You are on page 1of 21

Article

Neuronal paxillin and drebrin mediate BDNF-induced


force transduction and growth cone turning in a soft-
tissue-like environment
Graphical abstract Authors
Chen Chen, Chien-Hsin Chu, Ying Chu, ...,
Bi-Chang Chen, Hsiung-Lin Tu,
Pei-Lin Cheng

Correspondence
plcheng@imb.sinica.edu.tw

In brief
Chen et al. report that growth cones of
0.1 kPa neurons exhibit a highly
coordinated T-zone activity. Neurotro-
phic-factor-directed growth cone move-
ment on soft substrates depends on
drebrin phosphorylation and paxillin-
drebrin association, but not on Src activ-
ity, a mechanism distinct from the one
observed on conventional glass
coverslips.

Highlights
d Substrate stiffness changes molecular architectures of
growth cone subdomains

d T-zone factor drebrin modulates growth cone movement on


soft-tissue-like substrates

d Drebrin facilitates BDNF-elicited pressing force and


attraction on soft substrates

Chen et al., 2022, Cell Reports 40, 111188


August 16, 2022 ª 2022 The Author(s).
https://doi.org/10.1016/j.celrep.2022.111188 ll
ll
OPEN ACCESS

Article
Neuronal paxillin and drebrin mediate
BDNF-induced force transduction and growth
cone turning in a soft-tissue-like environment
Chen Chen,1,4 Chien-Hsin Chu,1,4 Ying Chu,1,4 Ting-Ya Chang,1 Sheng-Wen Chen,1 Shu-Yang Liang,1 Yun-Chi Tsai,2
Bi-Chang Chen,2 Hsiung-Lin Tu,3 and Pei-Lin Cheng1,5,*
1Institute of Molecular Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
2Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
3Institute of Chemistry, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
4These authors contributed equally
5Lead contact

*Correspondence: plcheng@imb.sinica.edu.tw
https://doi.org/10.1016/j.celrep.2022.111188

SUMMARY

Soft tissue environments govern neuronal morphogenesis. However, the precise molecular mechanisms un-
derlying chemotropism-directed axonal growth cone movement in extremely soft environments remain un-
clear. Here, we show that drebrin, a growth cone T-zone protein, modulates growth cone turning in response
to brain-derived neurotrophic factor (BDNF) coated on a soft substrate. Structurally, axonal growth cones of
rodent hippocampal neurons grown on 0.1 kPa hydrogels possess an expanded T zone in which drebrin is
highly integrated with both F-actin and microtubules. Biochemically, we identify paxillin as interacting with
drebrin in cells grown on 0.1 kPa hydrogels but not on glass coverslips. When grown on 0.1 kPa substrates,
growth cones asymmetrically exposed to BDNF-bound stripes exhibit enhanced paxillin-drebrin interaction
on the side facing the stripes, an activity that is PKA and AAK1 dependent but independent of Src kinase.
Functionally, we show that BDNF-induced growth cone turning and force generation on soft substrates
require drebrin phosphorylation and paxillin-drebrin association.

INTRODUCTION bule bundles emanating from the axonal shaft; (2) the peripheral
domain (P domain), distributed around the leading edge and
Axon pathfinding activity requires orientation of growth cones in mainly composed of lamellipodia and filopodia; and (3) the tran-
brain tissue, where the elastic modulus can be as low as 0.1– sition zone (T zone), representing the interface between C and P
1 kPa (Balgude et al., 2001; Koser et al., 2016; Tyler, 2012). domains (Lowery and Van Vactor, 2009) and containing microtu-
Growth cone attraction or repulsion on a soft substrate may bule-actin crosslinking molecules such as non-muscle myosin II
not necessarily employ the same set of force transduction ma- and developmentally regulated brain protein, or drebrin (Bur-
chineries as those functioning in motility of non-neuronal cells nette et al., 2008; Dent and Gertler, 2003; Geraldo and Gor-
on more rigid substrates (Koch et al., 2012; Koser et al., 2016). don-Weeks, 2009; Shin et al., 2014; Shirao et al., 2017; Shirao
However, in vitro mechanistic studies of growth cone dynamics and Obata, 1985). Drebrin belongs to a class of neuron-specific
have mostly been performed on tissue culture plastic or on glass F-actin side binding/bundling proteins and has two major iso-
coverslips whose mechanical properties do not resemble those forms, namely, embryonic-type drebrin E and adult-type drebrin
of embryonic brain tissues. Hence, mechanisms that drive force A (Geraldo et al., 2008; Ishikawa et al., 1994; Shirao, 1995; Shirao
generation and directed movements of highly mobile growth et al., 2017). Drebrin A accumulates within dendritic spines to
cones in response to chemical factors bound to soft-tissue-like enable postsynaptic protein assembly (Hayashi et al., 1996;
environment remain to be addressed. Mizui et al., 2005; Takahashi et al., 2003), while drebrin E is a
Mechanistically, growth cone movement behaviors evoked by typical T-zone-residing molecule expressed during neurogene-
chemical and physical instructive cues require actin filopodia sis and facilitating neuronal motility (Dun et al., 2012; Geraldo
stability and microtubule capture at the leading edges (Dent et al., 2008; Mizui et al., 2009; Trivedi et al., 2017). Actin-stabiliz-
and Gertler, 2003; Geraldo and Gordon-Weeks, 2009; Kolodkin ing drebrin binds to the microtubule plus-end tracking protein
and Tessier-Lavigne, 2011; Lowery and Van Vactor, 2009; Ros- EB3 (Geraldo et al., 2008; Merriam et al., 2013), which may
ner et al., 2007). Structurally, growth cones are divided into three modulate consumption of chemical energy by microtubule
subregions: (1) the central domain (C domain), located at or near extension and/or non-muscle myosin II-mediated contraction
the distal end of the axonal shaft, which is composed of microtu- (Ketschek et al., 2007; Mizui et al., 2009; Rosner et al., 2007).

Cell Reports 40, 111188, August 16, 2022 ª 2022 The Author(s). 1
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
ll
OPEN ACCESS Article

(legend on next page)

2 Cell Reports 40, 111188, August 16, 2022


ll
Article OPEN ACCESS

These actions represent the molecular basis for growth cone scribes two types of mechanical force transmission linking sepa-
dynamics. rate cytoskeletal systems of growth cones: (1) a contractile force
To address whether and how the common force transmission driven by the motor protein myosin II in the T zone and (2) a thrust
machinery of growth cones functions in a soft-tissue-like hydro- force driven by F-actin polymerization in the P domain (Betz
gel, we examined differential cytoskeletal integration of growth et al., 2011; Forscher and Smith, 1988; Katoh et al., 1999; Le
cone molecules on 0.1 kPa polyacrylamide (PA) hydrogels Clainche and Carlier, 2008; Medeiros et al., 2006; Pollard and
compared with glass coverslips. Morphologically, we observed Borisy, 2003; Woo and Gomez, 2006). However, it had been un-
a compact fist-like actin-based structure for growth cones of clear whether this model applied to rodent hippocampal or
hippocampal neurons grown on 0.1 kPa hydrogels, unlike open cortical neurons grown in a compliant native tissue. To answer
palm-like structures exhibited by growth cones grown on cover- this question, we performed super-resolution structured illumi-
slips. Growth cones on 0.1 kPa hydrogels exhibited significantly nation microscopy (3D SIM; Figures 1A and 1B) to analyze
greater T-zone integration with the cytoskeletal P domain than morphology of growth cones of hippocampal neurons cultured
conventional cultures on coverslips. Drebrin knockdown (KD) 2–3 days in vitro (DIV). Polyacrylamide (PA) hydrogel (0.1 kPa)
in newly differentiated rat layer 2/3 cortical neurons promoted was designed to mimic mechanical properties of embryonic
relative shortening of commissural axons. Mechanistically, out brain (E = 0.1–1 kPa). At 2 DIV, most neurons grown on
of eight factors associated with cytoskeletal dynamics exam- 0.1 kPa exhibited a polarized morphology, with one minor neurite
ined, paxillin and the brain-derived neurotrophic factor (BDNF) longer than the others. By contrast, neurons grown on coverslips
receptor TrkB displayed substrate rigidity-specific enrichment showed an approximately 1-day delay in reaching the same
at the growth cone T zone. Paxillin directly associated with dre- extent of polarization relative to neurons grown on 0.1 kPa gels
brin, and their binding affinity increased when neurons were (Figure S1). Thus, we acquired images from DIV 1–2 cultures
grown on 0.1 kPa hydrogels. KD of either paxillin or drebrin in on 0.1 kPa gels and from DIV 2–3 cultures on coverslips and
hippocampal neurons lowered the stress exerted on 0.1 kPa hy- focused on comparing growth cones of either the nascent
drogels, and corresponding growth cones failed to respond to a axon or the most extended or enlarged minor neurites. Analysis
steep BDNF gradient coated on the 0.1 kPa surface. Importantly, of cytoskeletal organization of growth cones on 0.1 kPa hydro-
BDNF-elicited force generation and growth cone turning, which gels revealed a fist-like, compact, F-actin-rich P domain and
requires expression, phosphorylation, and interaction of paxillin an enlarged T zone (defined by drebrin staining) extensively over-
and drebrin, was not dependent on Src kinase activity, which lapping the C domain (Figures 1B and 1C). By contrast, growth
is required for motility of growth cones on glass coverslips. cones of neurons grown on glass coverslips exhibited a wide P
Together, our findings suggest that an extremely soft surface domain with significantly reduced spatial overlap among cyto-
transforms growth cone structures in a manner that favors skeletal components (Figures 1B and 1C). Furthermore, when
drebrin-paxillin interactions, which then enable generation of in- we examined the arrangement of microtubules in 16-h neuronal
ternal mechanical force required for efficient growth cone cultures using expansion microscopy (Figure S1), the direction of
orientation. microtubule bending near the cell periphery was outward on
0.1 kPa and inward in coverslip cultures. These observations
RESULTS support the idea that native brain tissue or 0.1 kPa substrate,
which resembles the stiffness of brain tissue, is a more permis-
The growth cone T zone on brain-tissue-mimicking sive environment for microtubule protrusions from the cell
hydrogels exhibits alternative molecular architecture body than is a glass coverslip.
In conventional Banker’s neuronal culture on coverslips (Dotti To assess the function of growth cone T-zone molecules in
et al., 1988), the classical F-actin retrograde flow model de- terms of axonal outgrowth of newborn neurons in neonatal

Figure 1. Axonal growth cones of hippocampal neurons grown on 0.1 kPa hydrogels exhibit a distinct T zone with a high degree of paxillin-
drebrin co-localization
(A) Top and side views of 3D SIM images of hippocampal neurons grown on 0.1 kPa PA hydrogels or glass coverslips for 2 days in vitro (DIV), immunostained for
the T-zone factor drebrin (green in merge panels), microtubules (blue), and phalloidin for F-actin (red). Scale bars, 5 mm.
(B) Traces of staining intensity reveal a larger drebrin-positive growth cone T zone (gray rectangles) in 0.1 kPa hydrogels than on glass coverslips.
(C) Pie charts show percentage of drebrin, phalloidin, and microtubule co-localization based on signal intensity at the growth cone C domain or T zone of neurons
grown on substrates of varying stiffness.
(D) Fluorescence images of P3 rat cortices transfected in utero at E17 with IRES constructs harboring mScarlet and scrambled shRNA (SC shRNA) or drebrin
shRNA (DBN shRNA). Scale bars, 200 mm. The bottom plot shows relative mScarlet level of transfected cortical axons between the initial position (-2) and indi-
cated positions (dashed boxes) of the corpus callosum (±SEM, n = 3 independent experiments, >2 cortices for each drebrin shRNA and littermate control groups;
***p < 0.001, multiple t test).
(E) Images showing hippocampal neurons grown on 0.1 kPa hydrogels and coverslips, immunostained by antibodies against drebrin (top panel), paxillin (top
panel), TrkB (bottom panel), non-muscle myosin II (NMII; bottom panel), and phalloidin at 2 DIV. Scale bars, 5 mm.
(F) Summary scatterplot showing Pearson’s coefficient for protein co-localization with drebrin on 0.1 kPa culture and coverslips, as indicated (±SEM, n > 7 growth
cones each groups; ****p < 0.0001, t test).
(G) Images of hippocampal neurons grown on 0.1 kPa hydrogels and coverslips, immunostained with drebrin and paxillin (PXN) antibodies at 2 DIV. Repre-
sentative surface rendering 3D images sampled from the growth cone showing the co-localization surface (yellow). Volume thickness is 3 mm. Scale bars, 5 mm.
(H) Histograms showing the percentage of drebrin co-localizing with cytoskeletal proteins (left panel) and paxillin (right panel) in the growth cone of neurons grown
on 0.1 kPa hydrogels than for those grown on glass coverslips (±SEM, n = 7–13 cells for each set of experiments; **p < 0.01; ****p < 0.0001 by unpaired t test).

