Katelyn J. Hoff's research while affiliated with University of Colorado and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (8)


Dasatinib and trametinib treatment inhibits cell growth, induces apoptosis, and increases BIM expression. (A) Absolute quantification of the IC50 value for dasatinib with 100 nM trametinib in 23 thyroid cancer cell lines. Viability curves were measured across the cell lines using CellTiter-Glo Assay (Promega) or Sulforohodamine B Assay (SRB). Dashed line indicates 90 nM IC50 cutoff used to determine sensitivity and resistance to dasatinib and trametinib. Results shown are mean ± SEM. (B) Cleaved caspase 3/7 activity over the course of 3 days was measured in 3 sensitive cells (BCPAP, 8505C, and Cal62) and 2 resistant cells (T238 and CUTC60), treated alone or in combination with dasatinib and trametinib at 50 nM and 100 nM concentrations, respectively, and graphed as area under the curve. Results shown are mean ± SEM. (C) BIM expression obtained from RPPA data in sensitive and resistant cell lines. “ns” indicates not significant, **** p < 0.0001, * p < 0.01 (D) Immunoblot analysis of 8505C, T238, CUTC60 cells treated with indicated concentrations of dasatinib, trametinib, or the combination for 24 h and analyzed by Western blot for expression of BIM isoforms. EL = extra long, L = long, S = short. Alpha tubulin or beta actin were used as a loading control. Numbers below represent densitometric analysis normalized to loading control, followed by DMSO-treated cells. The uncropped blots are shown in Figure S6.
Growth inhibition by dasatinib and trametinib is dependent on BIM. (A) 8505C cells reverse transfected with nontargeting siRNA (siNT) or siRNA targeting BIM (siBIM). Cells were treated with 50 nM dasatinib and 100 nM trametinib for 24 h to induce BIM. (B) CellTiter Glo assays for viability were performed on siNT or siBIM cells. Cells were treated with indicated concentration of dasatinib with or without 100 nM trametinib for 72 h. Dashed line indicates 50% viability. Data was normalized to DMSO-treated control set to 100%. Results shown are mean ± SEM. IC50 values were calculated, and fold change is listed above the bars. (C) Immunoblot analysis of T238 cells overexpressing an empty vector (EV) or BIM plasmid. Cells were treated with 0, 0.5, 0.75, or 1 ng/mL of doxycycline upon plating. BCPAP cells served as a positive control for BIM induction and were treated with DMSO, 50 nM dasatinib, 100 nM trametinib, or the combination. Alpha tubulin was used as a loading control. Numbers below represent densitometric analysis normalized to loading control and 0 doxycycline- or DMSO-treated cells. (D,E) CellTiter Glo assay for viability was performed on parental, empty vector, or BIM-expressing cells. Cells were treated with indicated concentrations of dasatinib with or without 100 nM trametinib for 72 h. Data was normalized to DMSO-treated control set to 100%. Results shown are mean ± SEM. IC50 values were calculated and listed above the bars. The uncropped blots are shown in Figure S6.
Constitutively active AKT increases resistance to combined dasatinib and trametinib. (A) 8505C empty vector or Myr AKT cells were treated with indicated concentrations of dasatinib, trametinib, or the combination for 24 h and analyzed by Western blot for confirmation of pAKT. (B) CellTiter Glo assay for viability was performed on 8505C cells transfected with empty vector or myristoylated AKT (Myr AKT). Cells were treated with indicated doses of dasatinib and with 100 nM trametinib for 72 h. Area under the curve was calculated, and the results shown are mean ± SEM 2-way ANOVA * p < 0.05 ** p < 0.005. (C) Cleaved caspase 3/7 activity was measured after parental, empty vector, and Myr AKT cells were treated with DMSO, 100 nM trametinib, 50 nM dasatinib, or the combination for 24 h. Results shown are mean ± SEM. 2-way ANOVA test **** p < 0.003 * p < 0.03 (D) 8505C empty vector or Myr AKT cells were treated with indicated concentrations of dasatinib, trametinib, or the combination for 24 h and analyzed by Western blot for expression of BIM. Alpha tubulin was used as a loading control. Numbers below represent densitometric analysis normalized to loading control, followed by DMSO-treated cells. The uncropped blots are shown in Figure S6.
The addition of the AKT inhibitor MK2206 increases the efficacy of dasatinib and trametinib. (A,B) CellTiter Glo assay for viability was performed on cells treated with indicated doses of dasatinib, trametinib, and 1 μM of MK2206 for 72 h. Dashed line indicates 50% viability. Data were normalized to DMSO-treated control set to 100%. Results shown are mean ± SEM. IC50 values were calculated in GraphPad Prism ordinary one-way ANOVA test *** p < 0.006 * p < 0.06.
The mRNA ratio of MCL1:BCL-XL predicts sensitivity to the pan BCL-2 inhibitor ABT-263. (A) mRNA RPKM values for MCL1 and BCL-XL were obtained from the Cancer Cell Line Encyclopedia (CCLE). Ordinary one-way ANOVA test **** p < 0.0001, “ns” indicates not significant. (B) CellTiter Glo assay for viability was performed on T238 and CUTC60 cells. Cells were treated with indicated doses of dasatinib, 2 μM ABT-263, with or without 100 nM trametinib for 72 h. Data was normalized to DMSO-treated control set to 100%. Dashed line indicates 50% viability. Results shown are mean ± SEM. IC50 values for dasatinib with 2 μM ABT-263 and dasatinib with 2 μM ABT-263 and 100 nM trametinib were calculated in GraphPad Prism. 90 nM cutoff for sensitivity is indicated by the dashed line. (C) CellTiter Glo assay for viability was performed on 8505C, BCPAP, and Cal62 cells. Cells were treated indicated doses of dasatinib, 2 μM ABT-263 or 2 μM of an MCL1 inhibitor (MCL1i = A-1210477) with 10 nM trametinib for 72 h. Data were normalized to DMSO-treated control set to 100%. Dashed line indicates 50% viability. Results shown are mean ± SEM.

