-vesicles clustered within punctate zones delimited by staining with the Smi31 antibody (Fig. 5A). Smi31 detects phosphorylated epitopes present in neurofilament H and mostly in the microtubule-associated molecule 1B (MAP1B) (Fischer and Romano-Clarke, 1990). Although the precise function of MAP1B phosphorylation is still debated, experimental evidence suggests a role in regulating microtubules and actin dynamics as well as being necessary for axonal growth (Dehmelt and Halpain, 2004; Del Rio et al., 2004). The perfect overlapping distribution of BAD-LAMP and Smi31 strongly suggested that microtubules or actin are likely to play in important role in the organization and the clustering of BAD-LAMP-positive vesicles, however BAD-LAMP distribution is not dependent on the neuronal polarity.

BAD-LAMP-positive vesicles were found in the close vicinity of the microtubule network, mirroring, by their accumulation, the intensity of the tubules bundling (Fig. 5B). A treatment with the microtubule depolymerizing agent nocodazole was thus carried out (Fig. 5B). Nocodazole induced a strong redistribution of BAD-LAMP-containing vesicles and a loss of BAD-LAMP staining intensity in cortical neurons. Thus the microtubule network influences the positioning of BAD-LAMP vesicles. Lipid microdomain organization and BAD-LAMP distribution in cortical neurons are therefore linked, use the microtubule network and possibly depend on MAP1B phosphorylation for their regulation.

BAD-LAMP defines a specific subset of early endosomes

The unusual distribution of BAD-LAMP vesicles led us to investigate their relationship with other types of sub-cellular compartments. We focused primarily on endocytic organelles, likely to be relevant to a transmembrane molecule bearing a YxxΦ motif in its cytoplasmic tail. BAD-LAMP could not be detected in classical endosomal compartments as judged from its lack of co-localization with LAMP2 (late endosomes and lysosomes) and internalized transferrin-FITC (sorting and recycling endosomes) (Fig. 6A,B) or syntaxin 13 (Hirling et al., 2000). BAD-LAMP was not found in more specialized endocytic compartment such as TI-VAMP-positive vesicles (Coco et al., 1999) (Fig. 6C). Co-labeling with synaptic vesicle proteins such as synaptotagmin 1, RAB3a, VAMP2 revealed some level of co-localization with BAD-LAMP in the growth cones (Fig. 6D-F). Interestingly, co-localization was not observed in other cellular areas and a similar overlapping distribution in the growth cone was observed with TI-VAMP, which is not found enriched in synaptic vesicles (Coco et al., 1999). Thus, this co-distribution in the growth cone probably reflects the difficulty of segregating, at this optical resolution, individual carrier vesicles congregating in the same area of the cone, rather than a true co-localization in the same vesicles. Pre-and post-synaptic transport carriers are derived from trans-Golgi network (TGN) vesicles, which aggregate at initial contacts between axons and dendrites (Sytnyk et al., 2002). We, therefore, examined the possible association of BAD-LAMP with other known vesicular markers of these pathways, such as syntaxin 6 or N-CAM (Sytnyk et al., 2004) (Fig. 6E). We failed to detect co-localization of BAD-LAMP with any of these markers (see supplementary material Fig. S2), suggesting that the molecule is sorted in an uncharacterized type of vesicles, which can accumulate in the growth cone of developing axon, as well as in defined and organized domains along the cellular processes.

BAD-LAMP distribution at the plasma membrane as well as in localized intracellular vesicles suggested a possible shuttling of the molecule between the cell surface and the vesicles. The co-localization of GM1 and BAD-LAMP upon cholesterol depletion suggests that BAD-LAMP vesicles are accessible to plasma membrane constituents under specific conditions. To address this issue, cortical neurons were surface biotinylated at 4°C prior to incubation at 37°C. Biotinylated surface proteins could either diffuse, or be internalized, and their intermixing with BAD-LAMP-positive compartments was evaluated at different time points by confocal microscopy (Fig. 7). Biotinylated proteins were detected rapidly co-localizing with BAD-LAMP after 5 minutes of internalization. This significant overlapping distribution decreased after 45 minutes, suggesting that BAD-LAMP-containing organelles could represent a subset of early endocytic vesicles, rapidly accessible from the neuronal surface and serving as an intermediate step for the intracellular sorting of specific surface molecules present in developing neurons.

Fig. 6.

Confocal immunofluorescence microscopy analysis of BAD-LAMP transport in cortical neurons. (A) Staining for LAMP2 (green) and BAD-LAMP (red). (B) Internalized transferrin-FITC (green) in early and recycling endosomes and BAD-LAMP (red). (C) Staining for Ti-VAMP (green) and BAD-LAMP (red). (D) Staining of a growth cone for synaptotagmin 1 (SYT1, green) and BAD-LAMP (red). (E) Staining of a growth cone for VAMP2 (green) and BAD-LAMP (red). (F) Staining for syntaxin 6 (green) and BAD-LAMP (red). BAD-LAMP does not display any significant co-localization with LAMP1 and internalized transferrin. Bars, 20 μm in A,B,C,F, 10 μm in D,E.

Fig. 6.