Cell Reports 40, 111188, August 16, 2022 3


ll
OPEN ACCESS Article

cortex in vivo, we performed in utero electroporation to express cultured on substrates of varying stiffness. Our in vivo co-IP anal-
constructs encoding short hairpin RNA (shRNA) against drebrin ysis detected paxillin associating with drebrin in immunoprecip-
or scrambled control in a subpopulation of neural progenitor itates from brain lysates, but not from hepatic or cardiac tissues
cells of 17-day rat embryos (E17; Figure 1D). To extend analysis (Figure 2A). Interestingly, paxillin and drebrin exhibit a similar
of growth cone phenotypes, we obtained brain slices from expression time course in the developing brain cortex, both
newborn (postnatal day 3 [P3]) rat cortex when most commis- peaking at P0 and then progressively declining (Figure 2B), sug-
sural axons have crossed the midline. Nearly all (100%) gesting they are temporally and spatially associated with each
newborn cortical neurons at P3 arriving at cortical layer 2/3 ex- other at early stages of neuronal development.
hibited a neurite-bearing, polarized morphology (Figure 1D), Next, we asked whether the degree of paxillin-drebrin associ-
with dendritic arbors oriented toward the pial surface and an ation reflects mechanical stiffness of substrates. Levels of dre-
axon oriented radially in the cortical plate. At the same time, brin immunoprecipitated by an anti-paxillin antibody from neu-
most (76%) control commissural axon tips (namely, those ex- rons grown on 0.1 kPa hydrogels were significantly higher
pressing mScarlet tracer) had reached the contralateral side of than that seen in comparable analysis of neurons grown on cov-
the corpus callosum (Figure 1D). By contrast, drebrin KD cortical erslips (Figure 2C), suggesting higher binding affinity in a soft
neurons exhibited retardation of axon growth after crossing environment. Moreover, histidine-tagged drebrin co-purified
the midline. Quantitative analysis indicated that drebrin KD with GST-tagged paxillin in an in vitro binding assay, suggesting
decreased axon length by 20% on the contralateral side of direct interaction (Figure 2D). In vitro domain-mapping analysis
corpus callosum at P3 (Figure 1D). The shortened axon pheno- indicated that the paxillin C terminus, which harbors the LIM
type was also observed in drebrin KD cortical neurons at P6 (Fig- domains, is required and sufficient for drebrin interaction
ure S2). Consequently, the extent of branching and innervation of (Figures S4A and S4B). Notably, the paxillin N-terminal LD motif
drebrin KD callosal axons to contralateral cortical layer 2/3 at alone is insufficient for drebrin interaction but had a modulatory
P16 was significantly less than that seen in control axons (Fig- effect on binding affinity of the paxillin LIM domain for drebrin
ure S2). These results suggest that T-zone factors are important (Figure S4). Domain-mapping experiments showed that the dre-
for proper growth and targeting of axonal projections of newly brin N-terminal actin-depolymerizing factor homology (ADFH)
generated cortical neurons in vivo. domain is required for paxillin association (Figures S4C and
To identify compliant substrate-dependent T-zone accessory S4D). Importantly, confocal imaging of drebrin KD or control
factors, we assessed differential expression in growth cones on neurons grown on 0.1 kPa hydrogels indicated a relative loss
substrates with varying rigidity of eight factors linked to growth of immunostaining for paxillin at T zone in drebrin KD samples
cone movement or environmental sensing (Figure S3). Among (Figure 2E), suggesting that drebrin is required for paxillin accu-
them we found that paxillin modulates actin dynamics, and the mulation at the growth cone T zone on these substrates.
BDNF receptor TrkB preferentially accumulated at the T zone Together, these findings indicate that substrate softness enables
of neurons grown on 0.1 kPa hydrogels, a pattern not seen drebrin to recruit neuronal paxillin to the growth cone T zone.
when cells were grown on coverslips (Figures 1E–1H). Despite
the fact that T-zone formation is not dependent on substrate Expression of paxillin and drebrin is necessary for
elasticity (Figure 1E), levels of drebrin co-localizing with traction force generated by hippocampal neurons grown
the F-actin-stained P domain, the microtubule end-stained C on 0.1 kPa hydrogels
domain, and paxillin were significantly higher (3-fold) on To confirm functional association between paxillin and drebrin,
0.1 kPa hydrogels than on coverslips (Figures 1G and 1H). These we assessed growth cone force generation and motion in soft
enhanced molecular associations may be due in part to the environments. We took advantage of the fact that 0.1 kPa hydro-
compact architecture seen at the tip of 0.1 kPa growth cones gels preserve structural features of advancing growth cones to
(compare paxillin staining in Figures 1E and 1G). These findings analyze force transduction upon cell-substrate interactions.
suggest that a compliant environment ensures proper engage- First, we compared the degree of integration of the core compo-
ment and coordination of T-zone factors (such as myosin II, dre- nent of the force generation machinery, namely, non-muscle
brin, and paxillin) with the growth cone cytoskeleton. myosin II (NMII), at the growth cone T zone (Shin et al., 2014)
with P- and C-cytoskeletal domains in neurons grown either on
T-zone protein drebrin associates with paxillin in a 0.1 kPa hydrogels or coverslips (Figure 3A). We observed signif-
substrate elasticity-dependent manner icantly higher (3-fold) levels of NMII co-localizing with F-actin in
Given that paxillin accumulates at the T zone of growth cones on growth cones in neurons grown on 0.1 kPa hydrogels relative to
0.1 kPa hydrogels (Figures 1E and 1G), we postulated that pax- coverslips (Figure 3A3), a trend comparable with that seen with
illin physically and functionally interacts with factors modulating drebrin (Figures 1A–1C). These observations suggest more effi-
growth cone dynamics in such environments. Others had previ- cient growth cone motility in soft environments.
ously reported that embryonic drebrin is neuron specific (Shirao To investigate that possibility, we used traction force micro-
and Obata, 1986), and we had previously identified drebrin as a scopy (TFM) (0.5 frames per second over 10 min), which quan-
paxillin-associating factor via liquid chromatography-tandem tifies particle displacements due to stress generated by growth
mass spectrometry analyses (Chang et al., 2017). Thus we cones pressing against a 0.1 kPa hydrogel surface, to evaluate
examined physical association of paxillin with drebrin in rat em- changes in force transduction (Figure 3B). The average applied
bryonic brain lysates and in vivo co-immunoprecipitation (co-IP) force measured from control hippocampal neurons (at 2 DIV)
of cortical tissue lysates or lysates obtained from neurons was 0.4 pN/m2 for the area of the cell body and growth cone of

4 Cell Reports 40, 111188, August 16, 2022


ll
Article OPEN ACCESS

Figure 2. Paxillin directly binds drebrin in neurons grown on soft substrates


(A) Paxillin-associated complexes were immunoprecipitated (IP) from lysates of indicated E17.5 rat tissues using paxillin antibody (a-PXN) and detected by
western blot analysis. Normal rabbit immunoglobulin G (IgG) served as a negative control. Histograms show an increasing binding preference of paxillin toward
drebrin on soft (0.1 kPa) versus rigid (glass) substrates. Data represent mean intensity ± SEM (n > 3 independent experiments; ***p < 0.001; ****p < 0.0001 by
t test).
(B) Western blots (top panel) of lysates showing drebrin and paxillin protein expression at indicated developmental stages of cerebral cortex. Quantification
(bottom panel) of data shown in blots above. Data represent mean ± SEM from more than three independent experiments.
(C) Similar to (A), except cell lysate was obtained from cortical neuronal cultures grown on indicated substrates.
(D) Western blot showing direct paxillin-drebrin interaction. Bacterially expressed drebrin-His was purified by fast protein liquid chromatography and subjected to
a GST pull-down assay using GST-fused paxillin (GST-PXN) or GST alone. Precipitates were analyzed by western blotting with drebrin antibodies. Histogram
quantifies pull-down data (±SEM, n = 3; normalized to corresponding GST-PXN or GST inputs; ***p < 0.001 by t test).
(E) Representative confocal images of 2-DIV neurons on 0.1 kPa hydrogels incubated with scrambled control (SC) or drebrin siRNAs 30 min after cell plating,
followed by co-immunostaining for paxillin (PXN) and drebrin, and phalloidin staining for F-actin. Scale bars, 5 mm. Traces (bottom left) and histograms (bottom
right) showing the paxillin staining intensity in various cell compartments and the relative paxillin level between growth cone subdomains grown on 0.1 kPa hydro-
gels (±SEM; n = 20–25 cells for each group from three independent experiments; *p < 0.05; ns, not significant by ANOVA with multiple comparison test).

the longest neurite (Figure 3B2). After pretreating cells with eration on 0.1 kPa hydrogels (Figures 3B4 and 3B5). Ectopic
the NMII inhibitor blebbistatin (50 mM), those stress values expression of a drebrin construct harboring a phospho-
slightly but significantly decreased over the entire cell area mimicking mutation (YFP-DBNS142D) restored force generation
(Figures 3B2 and 3B3). However, pharmacological block of in- in drebrin KD growth cones (Figures 3B4 and 3B5), supporting
tegrin-regulated FAK-Src signaling by pretreatment with the the idea that FAK-Src signaling-independent force generation
Src kinase inhibitor PP2 (50 nM) did not alter the average force requires paxillin and drebrin expression.
generated by neuronal cell bodies or growth cones for the We propose that neurons grown on stiff versus soft substrates
10-min interval (Figures 3B2 and 3B3). These findings indicate differ in force generation and neurite extension modes. To
that cells on 0.1 kPa hydrogels exert forces in an NMII activity- examine this possibility, we assessed neurite growth of neurons
dependent but FAK-Src activity-independent manner. More- grown on substrates of varying rigidity in the presence and
over, drebrin or paxillin KD by respective small interfering absence of NMII inhibitor blebbistatin (Figure S5). Blebbistatin-
RNAs (siRNAs) significantly reduced stress (applied force) gen- treated hippocampal neurons grown on coverslips exhibited a