+1

BCL2L11 Induction Mediates Sensitivity to Src and MEK1/2 Inhibition in Thyroid Cancer
  • Article
  • Full-text available

January 2023

·

60 Reads

·

6 Citations

Cancers

Cancers

Madison M. Rose

·

Veronica L. Espinoza

·

Katelyn J. Hoff

·

[...]

·

Rebecca E. Schweppe

Simple Summary Thyroid cancer is the most common endocrine malignancy. Patients with advanced thyroid cancers have poor survival rates, largely because of limited therapeutic options to combat their aggressive nature, creating a compelling need to identify novel therapeutic targets. We and others have previously shown that Src is a clinically relevant target in thyroid cancer and that combined inhibition of Src and the MAP kinase pathway results in enhanced anti-tumor responses. The goals of this study were to identify the mechanism(s) mediating these anti-tumor effects and identify additional potential biomarkers of response to improve therapies for patients with advanced thyroid cancer. Abstract Patients with advanced thyroid cancer, including advanced papillary thyroid cancer and anaplastic thyroid cancer (ATC), have low survival rates because of the lack of efficient therapies available that can combat their aggressiveness. A total of 90% of thyroid cancers have identifiable driver mutations, which often are components of the MAPK pathway, including BRAF, RAS, and RET-fusions. In addition, Src is a non-receptor tyrosine kinase that is overexpressed and activated in thyroid cancer, which we and others have shown is a clinically relevant target. We have previously demonstrated that combined inhibition of Src with dasatinib and the MAPK pathway with trametinib synergistically inhibits growth and induces apoptosis in BRAF- and RAS-mutant thyroid cancer cells. Herein, we identified the pro-apoptotic protein BCL2L11 (BIM) as being a key mediator of sensitivity in response to combined dasatinib and trametinib treatment. Specifically, cells that are sensitive to combined dasatinib and trametinib treatment have inhibition of FAK/Src, MEK/ERK, and AKT, resulting in the dramatic upregulation of BIM, while cells that are resistant lack inhibition of AKT and have a dampened induction of BIM. Inhibition of AKT directly sensitizes resistant cells to combined dasatinib and trametinib but will not be clinically feasible. Importantly, targeting BCL-XL with the BH3-mimeitc ABT-263 is sufficient to overcome lack of BIM induction and sensitize resistant cells to combined dasatinib and trametinib treatment. This study provides evidence that combined Src and MEK1/2 inhibition is a promising therapeutic option for patients with advanced thyroid cancer and identifies BIM induction as a potential biomarker of response.

Download
Share

The molecular biology of tubulinopathies: Understanding the impact of variants on tubulin structure and microtubule regulation

November 2022

·

110 Reads

·

12 Citations

Frontiers in Cellular Neuroscience

Heterozygous, missense mutations in both α- and β-tubulin genes have been linked to an array of neurodevelopment disorders, commonly referred to as “tubulinopathies.” To date, tubulinopathy mutations have been identified in three β-tubulin isotypes and one α-tubulin isotype. These mutations occur throughout the different genetic domains and protein structures of these tubulin isotypes, and the field is working to address how this molecular-level diversity results in different cellular and tissue-level pathologies. Studies from many groups have focused on elucidating the consequences of individual mutations; however, the field lacks comprehensive models for the molecular etiology of different types of tubulinopathies, presenting a major gap in diagnosis and treatment. This review highlights recent advances in understanding tubulin structural dynamics, the roles microtubule-associated proteins (MAPs) play in microtubule regulation, and how these are inextricably linked. We emphasize the value of investigating interactions between tubulin structures, microtubules, and MAPs to understand and predict the impact of tubulinopathy mutations at the cell and tissue levels. Microtubule regulation is multifaceted and provides a complex set of controls for generating a functional cytoskeleton at the right place and right time during neurodevelopment. Understanding how tubulinopathy mutations disrupt distinct subsets of those controls, and how that ultimately disrupts neurodevelopment, will be important for establishing mechanistic themes among tubulinopathies that may lead to insights in other neurodevelopment disorders and normal neurodevelopment.