Confocal immunofluorescence microscopy analysis of BAD-LAMP transport in cortical neurons. (A) Staining for LAMP2 (green) and BAD-LAMP (red). (B) Internalized transferrin-FITC (green) in early and recycling endosomes and BAD-LAMP (red). (C) Staining for Ti-VAMP (green) and BAD-LAMP (red). (D) Staining of a growth cone for synaptotagmin 1 (SYT1, green) and BAD-LAMP (red). (E) Staining of a growth cone for VAMP2 (green) and BAD-LAMP (red). (F) Staining for syntaxin 6 (green) and BAD-LAMP (red). BAD-LAMP does not display any significant co-localization with LAMP1 and internalized transferrin. Bars, 20 μm in A,B,C,F, 10 μm in D,E.

BAD-LAMP sorting in transfected neurons

To further investigate the distribution of BAD-LAMP, we generated N-terminally FLAG-tagged and C-terminally GFP-tagged BAD-LAMP constructs and monitored their behavior by microscopy in co-transfection experiments of cortical neurons (Fig. 8). Surprisingly, endogenous BAD-LAMP expression and domain organization were strongly inhibited in electroporated neurons. Nevertheless, transfected FLAG-tagged BAD-LAMP was found enriched in vesicles clustered in specific zones along the neurites. Clearly, the tagged protein is not addressed in conventional endo/lysosomes as judged by its lack of co-localization with LAMP2 (Fig. 8A), internalized transferrin (not shown) or cholera toxin (see supplementary material Fig. S3). The exact location of FLAG-BAD-LAMP in the cell body was difficult to establish since its over-expression induced an accumulation of the molecule in the ER and Golgi network. Surprisingly, the C-terminally GFP-tagged, BAD-LAMP (BAD-GFP) was found accumulating in LAMP2-positive lysosomal compartments (Fig. 8A, arrowheads in Z1). Therefore, the BAD-LAMP cytoplasmic tail contains a cryptic lysosomal retention motif, which is revealed by the addition of the GFP moiety. This observation also suggests that the cytoplasmic tail of BAD-LAMP is actively interacting with, or modified by, molecules that promote its sorting away from traditional endocytic compartments. Co-localization of BAD-GFP and FLAG-tagged BAD-LAMP was observed in discrete vesicles in neurites (Fig. 8A, Z2 arrowheads), despite the fact that BAD-GFP was found mostly accumulating in large lysosomes in the cell body. This demonstrates that a small fraction of BAD-GFP can be sorted normally.

Fig. 7.

Surface biotinylation reveals the endocytic nature of BAD-LAMP-containing vesicles. Cortical neurons were surface biotinylated for 15 minutes at 4°C, prior to warming at 37°C for different times, fixation and visualization by confocal microscopy. (A) Prior to warming, no significant co-localization of biotinylated proteins with BAD-LAMP was observed. Co-localization was evaluated using the Image J image analysis software. A low Pearson's coefficient and strong negative pixel shift are both indicative of the absence of staining overlap (right). (B) After 5 minutes of endocytosis at 37°C, extensive co-localization of biotinylated proteins (green) was observed with BAD-LAMP (red) in neurites (arrowheads), as also shown by a higher Pearson's coefficient and the absence of pixel shift (right). (C) After 45 minutes of endocytosis co-localization of BAD-LAMP with biotin is decreased as shown by a decreased Pearson's coefficient and negative pixel shift (right). Bar, 10 μm.

Fig. 7.

Surface biotinylation reveals the endocytic nature of BAD-LAMP-containing vesicles. Cortical neurons were surface biotinylated for 15 minutes at 4°C, prior to warming at 37°C for different times, fixation and visualization by confocal microscopy. (A) Prior to warming, no significant co-localization of biotinylated proteins with BAD-LAMP was observed. Co-localization was evaluated using the Image J image analysis software. A low Pearson's coefficient and strong negative pixel shift are both indicative of the absence of staining overlap (right). (B) After 5 minutes of endocytosis at 37°C, extensive co-localization of biotinylated proteins (green) was observed with BAD-LAMP (red) in neurites (arrowheads), as also shown by a higher Pearson's coefficient and the absence of pixel shift (right). (C) After 45 minutes of endocytosis co-localization of BAD-LAMP with biotin is decreased as shown by a decreased Pearson's coefficient and negative pixel shift (right). Bar, 10 μm.

We next evaluated the internalization dynamics of BAD-LAMP by using the N-terminally FLAG-tagged construct and by monitoring FLAG antibody uptake after cold binding (Fig. 8B). The antibody was rapidly endocytosed after 5 minutes at 37°C. Inside the cell, it was detected in a different compartment from conventional endo/lysosomes as shown by the absence of co-localization with co-transfected BAD-GFP, LAMP2 (Z3 and arrows) and internalized cholera toxin (supplementary material Fig. S3A). After 30 minutes of synchronous uptake (Z4 and arrowheads), co-localization of the antibodies with BAD-LAMP-GFP and LAMP 2 indicated that BAD-LAMP can reach conventional endocytic compartments, after being internalized from the surface. Surprisingly, this co-localization was more evident in the more discrete LAMP2-positive organelles present in the neurite (late endosomes, arrowheads) than in the large lysosomes observed in the cell body (Fig. 8B).