Cell Reports 40, 111188, August 16, 2022 5


ll
OPEN ACCESS Article

Figure 3. Myosin II-mediated force generation of growth cones grown on 0.1 kPa hydrogels requires paxillin and drebrin expression, but not
Src kinase activity
(A) (A1) Myosin II distribution at the growth cone of 2-DIV hippocampal neurons grown on 0.1 kPa PA hydrogels or glass coverslips. Scale bars, 5 mm. Traces of
staining intensity reveal a compact growth cone with greater overlap of T-zone myosin II (NMII; marked by gray rectangles) with cytoskeletal proteins in neurons
grown on 0.1 kPa hydrogels compared with when grown on glass coverslips. (A2 and A3) Traces showing the staining intensity of a single cell (A2) and the aver-
aged intensity (n > 25 cells for each group; A3) of the indicated molecules in various cell compartments of neurons grown on 0.1 kPa hydrogels and coverslips.
Data represent mean ± SEM (*p < 0.05; ns, not significant by ANOVA with multiple comparison test).
(B) Myosin II-mediated contractile force measured by traction force microscopy. (B1) Schematic showing traction force microscopy and force-field visualization.
PIV, particle image velocimetry. FTTC, Fourier transform traction cytometry. (B2 and B4) Representative TFM images of vector fields plotted over magnitudes of
displacement (presented as pseudocolor maps in a linear scale) of 2-DIV hippocampal neurons grown on 0.1 kPa hydrogels in the presence or absence of phar-
macological inhibitors (B2; 50 mM myosin II inhibitor (±)-blebbistatin [(±)-Bleb] or 50 nM Src kinase inhibitor PP2) or siRNA-mediated knockdown of paxillin (PXN)
or drebrin (DBN) expression, and/or ectopic expression of phosphorylation mimicking YFP-DBNS142D (B4), as indicated. Scale bars, 20 mm. (B3 and B5) Histo-
grams summarizing averaged stress exerted by the entire image field (Whole Field), growth cone (Growth Cone), and cell body (Soma). Data represent mean ±
SEM (n > 10 cells for each set of experiments; *p < 0.05; **p < 0.01; ns, not significant by t test).

collapse of filopodia on growth cones as well as more curved and ity of growth cones on a soft substrate requires T-zone factors
elongated processes (both dendrites and axons) relative to un- and paxillin.
treated control neurites on coverslips (Figure S5), suggesting a
relaxation of the NMII-generated actin arc barrier that restricts Asymmetric exposure to BDNF biases paxillin
microtubule protrusion. In contrast, growth cone morphology distribution and growth cone turning on soft substrates
in cells grown on 0.1 kPa gels was little changed by blebbistatin independent of Src
treatment, and neurite length of those neurons was significantly Next, we asked whether the turning direction of growth cones
shortened (Figure S5), implying impaired regulation of microtu- grown on soft substrates is dictated by paxillin and drebrin. To
bule dynamics. These findings also support the notion that those do so, we established a turning assay for growth cones grown
mechanisms controlling neurite extension are different in neu- on substrates of varying rigidity (Figure 4). We counter-printed
rons grown on 0.1 kPa gel from those grown on coverslips. the chemotropic factor BDNF (Gallo et al., 1997; Oberstar
Concomitantly, drebrin and/or paxillin KD in neurons grown on et al., 1997) and/or fluorescent tag-conjugated BSA stripes
0.1 kPa hydrogels resulted in shortening of axons relative to con- (50 mm wide, spaced 100 mm apart) onto surfaces of 0.1 kPa hy-
trol (scrambled siRNA) neurons (Figure S6). Overall, these find- drogels or glass coverslips and then analyzed chemotropism of
ings indicate that proper neurite extension and mechanical activ- growth cones approaching the stripes as well as their molecular

6 Cell Reports 40, 111188, August 16, 2022


ll
Article OPEN ACCESS

Figure 4. Different signaling pathways mediate BDNF-induced growth cone attraction for neurons grown on 0.1 kPa hydrogels or glass
coverslips
(A) (A1) Schematic showing procedure used for BDNF/BSA-stripe printing onto surface of 0.1 kPa hydrogels. (A2) Fluorescent image of a stripe coated with
BDNF-AlexaFluor647. Right panels show fluorescence intensity across the stripe boundary. Scale bar, 3 mm.
(B) Images of growth cones growing near a stripe coated with fluorescently conjugated BDNF or control BSA on substrates of various elastic moduli, immu-
nostained with drebrin and paxillin antibodies (presented as pseudocolor maps in a linear scale) and phalloidin for F-actin at 2 DIV. Dashed line marks the central
axis of growth cones. Scale bars, 5 mm.
(C) Schematic diagrams (top panel) depict turning angles of the growth cone near BDNF or control BSA-coated stripes (blue rectangle). Plots show similar
percentages of growth cones turning toward (positive angles) BDNF stripes for neurons grown on substrates of varying stiffness. Only neurons with growth cones
located near the stripe boundary were counted.
(D) Quantitative measurement showing the percentage of drebrin co-localizing with paxillin on the side of growth cones facing toward or away from the BDNF or
BSA-coated stripes on substrates on varying stiffness. Data were calculated by dividing the co-localized volume on the indicated side by the total drebrin stained
volume, and presented as mean ± SEM (>18 cells for each group from three independent experiments; **p < 0.01; ns, not significant by t test).
(E) Similar to (D), except that preferential pattern of drebrin-paxillin co-localization (denoted as ‘‘PXNXdrebrin’’) is assessed based on a co-localization index (CI)
formula, as indicated. Data represent mean ± SEM (>18 cells for each group from three independent experiments; *p < 0.05; ns, not significant by t test).
(F) Similar to (C) and (E), except cells were subjected to bath application of pharmaceutical inhibitors of AAk1, PKA (KT 5720), or Src tyrosine kinase (PP2), as
indicated. Data represent mean ± SEM (>10 cells for each group from three independent experiments; **p < 0.01; ***p < 0.001; ns, not significant by t test).
(G) Similar to (F), except cells were cultured on BDNF-stripe-coated glass coverslips. Only neurons with growth cones located near the stripe boundary were
counted.

Cell Reports 40, 111188, August 16, 2022 7


ll
OPEN ACCESS Article

repositioning toward BDNF-coated stripes (Figures 4A–4E). As Using PLA analysis, we also detected direct interaction be-
shown in Figures 4B and 4C, BDNF-coated stripes significantly tween TrkB and paxillin in the cell body of neurons at 2 DIV grown
attracted growth cones grown on both 0.1 kPa hydrogels and on 0.1 kPa gels in the absence of BDNF treatment (Figure S6).
glass coverslips, with most (>70%) turning toward BDNF stripes. Interestingly, treatment of 0.1 kPa cultures with BDNF increased
In contrast, turning behavior of growth cones on 0.1 kPa gels or paxillin association (based on increases in PLA puncta density)
coverslips relative to control BSA-coated stripes was more with TrkB in both cell bodies and growth cones relative to con-
random (Figures 4B and 4C). When we compared protein distri- trols (Figure S7). To determine whether this activity resulted in
butions on sides of the growth cone (i.e., facing toward stripes phosphorylation of paxillin-drebrin complexes, we treated
versus away from them), we observed preferential paxillin and cortical neurons cultured on 0.1 kPa gels with and without
drebrin co-localization on the side facing BDNF stripes on BDNF and then subjected cell lysates to immunoprecipitation
0.1 kPa hydrogels (Figures 4D and 4E; co-localization index > 1), with an anti-paxillin antibody, followed by immunoblotting with
suggesting that asymmetric exposure to BDNF on soft surface antibodies against potential paxillin-associated factors,
promotes paxillin-drebrin association. We did not, however, including CIP4, vinculin, and drebrin, as well as S142 phosphor-
observe the same effects for growth cones grown on coverslips ylated drebrin (p-drebrinS142). Indeed, levels of drebrin and
(Figures 4D and 4E), implying that the paxillin-drebrin association p-drebrinS142 co-immunoprecipitated with paxillin increased in
is weak in neurons grown on glass. BDNF-treated cells (Figures S8A and S8B). While we also
Given that newborn neurons grown on compliant substrates observed greater association of paxillin with CIP4 in BDNF-
(E < 1 kPa) switch the relative dominance of endocytosis over treated cells, we did not detect vinculin/paxillin association in
adhesion signaling for neurite initiation (Chang et al., 2017), we 0.1 kPa cultures before or after BDNF treatment (Figures S8A
reasoned that molecular mediators and signaling pathways and S8B). These findings agree with our previous report that pax-
used for BDNF-induced chemotaxis differ among neurons grown illin expressed in neurons grown on 0.1 kPa substrates does not
on microenvironments of varying elasticity. To test this hypothe- preferentially bind focal adhesion molecules (Chang et al., 2017).
sis, we pretreated neuronal cultures grown on 0.1 kPa hydrogels In contrast, pretreating 0.1 kPa cultures with the TrkB inhibitor
or coverslips with inhibitors of the focal adhesion kinase Src K252a abolished BDNF-induced drebrin phosphorylation
(PP2), the endocytosis kinase AAK1, or the growth cone turning (Figures S8A and S8B), confirming that BDNF signaling induces
signaling kinase (Song et al., 1997) PKA (KT5720) (Figures 4F and phosphorylation of paxillin-drebrin complexes.
4G), and measured the BDNF-directed growth cone turning. To determine whether BDNF-induced paxillin association re-
Indeed, although Src kinase activity was required for growth quires drebrin phosphorylation at serine 142, we transfected
cone turning on coverslips (Figure 4G), attraction of a growth N2a cells with YFP-tagged wild-type (WT) drebrin or a tagged
cone to BDNF on 0.1 kPa hydrogels was abolished by blocking phosphorylation-deficient drebrinS142A construct (DBNS142A)
either AAK1 or PKA activities but not Src kinase activity (Fig- and treated both groups with and without BDNF. Co-IP of cell ly-
ure 4F). We also evaluated preferential paxillin/drebrin co-local- sates with an anti-YFP antibody revealed increased levels of
ization on the side of growth cone facing BDNF stripes, and its paxillin and S142-phosphorylated WT drebrin in BDNF-treated
dependence on endocytic activity. Consistently, BDNF was cells (Figures S8C and S8D), while control and BDNF-treated
insufficient to promote paxillin/drebrin co-localization in growth cells transduced with the DBNS142A mutant showed comparable
cones moving on hydrogels in the presence of an AAK1 inhibitor paxillin association (Figures S8C and S8D). Because this residue
or of KT5720, resulting in uniform paxillin distribution across the enables microtubule coupling to F-actin in growth cone filopodia
growth cone (Figure 4F, co-localization index = 1). In contrast, (Worth et al., 2013), p-drebrinS142 likely contributes to attraction
neither directed growth cone turning nor paxillin/drebrin co- to BDNF stripes by growth cones on 0.1 kPa hydrogels. To
localization (Figure 4F) in hydrogel-grown growth cones was assess this possibility, we calculated the percentage of axons
altered by Src kinase inhibition. These findings indicate that pax- that moved forward on or off BDNF stripes coated on 0.1 kPa hy-
illin/drebrin distribution in neurons responds to BDNF stimuli in a drogels, assessing only neurons with somata located off the
matrix compliance-specific manner, independent of activity of stripes (Figures S8E and S8F). At 3 DIV, 70% of neurons ex-
Src kinase. pressing WT drebrin exhibited axon tips moving along the
BDNF-coated surface, while neurons expressing DBNS142A ex-
Paxillin-drebrin interaction and drebrinS142 hibited randomly distributed axon terminals (50% on and
phosphorylation coordinate BDNF-induced growth cone 50% off BDNF-coated stripes) (Figures S8E and S8F), suggest-
turning behavior on 0.1 kPa hydrogels ing reduced attraction to BDNF stripes.
To quantify in situ direct paxillin-drebrin interaction in the pres- Finally, we asked whether BDNF-induced growth cone turning
ence of BDNF, we used a proximal ligation assay (PLA) to assess required drebrin-paxillin interaction. We found that preferential
proximity (<40 nm apart) of paxillin and drebrin proteins at the turning of 0.1 kPa hydrogel-grown growth cones toward BDNF
growth cone T zone. In this assay, a change in the number of stripes (76% toward/on versus 24% off the stripes; Figures 5B
PLA puncta observed reflects paxillin-drebrin interaction after and 5C), as well as preferential accumulation of paxillin on the
bath application of BDNF. As shown in Figure 5A, PLA signal sides of growth cones facing BDNF stripes, was absent in dre-
density in growth cones of neurons grown on 0.1 kPa hydrogels brin KD neurons (53% toward/on versus 47% off the stripes;
was significantly greater following BDNF treatment relative to un- Figures 5B and 5C). Importantly, attraction of growth cones
treated samples, an effect eliminated by pretreatment with turning on 0.1 kPa hydrogels to BDNF stripes was abrogated
K252a, which inhibits the BDNF receptor TrkB. in neurons expressing a paxillin LIM3-4 deletion variant, which

8 Cell Reports 40, 111188, August 16, 2022


ll
Article OPEN ACCESS

Figure 5. BDNF-induced growth cone turning requires association of paxillin with drebrin
(A) BDNF promotes paxillin-drebrin interaction at the growth cone on 0.1 kPa gels. Left panel: schematic describing PLA analysis used to assess paxillin-drebrin
interaction. Middle panels: representative images of 2-DIV hippocampal neurons stained with phalloidin for actin (red in merge) plus PLA detection of paxillin and
drebrin antibody signals (green in merge) after 30 min of BDNF (20 nM) treatment, with or without 30 min of K252a (200 nM) pretreatment. Scale bars, 5 mm. Right
panel: histogram shows number of PLA puncta per 50 mm2 of growth cone area from all experiments (±SEM, n > 25 cells for each group from three independent
experiments; ****p < 0.0001 by Student’s unpaired t test). Growth cone area was defined by thresholding phalloidin-stained images and tracing growth cone out-
lines (dashed lines in top middle panels). Only puncta located inside or on the phalloidin-stained growth cone area were counted.
(B and C) BDNF-induced chemoattraction on 0.1 kPa hydrogels requires drebrin expression. Similar to Figure 4B, except that cells grown on BDNF-stripe-coated
0.1 kPa hydrogels were treated with scrambled siRNA (SC-siRNA) or drebrin siRNA (DBN-siRNA), and immunostained with drebrin and paxillin antibodies and
phalloidin for F-actin at 3 DIV, as indicated. Scale bars, 5 mm. Only neurons located off the stripe with neurite initiation off the stripe were counted.
(D) Quantitative assessment of paxillin asymmetry (as calculated by the formula shown at the top) showing that drebrin KD eliminates asymmetric paxillin
accumulation on the side of growth cones facing BDNF-coated stripes on 0.1 kPa hydrogels. Data represent mean ± SEM (>14 cells for each group from three
independent experiments; **p < 0.01 by t test).
(E–G) Similar to (B–D), except cells were transfected with plasmid encoding full-length (PXN_FL) or the LIM 3–4 domain deletion variant (PXN_DLIM3-4) of paxillin.
Data represent mean ± SEM (>10 cells for each group from three independent experiments; *p < 0.05 by t test). Scale bars, 5 mm.

Cell Reports 40, 111188, August 16, 2022 9


ll
OPEN ACCESS Article

(legend on next page)

10 Cell Reports 40, 111188, August 16, 2022


ll
Article OPEN ACCESS

cannot interact with drebrin (Figures 5E and 5F). Similarly, pref- lin-drebrin interaction, as neurons expressing paxillin DLIM3-4
erential paxillin/drebrin accumulation on the growth cone side did not exhibit BDNF-induced force generation (Figures 6A3
facing BDNF stripes was suppressed in neurons following and 6B3). Together, these findings indicate that paxillin in the
ectopic expression of the paxillin LIM3-4 deletion variant (Fig- growth cone links chemical guidance signals to generation of
ure 5G). Consistently, axons of neurons misexpressing a drebrin physical force, which then drives movement of axonal tips on
deletion mutant lacking the ADFH domain, a construct that soft environments.
cannot bind paxillin (Figures S4C and S4D), exhibited no prefer-
ence for growth on BDNF-coated stripes in 0.1 kPa cultures DISCUSSION
(45% on versus 55% off the stripes; Figures S4E and S4F). These
findings support the idea that paxillin-drebrin interaction medi- Axonal pathfinding in its most energy-efficient form is instructed
ates the effect of BDNF on growth cone motion. by the chemical and physical context of tissue environments
conducive to long-range neuronal targeting to designated brain
BDNF-elicited traction force requires paxillin and areas. Here, we assessed major force-generating components
drebrin expression and functional structures of axonal growth cones of hippocam-
Our observations indicate that paxillin-drebrin interaction at the pal neurons cultured on 0.1 kPa hydrogels. We observed fist-
growth cone T zone is facilitated by BDNF (Figure 5), which in like architectures of growth cones on 0.1 kPa hydrogels, strongly
turn elicits a pushing force in the growth cone against the resembling the typical morphology of rapidly extending axons
0.1 kPa substrate. To investigate this possibility, we used TFM in vivo or reported in three-dimensional (3D) cultures (Balgude
to examine changes in stress induced by growth cones on et al., 2001). This compact structure enhances the degree of
0.1 kPa hydrogels in response to BDNF stimuli. Bath application actin and microtubule engagement, as well as physical interac-
of 20 nM BDNF significantly enhanced stress generated by the tions between the T-zone factor drebrin and the accessory pro-
cell body and growth cone in neurons grown on 0.1 kPa hydro- tein paxillin. We found that direct drebrin-paxillin interaction is
gels (Figure 6A), an effect significantly blocked by K252a pre- dependent on the physical environment, since binding affinity
treatment (Figure 6A), supporting the idea that BDNF signaling of paxillin for drebrin is significantly greater in brain tissue and
is sufficient to generate neuronal mechanical force on a soft sub- on 0.1 kPa hydrogels than on stiffer 20 kPa hydrogels. Moreover,
strate. Finally, we asked whether BDNF-elicited neuronal force we found that BDNF stripes bound to 0.1 kPa hydrogel surfaces
contributes to growth cone motion on 0.1 kPa hydrogels, where exert a chemoattractive turning and force generation effect on
substrate adhesion is minimized. Since paxillin is critical for growth cones, actions that required paxillin and drebrin expres-
growth cone force generation (Figures 3B4 and 3B5) and turning sion and interaction but not the activity of Src kinase. These find-
behavior (Figure 5E) on 0.1 kPa hydrogels, we postulated that ings demonstrate that the force generation mechanism of neu-
paxillin functions in the force generation response to BDNF. To rons growing in soft substrates differs from that of neurons
confirm this possibility, we performed paxillin KD in neurons grown on coverslips and is mediated by paxillin-drebrin interac-
and assessed growth cone force generation capacity. Growth tion at the T zone of the axonal growth cone (Figure 6C).
cones of control neurons generated a differential force, with Several in vitro culture systems—such as synthetic hydrogel
stronger stress emanating from the side facing toward BDNF platforms, microfluidic systems, paper-based culture platforms
stripes, on 0.1 kPa hydrogels (Figure 6B). In contrast, paxillin (Ng et al., 2017; Pelham and Wang, 1997; Santos et al., 2020;
or drebrin KD neurons exerted much less force on 0.1 kPa hydro- Taylor et al., 2005), or stem cell-based brain organoids (Gopalak-
gels and their growth cones facing toward BDNF stripes ex- rishnan, 2019)—have been engineered to recapitulate various
hibited no preferential activity (Figure 6B), suggesting that paxil- stages of neuronal/brain development. By increasing the elastic-
lin expression is required for BDNF-induced force generation by ity of PA hydrogels to a range resembling the stiffness of
neurons on 0.1 kPa hydrogels. This activity likely requires paxil- brain tissue (0.1–1 kPa), and with proper surface coating with

Figure 6. BDNF is sufficient to induce asymmetric force generation by growth cones of neurons growing on 0.1 kPa hydrogels via a
mechanism mediated by paxillin-drebrin interaction
(A) BDNF signaling increases force exerted by growth cones of neurons grown on 0.1 kPa hydrogels. (A1) Representative TFM images of vector force fields
plotted over magnitudes of the displacement (presented as pseudocolor maps in a linear scale) for 2-DIV hippocampal neurons growing on 0.1 kPa hydrogels and
subjected (or not) to bath application of 20 ng/mL BDNF and/or pretreatment of the TrkB inhibitor K252a (200 nM). (A2) Histograms summarizing averaged stress
exerted (±SEM, n = 7–15 cells for each set of experiments; *p < 0.05; **p < 0.01; ns, not significant by t test) by the entire image field (Whole Field), growth cone
(Growth Cone), and cell body (Soma). (A3) Similar to (A2), except cells were transfected with plasmid encoding full-length (PXN_FL) or the LIM 3–4 domain deletion
variant (PXN_DLIM3-4) of paxillin. Histograms summarizing averaged stress exerted (±SEM, n = 21 cells; ns, not significant by t test) by the entire image field
(Whole Field), growth cone (Growth Cone), and cell body (Soma).
(B) Asymmetric exposure to BDNF biases the force distribution exerted by growth cones on 0.1 kPa hydrogels. (B1) Representative TFM images of growth cones
approaching BDNF- or control BSA-coated stripes (blue rectangle) on 0.1 kPa hydrogels. Cells were treated with scrambled siRNA (SC-siRNA), paxillin siRNA
(PXN-siRNA), or drebrin siRNA (DBN-siRNA). Scale bars, 5 mm. (B2 and B3) Quantitative measurement of force-field asymmetry in a 2.5 mm 3 2.5 mm area (dashed
boxes in B2) covering half of a growth cone facing toward versus away (stoward/saway) from BSA- or BDNF-coated stripes and pretreated with or without 200 nM
K252a, as indicated. Data represent mean ± SEM (n = 7–20 cells for each group from three independent experiments; *p < 0.05; **p < 0.01 by t test). Only neurons
with growth cones located near the stripe boundary were counted.
(C) Schematic illustration showing organization of growth cones on soft substrates and asymmetric distribution of T-zone factors associated with paxillin that
enable growth cone force generation toward a BDNF gradient, in a Src kinase-independent manner.