We next investigated the contribution of tyrosine 276 to BAD-LAMP trafficking by introducing a mutational change to alanine at this position (Tyr276Ala). The FLAG-tagged mutant was also found accumulating in the ER and Golgi network of transfected neurons. However, the fraction of the mutant that exited these organelles accumulated at the surface of the neurites in a manner very distinct from the normal molecule (wild type), which was mostly found in intracellular vesicles (supplementary material Fig. S3B). Similar results were obtained with a construct lacking the entire cytoplasmic tail of BAD-LAMP (not shown). Thus, tyrosine 276 is directly involved in intracellular addressing of BAD-LAMP and allows its internalization from the surface. FLAG antibody uptake after binding in the cold was performed in neurons expressing FLAG-BAD-LAMP Tyr276Ala. Transfected cells remained mostly antibody-decorated at the surface 30 minutes after warming at 37°C (supplementary material Fig. S3C). Thus, BAD-LAMP is probably cycling between the plasma membrane and a subset of endocytic vesicles.

Fig. 8.

Localization of FLAG-tagged BAD-LAMP in transfected cortical neurons. Cortical neurons co-transfected with BAD-GFP and FLAG-BAD-LAMP were visualized by confocal microscopy. (A) Staining for FLAG antibody (red), BAD-GFP (green) and LAMP2 (white). FLAG-BAD-LAMP does not co-localize with LAMP 2. BAD-GFP is targeted to lysosomes upon addition of the GFP moiety at the C-terminal end of BAD-LAMP. Bar, 20 μm; 10 μm for high magnification of Z1 and 5 μm for Z2. (B) Internalization of FLAG antibody (red) in transfected neurons for indicated times and staining for LAMP2 (white). High magnifications reveal a late accessibility of BAD-LAMP into LAMP2 positive compartments in neurites. Bars, 20 μm; 5 μm for high magnification.

Fig. 8.

Localization of FLAG-tagged BAD-LAMP in transfected cortical neurons. Cortical neurons co-transfected with BAD-GFP and FLAG-BAD-LAMP were visualized by confocal microscopy. (A) Staining for FLAG antibody (red), BAD-GFP (green) and LAMP2 (white). FLAG-BAD-LAMP does not co-localize with LAMP 2. BAD-GFP is targeted to lysosomes upon addition of the GFP moiety at the C-terminal end of BAD-LAMP. Bar, 20 μm; 10 μm for high magnification of Z1 and 5 μm for Z2. (B) Internalization of FLAG antibody (red) in transfected neurons for indicated times and staining for LAMP2 (white). High magnifications reveal a late accessibility of BAD-LAMP into LAMP2 positive compartments in neurites. Bars, 20 μm; 5 μm for high magnification.

BAD-LAMP recycles in HeLa cells

In order to further dissect the molecular mechanisms governing BAD-LAMP endocytosis, we studied the distribution and transport of transfected BAD-LAMP in a cell type easy to manipulate, such as HeLa and mouse NIH 3T3 cells. In HeLa cells, FLAG-BAD-LAMP was found at the cell surface and in internalized transferrin-containing vesicles distributed in the vicinity of the plasma membrane, whereas the Tyr276Ala mutant accumulated only at the cell surface (Fig. 9A). No co-localization was found in LAMP1-positive late endosomes or lysosomes, nor with co-transfected DC-LAMP tagged with GFP (Fig. 9B), another lysosomal resident of the LAMP family (de Saint-Vis et al., 1998). These observations were confirmed after Percoll density gradient subcellular fractionation of transfected HeLa cells (supplementary material Fig. S4). BAD-LAMP was mostly detected in the low density fractions of the gradient containing plasma membrane, ER and early endosomes, but it was absent from the high density fractions containing lysosomes, as indicated by β-hexosaminidase activity. Thus, most of transfected BAD-LAMP was found on the cell surface contrasting with transfected neurons in which BAD-LAMP mostly accumulated intracellularly, underlining the specificity of its sorting even when over-expressed.

Anti-FLAG antibody uptake in transfected cells saturated with FITC-transferrin (FITC-TF), confirmed that BAD-LAMP could be internalized rapidly in sorting endosomes (Fig. 9C). Interestingly, 15 minutes after uptake BAD-LAMP was found present in transferrin-positive recycling endosomes clustered around the microtubule organizing center, suggesting that BAD-LAMP could recycle to the plasma membrane after internalization. This hypothesis was supported by the poor co-localization of the antibody with LAMP1 after 45 minutes of uptake, indicating that the molecule does not efficiently reach late endocytic compartments. This underlines again a difference with neurons, in which the antibodies could be detected in discrete LAMP1-positive compartment 45 minutes after uptake.

We next investigated the molecular mechanisms involved in BAD-LAMP endocytosis. Experiments performed in cells co-transfected with wild-type GTPase dynamin II or dominant-negative mutant A44K, indicated that BAD-LAMP internalization is mediated in a dynamin-dependent manner, since antibody internalization was abolished in cells expressing dynamin A44K (Fig. 9D and control, supplementary material Fig. S4C). In order to further define the endocytic pathway used by BAD-LAMP to enter the cell, we used an RNA inhibition approach to reduce the expression of molecules involved in protein triage from the surface, such as the clathrin adaptor AP2 (Dugast et al., 2005; McCormick et al., 2005). Antibody uptake was monitored by immunostaining and FACS detection after binding at 4°C and internalization at 37°C. Cells co-transfected with FLAG-BAD-LAMP and control RNAi plasmid showed rapid internalization of the antibody (Fig. 9E), whereas RNAi depletion of AP2 clearly inhibited BAD-LAMP internalization as well as transferin uptake (supplementary material Fig. S4C). In cells depleted for AP2, higher surface levels of BAD-LAMP were also consistently detected (not shown), suggesting that BAD-LAMP is internalized constantly through a dynamin/AP2-dependent endocytic pathway.