Cell Reports 40, 111188, August 16, 2022 11


ll
OPEN ACCESS Article

positively charged macromolecules, hippocampal neurons flattens lamella, and obstructs microtubules protruding into the P
grown on hydrogels can attain structural properties of growth domain (Luo, 2000; Santos et al., 2020). Thus, neurite overexten-
cones undergoing active extension, as seen in classic ‘‘open- sion observed in coverslip cultures after NMII inhibition can be
book’’ preparations or by DiI tracking of spinal commissural attributed to: (1) reduction in RhoA/Rock-activated, NMII-gener-
axons ex vivo (Bovolenta and Dodd, 1990; Stoeckli and Land- ated contractile forces acting on cortical actin along the axonal
messer, 1995). Inverse durotaxis has been shown for growth shaft; and (2) relaxing the NMII-generated actin arc barrier that
cones on PA hydrogels displaying a gradient of rigidity, with restricts microtubule protrusion (Bradke and Dotti, 1999; Dupraz
axon bundles of retinal ganglion cells (RGCs) preferentially ex- et al., 2019; Santos et al., 2020). The same NMII inhibition ac-
tending toward the softer side of the gel (Koser et al., 2016). counts for lack of neurite overextension on 0.1 kPa PA-gel cul-
This feature has been demonstrated in vivo for zebrafish RGC tures, in agreement with previously reported data in 0.2 kPa
axons extending along the optic tract (Koser et al., 2016). collagen 3D cultures (Bradke and Dotti, 1999). We argue that
Here, we replicated the stiffness of brain environment using growth cone mechanics of neurons grown on 2D 0.1 kPa sub-
0.1 kPa PA hydrogels with minimal surface coating of laminin strates resemble those seen in 3D soft matrices, in which there
and showed that the intracellular T-zone components drebrin is a relatively low level of integrin-actin linkage (adhesion clutch
and paxillin, in the context of a particular cytoskeletal architec- engagement), endowing growth cones with an amoeba-like
ture, function like a counter-shaft to transmit force for bound movement independent of FAK-Src activity. Accordingly, NMII-
chemical guidance cues. mediated contractility may ensure that growing axons of
Despite the fact that most cell types grown on stiff (>10 kPa) 0.1 kPa neurons form a compact actomyosin arc to gather the
substrates use integrin-dependent pathways for directed cell ends of microtubule bundles and allow growing axons to
migration (Kerstein et al., 2017; Lo et al., 2000; Robles and Go- press/push aside obstacles in their path. Although we applied
mez, 2006), other cell types such as neurons and tumor cells the same coating solution (a mixture of poly-L-lysine [PLL] and
adapt to low-rigidity environments and do not rely on mature laminin) to coverslips and 0.1 kPa PA gels, resulting surface
focal adhesion for migration in response to gradients of bound PLL and laminin levels may differ. Thus, different adhesion sub-
chemical cues (Tyler, 2012). Our findings reveal an alternative strates and elasticity could have altered the NMII distribution
type of cell mechanics specifically evoked on compliant environ- and/or function (Ketschek et al., 2007; Turney et al., 2016). We
ments, one that exhibits three features enabling growth cone also noted that a snapshot of growth cone zip-zap behavior
advancement and navigation. First, the compact architecture does not accurately indicate the direction in which the growth
of the growth cone in soft substrates means greater mechanical cone is going on a uniform substrate. However, while the growth
proximity of the T zone and C and P domains. Second, the cone is exposed to a gradient of attractants or moves up a
T-zone molecule drebrin selectively associates with paxillin on gradient, it does exhibit a longer continuous movement and a
soft surfaces to generate stress or pressing force on the sub- more significant turning angle toward attractants (Mai et al.,
strate. Third, chemotaxis induced by bound neurotrophic factors 2009; Ming et al., 1997). Since the attractant gradient (such as
on soft surfaces is independent of that exerted by the integrin- BDNF stripes) biases growth cone behavior, comparison of the
FAK-Src signaling. Conversely, the primary model of growth sides of the growth cone facing both toward and away from
cone mechanics in soft substrates does not follow the actin- the BDNF stripe allows us to analyze potential asymmetric distri-
based treadmill model for the leading edge of migrating cells bution of T-zone molecules and force generation underlying
on rigid substrates (Forscher and Smith, 1988; Lowery and Van growth cone steering.
Vactor, 2009; Mitchison and Kirschner, 1988). Instead, neurons It is intriguing that compliant environments evoke systematic
grown on a soft substrate express extremely low levels, if any, changes in axonal growth cones, not only in terms of structure
of the focal adhesion protein vinculin relative to neurons grown and protein localization but in signaling required for force exer-
on stiffer substrates (Chang et al., 2017), implying that the integ- tion, which are all distinct from those observed in neurons grown
rin-dependent pathway is dispensable in compliant physical on rigid substrates. These differences reflect the low-adhesive,
contexts in terms of directing growth cone turning or force high-surface-tension characteristics of neuronal mechanics in
generation. brain. Here, we present a potential molecular mechanism that
The force exerted by rat cortical neurons (in the range of 0.5–1 underlies how axonal tips in the brain orient their movement
pN/mm2) is exceedingly small compared with that exerted by and force toward trace amounts of a chemical guidance cue,
many other cell types, such as fibroblasts, which exert force in namely through low-rigidity and specific interaction between
the range of 10 nN/mm2 (Dembo and Wang, 1999). Our findings neuronal paxillin and drebrin. Looking forward, our proposed
indicate that growth cone attraction on soft surfaces requires model, which integrates structural changes in axonal tips with
drebrin and paxillin, an association that leverages the actin- molecular interaction of T-zone factors, describes a synergistic
microtubule coordination/interface with NMII to induce force mechanism that drives microtubule-to-actin engagement and
generation in the T zone. Accordingly, we postulate that upon force generation enabling growth cone movement in the context
contact with soft surfaces, neurons undergo a structural and mo- of an extremely soft environment.
lecular transformation to maximize efficiency and growth cone
sensitivity and movement. Neurons grown on stiff substrates in- Limitations of the study
crease levels of RhoA/Rock and form point contacts at filopodia Acquiring high-resolution F-actin images of growth cones on
tips (Chang et al., 2017; Woo and Gomez, 2006). In that case, our 0.1 kPa PA hydrogels (200 mm thick and watery) using
NMII located at the T-zone actin arc mediates actin contraction, stimulated emission depletion microscopy remains technically

12 Cell Reports 40, 111188, August 16, 2022


ll
Article OPEN ACCESS

challenging, possibly due to the inhomogeneous refractive index AUTHOR CONTRIBUTIONS


between the cell and scaffold.
C.C., C.-H.C., Y.C., and T.-Y.C. performed, analyzed, and validated biochem-
Although our findings focus on paxillin/drebrin and their integ-
istry experiments, immunofluorescence staining, plasmid construction, and
rin-independent regulation of neurons grown on soft substrates, hydrogel preparation, and co-wrote the manuscript. S.-W.C. and P.-L.C. per-
it is important to recognize that post-translational modification formed the IUE experiment. S.-Y.L. performed primary neuronal cultures.
(either phosphorylation or acetylation) of paxillin can be induced Y.-C.T. and B.-C.C. supervised, performed, analyzed, and validated expan-
by FAK or Src activity and is known to be essential for movement sion microscopy. H.-L.T. supervised TFM analysis. P.-L.C. supervised,
of cells/growth cones on rigid surfaces coated with high concen- conceptualized, and co-wrote the manuscript. All authors read and contrib-
uted to the manuscript.
trations of positively charged PLL (Koch et al., 2012; Myers and
Gomez, 2011). Also, apart from paxillin’s association with T-zone
factors, other paxillin functions mediated by various interacting DECLARATION OF INTERESTS
partners—such as receptor-mediated endocytosis, trafficking,
The authors declare no competing interests.
or nuclear translocation—could play important roles in neuronal
differentiation in a compliant microenvironment (Chang et al.,
INCLUSION AND DIVERSITY
2017; Dong et al., 2009; Dubois et al., 2017; Sathe et al., 2016;
Xu et al., 2019). In addition to growth cone dynamics, physical We worked to ensure sex balance in the selection of non-human subjects. One
environmental factors may also modulate strength and efficacy or more of the authors of this paper self-identifies as a member of the LGBTQ+
of cell-cell contacts along axons and dendrites, providing community. One or more of the authors of this paper received support from a
another mechanism by which neuronal maturation can be sys- program designed to increase minority representation in science. While citing
tematically controlled. references scientifically relevant for this work, we also actively worked to pro-
mote gender balance in our reference list.

STAR+METHODS Received: September 12, 2021


Revised: March 23, 2022
Detailed methods are provided in the online version of this paper Accepted: July 20, 2022
Published: August 16, 2022
and include the following:

d KEY RESOURCES TABLE REFERENCES


d RESOURCE AVAILABILITY
Balgude, A.P., Yu, X., Szymanski, A., and Bellamkonda, R.V. (2001). Agarose
B Lead contact
gel stiffness determines rate of DRG neurite extension in 3D cultures. Bioma-
B Materials availability terials 22, 1077–1084.
B Data and code availability
Betz, T., Koch, D., Lu, Y.B., Franze, K., and Käs, J.A. (2011). Growth cones as
d EXPERIMENTAL MODEL AND SUBJECT DETAILS soft and weak force generators. Proc. Natl. Acad. Sci. USA 108, 13420–13425.
d METHOD DETAILS Bovolenta, P., and Dodd, J. (1990). Guidance of commissural growth cones at
B Plasmids, antibodies, and inhibitors the floor plate in embryonic rat spinal cord. Development 109, 435–447.
B Cell culture, protein lysate preparation, and immuno-
Bradke, F., and Dotti, C.G. (1999). The role of local actin instability in axon for-
staining mation. Science 283, 1931–1934.
B Expression of GST- and His-tagged proteins, and Burnette, D.T., Ji, L., Schaefer, A.W., Medeiros, N.A., Danuser, G., and For-
in vitro and in situ binding assays scher, P. (2008). Myosin II activity facilitates microtubule bundling in the
B Polyacrylamide gel preparation neuronal growth cone neck. Dev. Cell 15, 163–169.
B In utero electroporation Chang, T.Y., Chen, C., Lee, M., Chang, Y.C., Lu, C.H., Lu, S.T., Wang, D.Y.,
B Microfabrication and substrate patterning Wang, A., Guo, C.L., and Cheng, P.L. (2017). Paxillin facilitates timely neurite
B Expansion microscopy initiation on soft-substrate environments by interacting with the endocytic ma-
B Traction force microscopy, image acquisition, and im- chinery. Elife 6, e31101.

age processing Dembo, M., and Wang, Y.L. (1999). Stresses at the cell-to-substrate interface
d QUANTIFICATION AND STATISTICAL ANALYSIS during locomotion of fibroblasts. Biophys. J. 76, 2307–2316.
Dent, E.W., and Gertler, F.B. (2003). Cytoskeletal dynamics and transport in
growth cone motility and axon guidance. Neuron 40, 209–227.
SUPPLEMENTAL INFORMATION
Dong, J.M., Lau, L.S., Ng, Y.W., Lim, L., and Manser, E. (2009). Paxillin nu-
clear-cytoplasmic localization is regulated by phosphorylation of the LD4
Supplemental information can be found online at https://doi.org/10.1016/j.
motif: evidence that nuclear paxillin promotes cell proliferation. Biochem. J.
celrep.2022.111188.
418, 173–184.
Dotti, C.G., Sullivan, C.A., and Banker, G.A. (1988). The establishment of po-
ACKNOWLEDGMENTS
larity by hippocampal neurons in culture. J. Neurosci. 8, 1454–1468.

We thank Mu-Ming Poo and Hwai-Jong Cheng for discussions and critical Dubois, F., Alpha, K., and Turner, C.E. (2017). Paxillin regulates cell polariza-
reading of the manuscript. We thank the IMB Imaging Core Facilities (Institute tion and anterograde vesicle trafficking during cell migration. Mol. Biol. Cell
of Molecular Biology, Academia Sinica) for SIM imaging and TFM analysis. 28, 3815–3831.
This work was supported in part by a grant (MOST 110-2628-B-001-028-) Dun, X.P., Bandeira de Lima, T., Allen, J., Geraldo, S., Gordon-Weeks, P., and
from the Ministry of Science and Technology of Taiwan, and Career Develop- Chilton, J.K. (2012). Drebrin controls neuronal migration through the formation
ment Award (AS-CDA-107-L04) from Academia Sinica, Taiwan. and alignment of the leading process. Mol. Cell. Neurosci. 49, 341–350.

Cell Reports 40, 111188, August 16, 2022 13


ll
OPEN ACCESS Article
Dupraz, S., Hilton, B.J., Husch, A., Santos, T.E., Coles, C.H., Stern, S., Brake- Merriam, E.B., Millette, M., Lumbard, D.C., Saengsawang, W., Fothergill, T.,
busch, C., and Bradke, F. (2019). RhoA controls axon extension independent Hu, X., Ferhat, L., and Dent, E.W. (2013). Synaptic regulation of microtubule
of Specification in the developing brain. Curr. Biol. 29, 3874–3886.e9. dynamics in dendritic spines by calcium, F-actin, and drebrin. J. Neurosci.
Forscher, P., and Smith, S.J. (1988). Actions of cytochalasins on the organiza- 33, 16471–16482.
tion of actin filaments and microtubules in a neuronal growth cone. J. Cell Biol. Ming, G.L., Song, H.J., Berninger, B., Holt, C.E., Tessier-Lavigne, M., and Poo,
107, 1505–1516. M.M. (1997). cAMP-dependent growth cone guidance by netrin-1. Neuron 19,
Gallo, G., Lefcort, F.B., and Letourneau, P.C. (1997). The trkA receptor medi- 1225–1235.
ates growth cone turning toward a localized source of nerve growth factor. Mitchison, T., and Kirschner, M. (1988). Cytoskeletal dynamics and nerve
J. Neurosci. 17, 5445–5454. growth. Neuron 1, 761–772.
Geraldo, S., and Gordon-Weeks, P.R. (2009). Cytoskeletal dynamics in Mizui, T., Kojima, N., Yamazaki, H., Katayama, M., Hanamura, K., and Shirao,
growth-cone steering. J. Cell Sci. 122, 3595–3604. T. (2009). Drebrin E is involved in the regulation of axonal growth through actin-
myosin interactions. J. Neurochem. 109, 611–622.
Geraldo, S., Khanzada, U.K., Parsons, M., Chilton, J.K., and Gordon-Weeks,
P.R. (2008). Targeting of the F-actin-binding protein drebrin by the microtubule Mizui, T., Takahashi, H., Sekino, Y., and Shirao, T. (2005). Overexpression of
plus-tip protein EB3 is required for neuritogenesis. Nat. Cell Biol. 10, 1181– drebrin A in immature neurons induces the accumulation of F-actin and
1189. PSD-95 into dendritic filopodia, and the formation of large abnormal protru-
sions. Mol. Cell. Neurosci. 30, 630–638.
Gopalakrishnan, J. (2019). The Emergence of stem cell-based brain organoids:
trends and Challenges. Bioessays 41, e1900011. Myers, J.P., and Gomez, T.M. (2011). Focal adhesion kinase promotes integrin
adhesion dynamics necessary for chemotropic turning of nerve growth cones.
Hayashi, K., Ishikawa, R., Ye, L.H., He, X.L., Takata, K., Kohama, K., and
J. Neurosci. 31, 13585–13595.
Shirao, T. (1996). Modulatory role of drebrin on the cytoskeleton within den-
dritic spines in the rat cerebral cortex. J. Neurosci. 16, 7161–7170. Ng, S.S., Xiong, A., Nguyen, K., Masek, M., No, D.Y., Elazar, M., Shteyer, E.,
Winters, M.A., Voedisch, A., Shaw, K., et al. (2017). Long-term culture of hu-
Hsu, S., Thakar, R., Liepmann, D., and Li, S. (2005). Effects of shear stress on
man liver tissue with advanced hepatic functions. JCI Insight 2, 90853.
endothelial cell haptotaxis on micropatterned surfaces. Biochem. Biophys.
Res. Commun. 337, 401–409. Oberstar, J.V., Challacombe, J.F., Roche, F.K., and Letourneau, P.C. (1997).
Concentration-dependent stimulation and inhibition of growth cone behavior
Ishikawa, R., Hayashi, K., Shirao, T., Xue, Y., Takagi, T., Sasaki, Y., and Ko-
and neurite elongation by protein kinase inhibitors KT5926 and K-252a.
hama, K. (1994). Drebrin, a development-associated brain protein from rat em-
J. Neurobiol. 33, 161–171.
bryo, causes the dissociation of tropomyosin from actin filaments. J. Biol.
Pelham, R.J., Jr., and Wang, Y.l. (1997). Cell locomotion and focal adhesions
Chem. 269, 29928–29933.
are regulated by substrate flexibility. Proc. Natl. Acad. Sci. USA 94, 13661–
Katoh, K., Hammar, K., Smith, P.J., and Oldenbourg, R. (1999). Birefringence 13665.
imaging directly reveals architectural dynamics of filamentous actin in living
Pollard, T.D., and Borisy, G.G. (2003). Cellular motility driven by assembly and
growth cones. Mol. Biol. Cell 10, 197–210.
disassembly of actin filaments. Cell 112, 453–465.
Kerstein, P.C., Patel, K.M., and Gomez, T.M. (2017). Calpain-mediated Prote-
Robles, E., and Gomez, T.M. (2006). Focal adhesion kinase signaling at sites of
olysis of talin and FAK regulates adhesion dynamics necessary for axon guid-
integrin-mediated adhesion controls axon pathfinding. Nat. Neurosci. 9, 1274–
ance. J. Neurosci. 37, 1568–1580.
1283.
Ketschek, A.R., Jones, S.L., and Gallo, G. (2007). Axon extension in the fast
Rösner, H., Möller, W., Wassermann, T., Mihatsch, J., and Blum, M. (2007).
and slow lanes: substratum-dependent engagement of myosin II functions.
Attenuation of actinomyosinII contractile activity in growth cones accelerates
Dev. Neurobiol. 67, 1305–1320.
filopodia-guided and microtubule-based neurite elongation. Brain Res. 1176,
Koch, D., Rosoff, W.J., Jiang, J., Geller, H.M., and Urbach, J.S. (2012). 1–10.
Strength in the periphery: growth cone biomechanics and substrate rigidity
Rossner, M., and O’Donnell, R. (2004). What’s in a picture? The temptation of
response in peripheral and central nervous system neurons. Biophys. J. 102,
image manipulation. J. Cell Biol. 164, 11–13.
452–460.
Saito, T., and Nakatsuji, N. (2001). Efficient gene transfer into the embryonic
Kolodkin, A.L., and Tessier-Lavigne, M. (2011). Mechanisms and molecules of
mouse brain using in vivo electroporation. Dev. Biol. 240, 237–246.
neuronal wiring: a primer. Cold Spring Harb. Perspect. Biol. 3, a001727.
Santos, T.E., Schaffran, B., Broguière, N., Meyn, L., Zenobi-Wong, M., and
Koser, D.E., Thompson, A.J., Foster, S.K., Dwivedy, A., Pillai, E.K., Sheridan, Bradke, F. (2020). Axon growth of CNS neurons in three Dimensions is amoe-
G.K., Svoboda, H., Viana, M., Costa, L.d.F., Guck, J., et al. (2016). Mechano- boid and independent of adhesions. Cell Rep. 32, 107907.
sensing is critical for axon growth in the developing brain. Nat. Neurosci. 19,
Sathe, A.R., Shivashankar, G.V., and Sheetz, M.P. (2016). Nuclear transport of
1592–1598.
paxillin depends on focal adhesion dynamics and FAT domains. J. Cell Sci.
Laukaitis, C.M., Webb, D.J., Donais, K., and Horwitz, A.F. (2001). Differential 129, 1981–1988.
dynamics of alpha 5 integrin, paxillin, and alpha-actinin during formation and
Schneider, C.A., Rasband, W.S., and Eliceiri, K.W. (2012). NIH Image to Im-
disassembly of adhesions in migrating cells. J. Cell Biol. 153, 1427–1440.
ageJ: 25 years of image analysis. Nat. Methods 9, 671–675.
Le Clainche, C., and Carlier, M.F. (2008). Regulation of actin assembly associ-
Shin, E.Y., Lee, C.S., Yun, C.Y., Won, S.Y., Kim, H.K., Lee, Y.H., Kwak, S.J.,
ated with protrusion and adhesion in cell migration. Physiol. Rev. 88, 489–513.
and Kim, E.G. (2014). Non-muscle myosin II regulates neuronal actin dynamics
Lo, C.M., Wang, H.B., Dembo, M., and Wang, Y.L. (2000). Cell movement is by interacting with guanine nucleotide exchange factors. PLoS One 9, e95212.
guided by the rigidity of the substrate. Biophys. J. 79, 144–152.
Shirao, T. (1995). The roles of microfilament-associated proteins, drebrins, in
Lowery, L.A., and Van Vactor, D. (2009). The trip of the tip: understanding the brain morphogenesis: a review. J. Biochem. 117, 231–236.
growth cone machinery. Nat. Rev. Mol. Cell Biol. 10, 332–343.
Shirao, T., Hanamura, K., Koganezawa, N., Ishizuka, Y., Yamazaki, H., and Se-
Luo, L. (2000). Rho GTPases in neuronal morphogenesis. Nat. Rev. Neurosci. kino, Y. (2017). The role of drebrin in neurons. J. Neurochem. 141, 819–834.
1, 173–180. Shirao, T., and Obata, K. (1985). Two acidic proteins associated with brain
Mai, J., Fok, L., Gao, H., Zhang, X., and Poo, M.M. (2009). Axon initiation and development in chick embryo. J. Neurochem. 44, 1210–1216.
growth cone turning on bound protein gradients. J. Neurosci. 29, 7450–7458. Shirao, T., and Obata, K. (1986). Immunochemical homology of 3 developmen-
Medeiros, N.A., Burnette, D.T., and Forscher, P. (2006). Myosin II functions in tally regulated brain proteins and their developmental change in neuronal dis-
actin-bundle turnover in neuronal growth cones. Nat. Cell Biol. 8, 215–226. tribution. Brain Res. 394, 233–244.

14 Cell Reports 40, 111188, August 16, 2022


ll
Article OPEN ACCESS

Song, H.J., Ming, G.L., and Poo, M.M. (1997). cAMP-induced switching in Tseng, Q., Duchemin-Pelletier, E., Deshiere, A., Balland, M., Guillou, H., Filhol,
turning direction of nerve growth cones. Nature 388, 275–279. O., and Théry, M. (2012). Spatial organization of the extracellular matrix regu-
Stoeckli, E.T., and Landmesser, L.T. (1995). Axonin-1, Nr-CAM, and Ng-CAM lates cell-cell junction positioning. Proc. Natl. Acad. Sci. USA 109, 1506–1511.
play different roles in the in vivo guidance of chick commissural neurons.
Turney, S.G., Ahmed, M., Chandrasekar, I., Wysolmerski, R.B., Goeckeler,
Neuron 14, 1165–1179.
Z.M., Rioux, R.M., Whitesides, G.M., and Bridgman, P.C. (2016). Nerve growth
Takahashi, H., Sekino, Y., Tanaka, S., Mizui, T., Kishi, S., and Shirao, T. (2003). factor stimulates axon outgrowth through negative regulation of growth cone
Drebrin-dependent actin clustering in dendritic filopodia governs synaptic tar- actomyosin restraint of microtubule advance. Mol. Biol. Cell 27, 500–517.
geting of postsynaptic density-95 and dendritic spine morphogenesis.
J. Neurosci. 23, 6586–6595. Tyler, W.J. (2012). The mechanobiology of brain function. Nat. Rev. Neurosci.
13, 867–878.
Taylor, A.M., Blurton-Jones, M., Rhee, S.W., Cribbs, D.H., Cotman, C.W., and
Jeon, N.L. (2005). A microfluidic culture platform for CNS axonal injury, regen- Woo, S., and Gomez, T.M. (2006). Rac1 and RhoA promote neurite outgrowth
eration and transport. Nat. Methods 2, 599–605. through formation and stabilization of growth cone point contacts. J. Neurosci.
Trivedi, N., Stabley, D.R., Cain, B., Howell, D., Laumonnerie, C., Ramahi, J.S., 26, 1418–1428.
Temirov, J., Kerekes, R.A., Gordon-Weeks, P.R., and Solecki, D.J. (2017). Dre-
Worth, D.C., Daly, C.N., Geraldo, S., Oozeer, F., and Gordon-Weeks, P.R.
brin-mediated microtubule-actomyosin coupling steers cerebellar granule
(2013). Drebrin contains a cryptic F-actin-bundling activity regulated by
neuron nucleokinesis and migration pathway selection. Nat. Commun. 8,
Cdk5 phosphorylation. J. Cell Biol. 202, 793–806. https://doi.org/10.1083/
14484.
jcb.201303005.
Tsai, Y.C., Tang, W.C., Low, C.S.L., Liu, Y.T., Wu, J.S., Lee, P.Y., Chen, L.Q.,
Lin, Y.L., Kanchanawong, P., Gao, L., and Chen, B.C. (2020). Rapid high res- Xu, W., Gulvady, A.C., Goreczny, G.J., Olson, E.C., and Turner, C.E. (2019).
olution 3D imaging of expanded biological specimens with lattice light sheet Paxillin-dependent regulation of apical-basal polarity in mammary gland
microscopy. Methods 174, 11–19. morphogenesis. Development 146.