Fig. 9.

BAD-LAMP is targeted to early endosomes and recycles in HeLa cells. HeLa cells transfected with FLAG-BAD-LAMP were submitted to immunofluorescent staining and confocal microscopy visualization. (A) FLAG-tagged BAD-LAMP (anti-flag antibody, red) was found at the cell surface and in internalized transferrin-FITC-containing endosomes. Cytoplasmic tail tyrosine 276 mutant (Tyr276-Ala) was found accumulating at the surface of transfected cells (anti-flag antibody, red) with little intracellular distribution (transferrin-FITC, green). (B) Transfected FLAG-BAD-LAMP is not detected in LAMP1- (blue) and DC-LAMP (green)-positive late endosomes and lysososomes. (C) Kinetics of FLAG antibody uptake after cold binding on the surface of transfected HeLa cells. Only transfected cells accumulate the antibody (red) on their surface, which upon warming reaches rapidly sorting (5 minutes) and recycling (15 minutes) endosomes containing transferrin-FITC (green). No co-localization with LAMP1 (white, 45 minutes) could be observed, suggesting that BAD-LAMP and associated antibodies do not access the late endocytic pathway. (D) Co-expression of dynamin dominant negative mutant A44K (right panel, green) in FLAG-BAD-LAMP-transfected HeLa cells prevents the internalization of associated flag antibodies (right panel, red), whereas expression of wild-type dynamin has no effect (green, left panel). (E) Co-transfection of HeLa cells with FLAG-BAD-LAMP (anti-BAD-LAMP, red) and pSuper control plasmid (left) has no effect on the internalization of associated flag antibodies (green). Conversely RNAi inhibition of the clathrin adaptor AP2 blocks flag antibodies uptake (green). Bars, 20 μm.

Fig. 9.

BAD-LAMP is targeted to early endosomes and recycles in HeLa cells. HeLa cells transfected with FLAG-BAD-LAMP were submitted to immunofluorescent staining and confocal microscopy visualization. (A) FLAG-tagged BAD-LAMP (anti-flag antibody, red) was found at the cell surface and in internalized transferrin-FITC-containing endosomes. Cytoplasmic tail tyrosine 276 mutant (Tyr276-Ala) was found accumulating at the surface of transfected cells (anti-flag antibody, red) with little intracellular distribution (transferrin-FITC, green). (B) Transfected FLAG-BAD-LAMP is not detected in LAMP1- (blue) and DC-LAMP (green)-positive late endosomes and lysososomes. (C) Kinetics of FLAG antibody uptake after cold binding on the surface of transfected HeLa cells. Only transfected cells accumulate the antibody (red) on their surface, which upon warming reaches rapidly sorting (5 minutes) and recycling (15 minutes) endosomes containing transferrin-FITC (green). No co-localization with LAMP1 (white, 45 minutes) could be observed, suggesting that BAD-LAMP and associated antibodies do not access the late endocytic pathway. (D) Co-expression of dynamin dominant negative mutant A44K (right panel, green) in FLAG-BAD-LAMP-transfected HeLa cells prevents the internalization of associated flag antibodies (right panel, red), whereas expression of wild-type dynamin has no effect (green, left panel). (E) Co-transfection of HeLa cells with FLAG-BAD-LAMP (anti-BAD-LAMP, red) and pSuper control plasmid (left) has no effect on the internalization of associated flag antibodies (green). Conversely RNAi inhibition of the clathrin adaptor AP2 blocks flag antibodies uptake (green). Bars, 20 μm.

Interestingly, monitoring of surface anti-FLAG antibody by FACS also indicated that the molecule was rapidly internalized between 5 and 7.5 minutes after warming (supplementary material Fig. S4B). Surface levels of antibodies then re-increased after 10 minutes, to be diminished again but with a relatively slower internalization rate. These observations confirm that BAD-LAMP and associated antibodies constantly recycle to the plasma membrane with a relatively high efficiency.

BAD-LAMP sequence analysis clearly indicates that it represents a new member of the LAMP family. However, its expression pattern and intracellular distribution are unconventional compared to other LAMP family members, which show a widespread expression and specifically accumulate in the lysosomes.

Our observations on BAD-LAMP intracellular distribution are clearly indicative of a strong regulation of its trafficking in a subset of early endosomes. Although we have not been able to identify molecular markers able to identify these organelles, the absence of transferrin or synaptotagmin 1, as well as late endosomal markers such as LAMP1 suggests that these vesicles represent a distinct class of neuronal endosomes. The kinetics of biotinylated proteins and antibody uptake indicate that they can serve as sorting platforms, prior to transport to other organelles, which are positive for LAMP1, but only represent a minor fraction of the neuronal organelles containing LAMP1.

We have shown that BAD-LAMP, through an interaction with its YKHM domain, requires dynamin and AP2 to be internalized and sorted towards the early endocytic recycling pathway of transfected HeLa cells. LAMP1 has also been shown to require the AP2 adaptor, but its sorting is directed towards lysosomes (Janvier and Bonifacino, 2005). Interestingly, modification of the BAD-LAMP C-terminal domain by GFP deeply affects its transport in neurons and demonstrates the existence of an active sorting pathway in these cells, which normally prevents the accumulation of BAD-LAMP in the lysosomes. The YKHM domain is a relatively weak consensus endosomal/lysosomal addressing signal (Bonifacino and Traub, 2003), although it is also found in CTLA-4, a molecule known to recycle upon activation of T cells (Linsley et al., 1996). As suggested by its early endosomes distribution, we could show that BAD-LAMP also recycles in transfected HeLa cells. Whether this is the case in neurons remains to be further investigated, although it clearly indicates that the `YKHM' domain is not normally used as a lysosomal addressing signal.

One of the features of BAD-LAMP-containing organelles is their clustered distribution. This distribution mirrors the organization of the different microdomains at the cell surface. Whether BAD-LAMP-containing organelles participate in the maintenance of this organization within the neuritic plasma membrane remains to be proved. Nevertheless their sensitivity to cholesterol-depleting drugs suggests that microdomains and BAD-LAMP-containing vesicles are functionally linked. Strikingly, the clustering the BAD-LAMP-containing vesicles is also defined by the distribution of the phosphorylated epitopes (SMI31) found on the microtubule-associated protein MAP1B or neurofilament H (Fischer and Romano-Clarke, 1990). MAP1B in the cortex has been strongly implicated in synapse formation and function (Kawakami et al., 2003). Such a role has been recently functionally demonstrated through the observation that mice lacking the phosphorylated form of MAP1B specifically in the hippocampus, show deficits in long-term potentiation in the Schaeffer collaterals pathway (Zervas et al., 2005). Therefore, it is conceivable that MAP1B is implicated in the positioning and transport of BAD-LAMP vesicles at sites of postsynaptic densities on the dendrites of cortical neurons, and that this process could be essential for stabilization, function and plasticity of cortical synapses. Indeed, BAD-LAMP expression is temporally and spatially restricted in cortical neurons of layers II, III and V. Whereas the generation and migration of cortical neurons in rodents is an embryonic process, synaptogenesis in the cortex occurs in the postnatal animal with a peak between P10 and P15 to approach adult values (Micheva and Beaulieu, 1996). This increase in functional synapses in the cortex is strikingly mirrored by the expression of BAD-LAMP during corticogenesis. Thus, it appears very possible that BAD-LAMP, together with MAP1B, is involved in the terminal maturation steps and/or function of defined cortical neurone populations.

Most of our observations point towards a link between BAD-LAMP and endocytosis. The transformation of a transient contact between two neurons into a stable and functional synapse requires major changes in the membrane composition of the respective neuronal surface areas. Endocytic processes have been implicated in the regulation of synaptic function and plasticity in vertebrates (Vissel et al., 2001) and in Drosophila (Dickman et al., 2006). For example, NMDA receptors are subject to constitutive (Roche et al., 2001) as well as agonist-induced (Vissel et al., 2001) internalization through clathrin-mediated endocytosis. Interestingly, in situ hybridization for NMDAR1 resulted in strong cellular labeling in neurons of layers II/III, V and VI (Rudolf et al., 1996), resembling the pattern we found for BAD-LAMP in the postnatal cortex. The BAD-LAMP-containing endocytic compartment could therefore play a regulatory role in these events by maintaining specific zones in the neuronal projections.

Bioinformatics

The BAD-LAMP protein sequence ID in Ensembl database is ENSMUSP00000061180. All LAMPs sequences were aligned using CLUSTALW package (EBI) and results were treated with TreeView for phylogeny. Image analysis was performed with the Image J software and the plugin JacoP.

Animals and tissues

All animals were treated according to protocols approved by the French Ethical Committee. CD1 mice (Iffa-Credo, Town?, France) were used to determine the Bad-lamp expression pattern. Disabled 1 deficient Scrambler mice were purchased from Jackson Laboratories. The day of the vaginal plug appearance was considered as embryonic day (E)0.5 and the day of the birth as postnatal day (P)0. For in situ hybridization and immunohistochemistry, postnatal and adult brains were collected after the animals were anaesthetized with a lethal dose of Rompun/Imalgen 500 and intracardially perfused with 4% paraformaldehyde (PFA). Brains were further fixed in 4% PFA overnight. Adult brains were sectioned at 80 μm on a vibratome whereas P2-P12 brains were cryoprotected in 20% sucrose/PBS, frozen in OCT compound and sectioned at 16 μm on a cryostat. Sections collected on Superfrost slides were treated as described below.