Cell Reports 40, 111188, August 16, 2022 15


ll
OPEN ACCESS Article

STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER


Antibodies
anti-FLAG M2 Sigma-Aldrich Cat#F1804; RRID:AB_262044
chicken polyclonal bIII tubulin (AB9354) Merck Millipore Cat# AB9354; RRID:AB_570918
anti-Actin (MAB1501) Merck Millipore Cat# MAB1501; RRID:AB_2223041
anti-alpha Tubulin (ab89984) Abcam Cat# ab89984; RRID:AB_10672056
anti-non-muscle Myosin IIB [3H2] (ab684) Abcam Cat#ab684
anti-drebrin (ab11068) Abcam Cat# ab11068; RRID:AB_2230303
anti-paxillin clone 349 (612405) BD Biosciences Cat# 612405; RRID:AB_647289
Paxillin Polyclonal Antibodies( ThermoFisher Cat# PA5-34910;
Scientific/ Invitrogen RRID: AB_2552260
c-Myc Monoclonal Antibody (9E10) Invitrogen Cat#MA1-980; RRID: AB_558470
Piezo1 Antibody Novus Biologicals Cat#NBP2-10504
Piezo2 Antibody Novus Biologicals Cat#NBP1-79321; RRID:AB_11030107
TrkB Polyclonal Antibody ThermoFisher Scientific Cat# PA5-78405; RRID:AB_2736725
CD171 (L1CAM) Polyclonal Antibody ThermoFisher Scientific Cat# PA5-85876; RRID:AB_2802677
YAP Antibody (63.7) Santa Cruz Biotechnology Cat# sc-101199; RRID:AB_1131430
Anti-Integrin b1 Antibody, activated, clone HUTS-4 Millipore Cat# MAB2079Z; RRID:AB_2233964
Talin (C-20) Santa Cruz Biotechnology Cat# sc-7534; RRID:AB_661610
Anti-GFP/YFP antibody (ab6556) Abcam Cat# ab6556;
RRID: AB_305564
Anti-phospho Drebrin (Ser142), clone 3C14 Antibody Millipore Cat#MABN833
Bacterial and virus strains
pLenti-C-mGFP-PXN OriGeneTechnologies Cat#PS100071
pGFP-C-shLenti shRNA Vector OriGeneTechnologies Cat#TR30023
ECOS101 DH5a competent cell Yeastern Biotech Co. Cat#FYE678
Biological samples
Sprague Dawley timed-pregnant rats BioLASCO N/A
Chemicals, peptides, and recombinant proteins
Recombinant Human/Murine/Rat BDNF PeproTech Cat#450-02
BSA Alexa Fluor 647 conjugate Invitrogen Cat#A34785
Forskolin, 7-Deacetyl-7-[O-(N-methylpiperazino)- Calbiochem Cat#344273
g-butyryl]-, Dihydrochloride
KT5720 Calbiochem Cat#420320
Pim1/AKK1-IN-1, also named LKB1/AAK1 dual inhibitor MedChemExpress Cat#HY-10371
PP2 Abcam Cat#ab120308
PP3 Abcam Cat#ab120617
K252a Abcam Cat#ab120419
Gem21 NeuroPlexTM GEMINI Bio-products Cat#400-160
poly-L-Lysine Sigma Aldrich Cat#P2636-1G
laminin Corning Cat#354232
SYLGARD 184 SILICONE ELASTOMER KIT Dow Corning Material#1673921
N0 -N0 -methylenebisacrylamide solution Sigma Aldrich Cat#M1533
Acrylamide solution Sigma Aldrich Cat#A4058
(3-aminopropyl)triethoxysilan Sigma Aldrich Cat#A3638
(Continued on next page)

e1 Cell Reports 40, 111188, August 16, 2022


ll
Article OPEN ACCESS

Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Critical commercial assays
DuolinkR In Situ Detection Reagents Red, Anti-Rabbit MINUS, Sigma Aldrich Cat#DUO92008, DUO92005, DUO92001
Anti-Mouse PLUS
Deposited data
Mendeley DATA https://data.mendeley.com https://doi.org/10.17632/cpb3skbrhn.4
Experimental models: Cell lines
Human Embryonic Kidney 293T cells ATCC Cat# CRL-3216; RRID:CVCL_0063
Oligonucleotides
paxillin siRNA C: 50 -GAUGAGCAGUCCACAGCGA -30 This paper N/A
paxillin siRNA D: 50 -CAGCCUAGUGGUUCCAGAU -30 This paper N/A
drebrin siRNA #1: 50 -GCACGAAAUUUAAAACAUG-30 This paper N/A
drebrin siRNA #2: 50 -CCAACCUUCUUAAUUUCGA-30 This paper N/A
drebrin siRNA #3: 50 -CUUUCAGGCCACUUCGAGA-30 This paper N/A
drebrin siRNA #4: 50 -GCAGUGUGGAGGAUAUCGA-30 This paper N/A
Acell non-targeting Pool Dharmacon DAMD-001910-10-20
Recombinant DNA
Paxillin-pEGFP Laukaitis et al. (2001) RRID:Addgene_15233
drebrin-YFP Geraldo et al. (2008) RRID:Addgene_40359
S142A drebrin-YFP Geraldo et al. (2008) RRID:Addgene_58335
S142D drebrin-YFP Geraldo et al. (2008) RRID:Addgene_58336
drebrin-his Geraldo et al. (2008) RRID:Addgene_40363
GatewayTM pDONR/Zeo Vector and GatewayTM pDESTTM15 ThermoFisher Scientific Cat#11803-012, Cat#K37120, Cat# K37020
Vector, GatewayTM Cloning System
Software and algorithms
ImageJ Schneider et al. (2012) https://ImageJ.nih.gov/ij/
Imaris 9.3.1 (Bitplane) Oxford Instruments https://imaris.oxinst.com
Other
HisTrap column GE Healthcare Cat#17-5247-01
Glutathione Sepharose 4B GE Healthcare Cat#17-0756-01
square-wave electroporation generator BTX Inc. model ECM 830
carboxylate-modified FluoSpheresTM microspheres (0.2 mm, ThermoFisher Scientific Cat#F8805
blue fluorescing (365/415)

RESOURCE AVAILABILITY

Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Pei-Lin
Cheng (plcheng@imb.sinica.edu.tw).

Materials availability
All unique/stable reagents generated in this study are available from the lead contact with a completed materials transfer agreement.

Data and code availability


d Original western blot images have been deposited at Mendeley and are publicly available as of the date of publication. The DOI
is listed in the key resources table. Microscopy data reported in this paper will be shared by the lead contact upon request.
d This paper does not report original code
d Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

Cell Reports 40, 111188, August 16, 2022 e2


ll
OPEN ACCESS Article

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Hippocampal and cortical neurons were prepared from embryonic day 17.5 (E17.5) Sprague–Dawley (SD) rat embryos (Dotti et al.,
1988) and cultured in neurobasal medium supplemented with Gem21 NeuroPlexTM (GEMINI Bio-products, West Sacramento, CA)
on 0.1 kPa PA hydrogel substrate or glass coverslips coated with poly-L-lysine (Sigma Aldrich, St. Louis, MO) and laminin (Corning
Incorporated, Corning, NY). Timed (14 days) pregnant 6-month-old SD rats were housed in IMB animal facility under standardized
conditions (20–24 C room temperature, 55% humidity and a day/night cycle of 12:12 h, light turned on at 7 am every morning,
receiving pelleted standard diet and drinking water) util the day of experiment. Animal protocols were approved by the Academia
Sinica Institutional Animal Care and Utilization Committee. Human Embryonic Kidney 293T cells (HEK293T; ATCC Cat# CRL-
3216) for biochemical analysis were cultured in Dulbecco’s Modification of Eagle’s Medium (ThermoFisher Scientific, Waltham,
MA) supplemented with 10% fetal bovine serum (Biological Industries, Beit Haemek, Israel). Hippocampal and cortical neurons
were maintained in an incubator with 5% CO2 at 37 C for 5–72 h. Transfection was performed using Lipofectamine 2000
(ThermoFisher Scientific, Waltham, MA) according to the manufacturer’s instructions. Cortical cultures at high density (>1 x 106
per 42mm gel/coverslips) were used to obtain a sufficient number of cells for biochemical assays.

METHOD DETAILS

Plasmids, antibodies, and inhibitors


Paxillin full-length and truncated variants were subcloned from pLenti-C-mGFP-PXN (OriGene Technologies, Rockville, MD) into
plasmids pCAGGS-GFP by PCR-based methods. Paxillin-pEGFP plasmid (Plasmid # 15,233; (Laukaitis et al., 2001), drebrin-YFP
(Plasmid # 40,359; Geraldo et al., 2008), and drebrin-his (Plasmid # 40,363; Geraldo et al., 2008) were obtained from Addgene (Water-
town, MA). FLAG-tagged drebrin was generated by cloning a PCR product containing the coding sequence of drebrin-YFP into
pLenti-C-Myc-DDK-CIP4 (Chang et al., 2017). Paxillin siRNA pool, a mixture of two siRNAs (paxillin C: 50 -GAUGAGCAGUCCA
CAGCGA -30 , paxillin D: 50 -CAGCCUAGUGGUUCCAGAU -30 ), drebrin siRNA pool (drebrin siRNA #1: 50 -GCACGAAAUUUAAAA
CAUG-30 , drebrin siRNA #2: 50 -CCAACCUUCUUAAUUUCGA-30 , drebrin siRNA #3: 50 -CUUUCAGGCCACUUCGAGA-30 , and drebrin
siRNA #4: 50 -GCAGUGUGGAGGAUAUCGA-30 ), and a scramble siRNA (Acell non-targeting siRNA #1: 50 -UGGUUUACAUGUCGA
CUAA-30 ) were purchased from Dharmacon (Lafayette, CO). GST-fusion protein expression plasmid encoding full-length or trun-
cated versions of paxillin were cloned into GatewayTM pDONR/Zeo Vector and GatewayTM pDESTTM15 Vector (ThermoFisher
Scientific, Waltham, MA) using a GatewayTM Cloning System. All constructs were sequenced to verify sequence integrity. Sources
of antibodies and chemicals are as follows: mouse monoclonal FLAG M2 (F1804) was purchased from Sigma-Aldrich (St. Louis, MO);
chicken polyclonal bIII tubulin (AB9354) and anti-Actin (MAB1501) antibodies were from Merck Millipore (Burlington, MA); anti-drebrin
(ab11068), anti-alpha Tubulin (ab89984), and anti-non-muscle Myosin IIB (ab684) were from Abcam (Cambridge, MA); anti-paxillin
clone 349 (612,405) was from BD Biosciences (Franklin Lakes, NJ); mouse c-Myc tag monoclonal antibody, Alexa Fluor 647 phalloi-
din, and Alexa Fluor 647 phalloidin were from ThermoFisher Scientific (Waltham, MA); secondary antibodies conjugated with Alexa
Fluor 488, Alexa Fluor 546, or Alexa Fluor 633 were purchased from ThermoFisher Scientific; Forskolin and KT5720 were purchased
from Calbiochem (San Diego, CA); Pim1/AKK1-IN-1, also named LKB1/AAK1 dual inhibitor, was purchased from MedChemExpress
(Monmouth Junction, NJ); and PP2 and K252a were from Abcam (Cambridge, MA).