Molecular biology

Northern blot analysis was done with FirstChoice Northern Blot Mouse Blot I (Ambion) using a probe corresponding to exons 4, 5 and 6 of BAD-LAMP (clone IMAGE 2588577). 2 mg of Trizol extracted total mouse cortex RNA was used for reverse transcription with oligo(dT) primers. The cDNAs coding for BAD-LAMP were amplified after 30 cycles of PCR using Taq polymerase. Sense primer was ACC GGC CAC TTT GAG GGA and antisense GGG GCG GCC TTT GCA GCA (1.5 kb). PCR products were cloned into pGEM-Teasy plasmid (Promega). BAD-LAMP-GFP fusion construct was constructed using pEGFP-NI vector (Clontech). FLAG-BAD-LAMP was constructed using pTEJ-8-HA-FLAG plasmid (Didier Marguet, Marseille, France). A tyrosine mutant of BAD, FLAG-BAD-Tyr-276-Ala was produced by targeted PCR mutagenesis. FLAG-BAD-LAMP cDNA were transferred into pCX-MCS2 plasmid, a pCAAGS derived plasmid with an extended cloning site (a kind gift from Xavier Morin, Marseille, France). Dynamin-GFP wt plasmid and dynamin-GFP A44K were kindly given by M. McNiven, Rochester, MN. RNAi constructs pSUPER AP2 μ2 and pSUPER control were a gift from Philippe Benaroch, Paris, France.

In situ hybridization and immunohistochemistry

IMAGE clone 2588577 was used to make an antisense RNA probe. Antisense RNA probes for Bad-lamp, Cux2 (Zimmer et al., 2004) and ER81 (Lin et al., 1998) were generated using the Dig-RNA labelling kit (Roche). Single in situ hybridization and combined in situ hybridization with immunohistochemistry were described previously (Tiveron et al., 1996; Zimmer et al., 2004) for all probes and the NeuN monoclonal mouse IgG (MAB377; Chemicon).

Antibodies and immunocytochemistry

A polyclonal rabbit anti-BAD-LAMP was raised in rabbit against two peptides of the BAD-LAMP cytoplasmic tail, KMTANQVQIPRDRSQC and KQIPRDRSQYKHMC. Anti-synaptotagmin 1 and anti-RAB3a/b antibodies were obtained from P. Di Camilli, New Haven, CT, anti-FLAG M2 antibody and anti-β-tubulin-Cy3 were obtained from Sigma, anti-VAMP2 from SYSY, anti-syntaxin 6 from BD Transduction Laboratories; Aanti-PrP (6H4) was from Prionics (Schlieren, Switzerland), anti-syntaxin 13 from Stressgen (Ann Arbor, MI); anti-Thy1 from Michel Pierres, Marseille, France; human Alexa Fluor 568-Tf from Molecular Probes; mouse FITC-Tf from Rockland (Gilbertsville, PA), Cy3-β-tubulin from Sigma, FITC-cholera toxin B subunit (GM1 staining) from Sigma; anti-NCAM H28 from C. Goridis, Paris, France; Anti-Ti-VAMP from T. Galli, Paris, France; Rat anti-mouse LAMP2 from I. Mellman (New Haven, CT) and anti-human LAMP1 from Abcam. All FITC and Cy3-5 secondary antibodies were from Jackson ImmunoResearch. All Alexa secondary antibodies were from Molecular Probes. Immunofluorescence and confocal microscopy was performed with a Zeiss LSM 510 microscope as described previously (Cappello et al., 2004). Vibratome adult brain sections were immunostained with rabbit anti-BAD-LAMP and mouse anti-MAP2.

Cell culture

HeLa cells were grown in DMEM containing 10% FCS. Cortical neurons were prepared from E15.5 embryonic cortices. Cortices were dissected out in HBSS, treated for 15 minutes at 37°C in Trypsin/EDTA-HBSS (Invitrogen), washed once in NeuroBasal medium (NB; Invitrogen) complemented with 10% horse serum to block trypsin activity and washed once more in NB alone. Cortical neurons were dissociated, plated on glass coverslips in NB with B27 complement, 2 mM L-glutamine and 50 μg/ml penicillin/streptomycin (Invitrogen) and cultured for 3 days at 37°C, 5% CO2. Coverslips were coated overnight with poly-L-lysine (10 μg/ml).

Transfection and internalization experiments

Neurons were electroporated using Amaxa Nucleofactor Kit according to the manufacturer's instructions. HeLa cells were grown on coverslips and transfected using Lipofectamine 2000 (Invitrogen) using the manufacturer's protocol. After 8-24 hours of transfection the HeLa cells were processed to study internalization kinetics or fixed using 3% paraformaldehyde. Internalization assays were performed using FITC-conjugated transferrin or unconjugated antibodies. The cells were first incubated for 20 minutes at 37°C in DMEM/100 mM HEPES to eliminate endogenous transferrin. Cells were incubated for 15 minutes at 4°C with ligand and/or antibody and washed twice in ice-cold PBS before incubation with DMEM, 1% BSA, 100 mM Hepes at 37°C, for different times prior to fixation and immunocytochemistry. Neurons were processed identically in NB medium. Cortical neurone biotinylation was performed using EZ-Link Sulfo-NHS-Biotin kit (Pierce) with a 15-minute reaction time at 4°C, followed by three washes with ice-cold PBS containing 10 mM glycine. Cells were incubated for 5 and 45 minutes at 37°C to allow endocytosis of biotinylated membrane proteins, prior to fixation and immunostaining.

Immunoblots and immunoprecipitation

1% Triton X-100 cell extracts complemented with protease inhibitors cocktail (Roche) were immunoblotted after separation by 12% or a 7-17% gradient SDS-PAGE. Immunoprecipitation with anti-FLAG antibody and N-glycosidase F or endoglycosidase H (Calbiochem) treatment were performed as described previously (Cappello et al., 2004).