Cell culture, protein lysate preparation, and immunostaining


To prepare cell lysates, protein was extracted with M-PERTM (Mammalian Protein Extraction Reagent, ThermoFisher Scientific,
Waltham, MA) containing protease inhibitor cocktail (Roche, Basel, Switzerland) and the phosphate inhibitor PhosSTOP (Roche,
Basel, Switzerland). To prepare tissue lysates, we briefly rinsed the brain, heart, and liver of rat E17 embryos with HBSS
(ThermoFisher Scientific, Waltham, MA) and then lysed tissues in RIPA buffer (Sigma Aldrich, St. Louis, MO) with a protease inhibitor
cocktail and PhosSTOP.
For immunostaining, cultured hippocampal or cortical neurons were fixed for 12 min in 4% paraformaldehyde, permeabilized for
12 min in 0.3% Triton X-100, and blocked with 3% BSA for 1–2 h. Fixed cells were processed further for immunostaining according to
standard procedures and imaged with a confocal microscope (Carl Zeiss LSM700) equipped with a 403 water-immersion objective
(NA1.1; Carl Zeiss). Brightness and contrast adjustments of confocal images in figures were performed using ImageJ software (NIH;
(Schneider et al., 2012). For quantitative measurement of co-localization, we acquired images using a super-resolution structured
illumination microscopy (SR-SIM) system (Zeiss ELYRA PS.1) equipped with a Plan Apo 633 oil-immersion objective (NA 1.4,
Carl Zeiss). The axial step size was set to 125 nm. Surface rendering of three-dimensional images was performed using ZEN software
(Zeiss) and Imaris software (Bitplane) without Z-correction. Images were analyzed and processed for presentation in figures using
brightness and contrast adjustments in ImageJ software (NIH) and following the guidelines of (Rossner and Yamada, 2004).

Expression of GST- and His-tagged proteins, and in vitro and in situ binding assays
For protein expression and purification, GST- or His-tagged proteins were expressed in E. coli BL21(DE3) induced by 0.4 mM IPTG
overnight at 30 C in LB medium. Bacteria were lysed by sonication in short pulses of 15 s in lysis buffer [(50 mM Tris-HCl pH = 7.4,
50 mM NaCl, 5 mM DTT, 1 mM phenylmethylsulfonyl fluoride, 1% Triton X-100 with 1 mM EDTA, 1 mM dithiothreitol and protease

e3 Cell Reports 40, 111188, August 16, 2022


ll
Article OPEN ACCESS

inhibitors (Complete EDTA-free; Roche)]. Cell debris was removed by centrifugation at 9000 g for 10 min at 4 C. The resulting super-
natant was applied onto a Glutathione Sepharose 4B or HisTrap column (GE Healthcare). After washing, GST- and His-tagged pro-
teins were eluted in buffer containing 10 mM reduced glutathione and 300 mM imidazole, respectively. We added 1 M imidazole to
samples shortly after elution to prevent precipitation of His-tagged protein. Protein concentrations were measured using the Bio-Rad
DC Protein Assay. Protein purity was further assessed by fast protein liquid chromatography, followed by SDS-PAGE and Coomassie
blue staining.
For GST pull-down assays, cell lysate/His-tagged protein and GST fusion proteins were incubated with glutathione-agarose
beads. Complexes recovered from beads were resolved by SDS-PAGE and analyzed by Western blotting. In situ detection of protein
interaction at the growth cone was performed using Duolink PLA technology (Sigma Aldrich, St. Louis, MO), following the manufac-
turer’s instruction manual. PLA signals were visualized and quantified from confocal images.

Polyacrylamide gel preparation


Polyacrylamide gels with tunable mechanical stiffness were generated using a protocol slightly modified from that previously pub-
lished (Chang et al., 2017). Briefly, uniform polyacrylamide gels were fabricated in a three-layer assembly of 200 mm thickness. The
bottom layer was a hydrophilic amino-silanized coverslip prepared according to the following protocol. A thin film of sodium hydrox-
ide was allowed to form on the coverslips at approximately 90 C. The entire surface of each coverslip was then immersed in a suf-
ficient volume of (3-aminopropyl) triethoxysilane (Sigma-Aldrich, Saint Louis, MO) for 5 min. The (3-aminopropyl) triethoxysilane was
then completely rinsed off to prevent precipitation before adding 0.5% (v/v) glutaraldehyde (Sigma-Aldrich, Saint Louis, MO) in PBS
onto the silanized coverslips for 30 min. Fluids were removed by suction before the amino-silanized coverslips were air-dried and
sterilized with 70% ethanol for 16 h prior to gel preparation. The top layer was a laminin-coated coverslip prepared by sterilizing
an acid-washed coverslip with 70% ethanol and then coating it with 5 mg/mL of poly-L-Lysine (Sigma Aldrich, Saint Louis, MO)
and 0.08 mg/mL of laminin (Corning, NY) by absorption at 37 C for 1 h. For the middle layer of 0.1 kPa hydrogels, a prepolymer mixture
(75 mL 4% acrylamide, 25 mL 2%N0 -N0 -methylenebisacrylamide, and 900 mL deionized water) was prepared. Polymerization of the
mixture was carried out by adding 10 mL of 10% ammonium persulfate, 2 mL of TEMED (Bio-Rad Laboratories, Hercules, CA),
and sufficient deionized water to yield a final volume of 1000 mL. The resulting prepolymer-catalyst mixture was dropped onto hydro-
phobic amino-silanized coverslips. The three-layer assembly was formed by transferring pre-coated poly-L-Lysine and laminin cov-
erslips to the surfaces of the polyacrylamide gels during polymerization. After 30 min, the top layer was gently peeled off and washed
three times with HEPES solution to remove unreacted monomers and excess coating.

In utero electroporation
In utero electroporation followed previously described procedures (Saito and Nakatsuji, 2001), with minor modifications. Timed-
pregnant Sprague-Dawley rats were anesthetized at E17.5 with isoflurane, and the uterine horns were exposed by way of a laparot-
omy. Saline solution containing the expression plasmid of interest (2 mg/mL) together with the dye Fast Green (0.3 mg/mL; Sigma-
Aldrich, Saint Louis, MO) was injected (1–2 mL) through the uterine wall into one of the lateral ventricles of the embryos. The embryo’s
head was electroporated by tweezer-type circular electrodes across the uterus wall, and five electrical pulses (50 V, 50 ms duration at
100 ms intervals) were delivered with a square-wave electroporation generator (model ECM 830, BTX Inc.). Uterine horns were then
returned to the abdominal cavity, the wall and skin were sutured, and embryos continued normal development. Control embryos
were electroporated with the mScarlet construct together with the GFP construct (1:2 ratio), and experimental embryos were electro-
porated with the mScarlet construct (Kindly provided by Dr. Shu-Ling Chu, ICOB, Academia Sinica) together with drebrin shRNA
(#1- #4), or control scramble-shRNA construct. Control and experimental cortices (at P3 or P16) were obtained from the same litter
and the injections were always made into the left and right ventricles, respectively, for later identification. Maximum intensities of the
z-projection of tile images (20 mm thickness) were attained from spinning-disk confocal microscopy (Zeiss). Animal protocols were
approved by the Animal Care and Use Committee of Academia Sinica.

Microfabrication and substrate patterning


Microfabrication and substrate coating methods followed those previously described (Chang et al., 2017; Hsu et al., 2005). Briefly,
the poly(dimethylsiloxane) (PDMS) cuboids used to generate microchannels were prepared from Sylgard 184 base and curing agent
(Dow Corning, Midland, MI). These were polymerized on a silicon wafer etched with parallel stripes (50 mm width each) spaced
100 mm apart. Solution containing substrate factors was filled into the microchannels formed by placing the PDMS cuboids over
the poly-L-lysine-coated glass coverslip, and overnight incubation allowed the substrate factor to be coated onto the coverslip. Sub-
strate solutions were prepared with 0.5 ng/mL fluorescently-conjugated BDNF. In all coating solutions, 5 mg/mL of fluorescently-con-
jugated BSA was added as a marker of stripes.

Expansion microscopy
Lattice light sheet microscopy and sample preparation for expanded neuronal cultures were slightly modified from a previously re-
ported protocol (Tsai et al., 2020). Briefly, 0.1 kPa polyacrylamide gels were soaked in ExM monomer solution [1 3 PBS, 2 M NaCl,
8.625% (w/w) sodium acrylate, 2.5% (w/w) acrylamide, 0.15% (w/w) N,N0 -methylenebisacrylamide) at 4 C overnight. Full gelation
was achieved further by adding freshly prepared ExM monomer solution plus ammonium persulfate and tetramethyl ethylenediamine

Cell Reports 40, 111188, August 16, 2022 e4


ll
OPEN ACCESS Article

[up to 0.2% (w/w) each] onto the sample, incubated at room temperature for 20 min followed by 1 h of incubation at 37 C. The LLS
microscopy configuration and image processing were performed as described in (Tsai et al., 2020), with optical resolution in x, y, z =
1024 3 1024 3 201, voxel size in x, y, z = 0.103 mm 3 0.103 mm x 3 mm, field of view = 1024 3 0.103 mm = 105.472 mm.

Traction force microscopy, image acquisition, and image processing


Hippocampal neurons were grown on 0.1 kPa PA hydrogels embedded with carboxylate-modified FluoSpheres microspheres
(0.2 mm, blue fluorescing (365/415), ThermoFisher Scientific, Waltham, MA) for traction force microscopy (TFM) experiments. The
concentration of fluorescent beads is 4% of the hydrogel precursor mixture. A time series of z stack images [300 two-plane (2 mm
axial step) stacks over 10 min] was acquired at 0.5 Hz by DeltaVision Core Deconvolution Microscopy (General Electric, Boston,
MA) using an Olympus IX71 microscope and a 60x oil-immersion objective (NA 1.42, Olympus). Analysis of TFM images was con-
ducted as previously described (Tseng et al., 2012). The bead displacement field was calculated and visualized using the ImageJ
plugin for iterative particle image velocimetry (PIV), followed by the Fourier transform traction cytometry (FTTC) plugin to reconstruct
the force vector field.

QUANTIFICATION AND STATISTICAL ANALYSIS

Statistical analysis was performed by one-way ANOVA or unpaired two-tailed Student’s t-test. Statistical significance was deter-
mined as p < 0.05. Statistical analysis was conducted using GraphPad Prism 9 (GraphPad, San Diego, CA).

e5 Cell Reports 40, 111188, August 16, 2022

You might also like