This work was supported by grants to P.P. from CNRS-INSERM, the Ministère de la Recherche et de la Technologie (ACI), La Ligue Nationale Contre le Cancer and the Human Frontier of Science Program. A.D. is supported by the MRT and ARC. P.P. is part of the EMBO Young Investigator Program. H.C. was supported by the French Fondation pour la Recherche sur le Cerveau (FRC), the Association Francaise contre le Myopathies (AFM) and the European Community through the NOE NeuroNE. We thank the PICsL imaging core facility for expert technical assistance. We are grateful to Vilma Arce for expert technical advice.

Andrejewski, N., Punnonen, E. L., Guhde, G., Tanaka, Y., Lullmann-Rauch, R., Hartmann, D., von Figura, K. and Saftig, P. (
1999
). Normal lysosomal morphology and function in LAMP-1-deficient mice.
J. Biol. Chem.
274
,
12692
-12701.
Bonifacino, J. S. and Traub, L. M. (
2003
). Signals for sorting of transmembrane proteins to endosomes and lysosomes.
Annu. Rev. Biochem.
72
,
395
-447.
Cappello, F., Gatti, E., Camossetto, V., David, A., Lelouard, H. and Pierre, P. (
2004
). Cystatin F is secreted, but artificial modification of its C-terminus can induce its endocytic targeting.
Exp. Cell Res.
297
,
607
-618.
Coco, S., Raposo, G., Martinez, S., Fontaine, J. J., Takamori, S., Zahraoui, A., Jahn, R., Matteoli, M., Louvard, D. and Galli, T. (
1999
). Subcellular localization of tetanus neurotoxin-insensitive vesicle-associated membrane protein (VAMP)/VAMP7 in neuronal cells: evidence for a novel membrane compartment.
J. Neurosci.
19
,
9803
-9812.
de Saint-Vis, B., Vincent, J., Vandenabeele, S., Vanbervliet, B., Pin, J. J., Ait-Yahia, S., Patel, S., Mattei, M. G., Banchereau, J., Zurawski, S. et al. (
1998
). A novel lysosome-associated membrane glycoprotein, DC-LAMP, induced upon DC maturation, is transiently expressed in MHC class II compartment.
Immunity
9
,
325
-336.
Dehmelt, L. and Halpain, S. (
2004
). Actin and microtubules in neurite initiation: are MAPs the missing link?
J. Neurobiol.
58
,
18
-33.
Del Rio, J. A., Gonzalez-Billault, C., Urena, J. M., Jimenez, E. M., Barallobre, M. J., Pascual, M., Pujadas, L., Simo, S., La Torre, A., Wandosell, F. et al. (
2004
). MAP1B is required for Netrin 1 signaling in neuronal migration and axonal guidance.
Curr. Biol.
14
,
840
-850.
Dickman, D. K., Lu, Z., Meinertzhagen, I. A. and Schwarz, T. L. (
2006
). Altered synaptic development and active zone spacing in endocytosis mutants.
Curr. Biol.
16
,
591
-598.
Dugast, M., Toussaint, H., Dousset, C. and Benaroch, P. (
2005
). AP2 clathrin adaptor complex, but not AP1, controls the access of the major histocompatibility complex (MHC) class II to endosomes.
J. Biol. Chem.
280
,
19656
-19664.
Eskelinen, E. L., Illert, A. L., Tanaka, Y., Schwarzmann, G., Blanz, J., von Figura, K. and Saftig, P. (
2002
). Role of LAMP-2 in lysosome biogenesis and autophagy.
Mol. Biol. Cell
13
,
3355
-3368.
Eskelinen, E. L., Tanaka, Y. and Saftig, P. (
2003
). At the acidic edge: emerging functions for lysosomal membrane proteins.
Trends Cell Biol.
13
,
137
-145.
Fischer, I. and Romano-Clarke, G. (
1990
). Changes in microtubule-associated protein MAP1B phosphorylation during rat brain development.
J. Neurochem.
55
,
328
-333.
Gruenberg, J. and Stenmark, H. (
2004
). The biogenesis of multivesicular endosomes.
Nat. Rev. Mol. Cell Biol.
5
,
317
-323.
Hirling, H., Steiner, P., Chaperon, C., Marsault, R., Regazzi, R. and Catsicas, S. (
2000
). Syntaxin 13 is a developmentally regulated SNARE involved in neurite outgrowth and endosomal trafficking.
Eur. J. Neurosci.
12
,
1913
-1923.
Janvier, K. and Bonifacino, J. S. (
2005
). Role of the endocytic machinery in the sorting of lysosome-associated membrane proteins.
Mol. Biol. Cell
16
,
4231
-4242.
Kawakami, S., Muramoto, K., Ichikawa, M. and Kuroda, Y. (
2003
). Localization of microtubule-associated protein (MAP) 1B in the postsynaptic densities of the rat cerebral cortex.
Cell. Mol. Neurobiol.
23
,
887
-894.
Kennedy, M. J. and Ehlers, M. D. (
2006
). Organelles and trafficking machinery for postsynaptic plasticity.
Annu. Rev. Neurosci.
29
,
325
-362.
Lin, J. H., Saito, T., Anderson, D. J., Lance-Jones, C., Jessell, T. M. and Arber, S. (
1998
). Functionally related motor neuron pool and muscle sensory afferent subtypes defined by coordinate ETS gene expression.
Cell
95
,
393
-407.
Linsley, P. S., Bradshaw, J., Greene, J., Peach, R., Bennett, K. L. and Mittler, R. S. (
1996
). Intracellular trafficking of CTLA-4 and focal localization towards sites of TCR engagement.
Immunity
4
,
535
-543.
Madore, N., Smith, K. L., Graham, C. H., Jen, A., Brady, K., Hall, S. and Morris, R. (
1999
). Functionally different GPI proteins are organized in different domains on the neuronal surface.
EMBO J.
18
,
6917
-6926.
Maxfield, F. R. and Tabas, I. (
2005
). Role of cholesterol and lipid organization in disease.
Nature
438
,
612
-621.
McCormick, P. J., Martina, J. A. and Bonifacino, J. S. (
2005
). Involvement of clathrin and AP-2 in the trafficking of MHC class II molecules to antigen-processing compartments.
Proc. Natl. Acad. Sci. USA
102
,
7910
-7915.
Micheva, K. D. and Beaulieu, C. (
1996
). Quantitative aspects of synaptogenesis in the rat barrel field cortex with special reference to GABA circuitry.
J. Comp. Neurol.
373
,
340
-354.
Rice, D. S. and Curran, T. (
1999
). Mutant mice with scrambled brains: understanding the signaling pathways that control cell positioning in the CNS.
Genes Dev.
13
,
2758
-2773.
Roche, K. W., Standley, S., McCallum, J., Dune Ly, C., Ehlers, M. D. and Wenthold, R. J. (
2001
). Molecular determinants of NMDA receptor internalization.
Nat. Neurosci.
4
,
794
-802.
Rudolf, G. D., Cronin, C. A., Landwehrmeyer, G. B., Standaert, D. G., Penney, J. B., Jr and Young, A. B. (
1996
). Expression of N-methyl-D-aspartate glutamate receptor subunits in the prefrontal cortex of the rat.
Neuroscience
73
,
417
-427.
Saftig, P., Tanaka, Y., Lullmann-Rauch, R. and von Figura, K. (
2001
). Disease model: LAMP-2 enlightens Danon disease.
Trends Mol. Med.
7
,
37
-39.
Santuccione, A., Sytnyk, V., Leshchyns'ka, I. and Schachner, M. (
2005
). Prion protein recruits its neuronal receptor NCAM to lipid rafts to activate p59fyn and to enhance neurite outgrowth.
J. Cell Biol.
169
,
341
-354.
Shikano, S., Bonkobara, M., Zukas, P. K. and Ariizumi, K. (
2001
). Molecular cloning of a dendritic cell-associated transmembrane protein, DC-HIL, that promotes RGD-dependent adhesion of endothelial cells through recognition of heparan sulfate proteoglycans.
J. Biol. Chem.
276
,
8125
-8134.
Sunyach, C., Jen, A., Deng, J., Fitzgerald, K. T., Frobert, Y., Grassi, J., McCaffrey, M. W. and Morris, R. (
2003
). The mechanism of internalization of glycosylphosphatidylinositol-anchored prion protein.
EMBO J.
22
,
3591
-3601.
Sytnyk, V., Leshchyns'ka, I., Delling, M., Dityateva, G., Dityatev, A. and Schachner, M. (
2002
). Neural cell adhesion molecule promotes accumulation of TGN organelles at sites of neuron-to-neuron contacts.
J. Cell Biol.
159
,
649
-661.
Sytnyk, V., Leshchyns'ka, I., Dityatev, A. and Schachner, M. (
2004
). Trans-Golgi network delivery of synaptic proteins in synaptogenesis.
J. Cell Sci.
117
,
381
-388.
Tanaka, Y., Guhde, G., Suter, A., Eskelinen, E. L., Hartmann, D., Lullmann-Rauch, R., Janssen, P. M., Blanz, J., von Figura, K. and Saftig, P. (
2000
). Accumulation of autophagic vacuoles and cardiomyopathy in LAMP-2-deficient mice.
Nature
406
,
902
-906.
Tiveron, M. C., Hirsch, M. R. and Brunet, J. F. (
1996
). The expression pattern of the transcription factor Phox2 delineates synaptic pathways of the autonomic nervous system.
J. Neurosci.
16
,
7649
-7660.
Vissel, B., Krupp, J. J., Heinemann, S. F. and Westbrook, G. L. (
2001
). A use-dependent tyrosine dephosphorylation of NMDA receptors is independent of ion flux.
Nat. Neurosci.
4
,
587
-596.
Zervas, M., Opitz, T., Edelmann, W., Wainer, B., Kucherlapati, R. and Stanton, P. K. (
2005
). Impaired hippocampal long-term potentiation in microtubule-associated protein 1B-deficient mice.
J. Neurosci. Res.
82
,
83
-92.
Zimmer, C., Tiveron, M. C., Bodmer, R. and Cremer, H. (
2004
). Dynamics of Cux2 expression suggests that an early pool of SVZ precursors is fated to become upper cortical layer neurons.
Cereb. Cortex
14
,
1408
-1420.

Supplementary information