Figure 7. Potential models for the effect of α-tubulin V409I/A on XMAP215/Stu2 and microtubule polymerization. Wild-type (WT) tubulin undergoes a series of conformational states as it transitions from a curved free heterodimer into the straight microtubule lattice, which is essential for tubulin binding to a variety of microtubule-associated proteins (MAPs), such as XMAP215/Stu2 (center). Our data suggest that V409I, and more severely V409A, have decreased XMAP215/Stu2 activity at microtubule plus ends, yet have faster microtubule polymerization rates. Model 1: V410I/A tubulin unbinds from TOG (tumor overexpressed gene) domains faster, thus increasing the concentration of free tubulin to promote faster microtubule polymerization. However, if the faster microtubule polymerization rates were solely driven by a decrease in XMAP215/Stu2 and/or free tubulin concentration, we would not expect to see an increase in intrinsic polymerization rates in V409A as compared to WT, as we do in our reconstitution experiments. Therefore, we favor the second model. Model 2: the V410I/A tubulin heterodimer is intrinsically altered in a way that disrupts normal XMAP215/Stu2 regulation and drives faster microtubule polymerization. One way in which tubulin could be intrinsically changed is through adopting a straighter, lattice-favoring conformation.
Tubulinopathy mutations in TUBA1A that disrupt neuronal morphogenesis and migration override XMAP215/Stu2 regulation of microtubule dynamics

May 2022

·

23 Reads

·

6 Citations

eLife

Heterozygous, missense mutations in a- or b-tubulin genes are associated with a wide range of human brain malformations, known as tubulinopathies. We seek to understand whether a mutation’s impact at the molecular and cellular levels scale with the severity of brain malformation. Here we focus on two mutations at the valine 409 residue of TUBA1A, V409I and V409A, identified in patients with pachygyria or lissencephaly, respectively. We find that ectopic expression of TUBA1A -V409I/A mutants disrupt neuronal migration in mice and promote excessive neurite branching and a decrease in the number of neurite retraction events in primary rat neuronal cultures. These neuronal phenotypes are accompanied by increased microtubule acetylation and polymerization rates. To determine the molecular mechanisms, we modeled the V409I/A mutants in budding yeast and found that they promote intrinsically faster microtubule polymerization rates in cells and in reconstitution experiments with purified tubulin. In addition, V409I/A mutants decrease the recruitment of XMAP215/Stu2 to plus ends in budding yeast and ablate tubulin binding to TOG domains. In each assay tested, the TUBA1A -V409I mutant exhibits an intermediate phenotype between wild type and the more severe TUBA1A -V409A, reflecting the severity observed in brain malformations. Together, our data support a model in which the V409I/A mutations disrupt microtubule regulation typically conferred by XMAP215 proteins during neuronal morphogenesis and migration, and this impact on tubulin activity at the molecular level scales with the impact at the cellular and tissue levels.


Bridging the Gap: The Importance of TUBA1A α-Tubulin in Forming Midline Commissures

January 2022

·

104 Reads

·

9 Citations

Developing neurons undergo dramatic morphological changes to appropriately migrate and extend axons to make synaptic connections. The microtubule cytoskeleton, made of α/β-tubulin dimers, drives neurite outgrowth, promotes neuronal growth cone responses, and facilitates intracellular transport of critical cargoes during neurodevelopment. TUBA1A constitutes the majority of α-tubulin in the developing brain and mutations to TUBA1A in humans cause severe brain malformations accompanied by varying neurological defects, collectively termed tubulinopathies. Studies of TUBA1A function in mammalian cells have been limited by the presence of multiple genes encoding highly similar tubulin proteins, which leads to α-tubulin antibody promiscuity and makes genetic manipulation challenging. Here, we test mutant tubulin levels and assembly activity and analyze the impact of TUBA1A reduction on growth cone composition, neurite extension, and commissural axon architecture during brain development. We present a novel tagging method for studying and manipulating TUBA1A in cells without impairing tubulin function. Using this tool, we show that a TUBA1A loss-of-function mutation TUBA1AN102D (TUBA1AND), reduces TUBA1A protein levels and prevents incorporation of TUBA1A into microtubule polymers. Reduced Tuba1a α-tubulin in heterozygous Tuba1aND/+ mice leads to grossly normal brain formation except a significant impact on axon extension and impaired formation of forebrain commissures. Neurons with reduced Tuba1a as a result of the Tuba1aND mutation exhibit slower neuron outgrowth compared to controls. Neurons deficient in Tuba1a failed to localize microtubule associated protein-1b (Map1b) to the developing growth cone, likely impacting stabilization of microtubules. Overall, we show that reduced Tuba1a is sufficient to support neuronal migration and cortex development but not commissure formation, and provide mechanistic insight as to how TUBA1A tunes microtubule function to support neurodevelopment.


Tubulinopathy mutations in TUBA1A that disrupt neuronal morphogenesis and migration override XMAP215/Stu2 regulation of microtubule dynamics

December 2021

·

15 Reads

Heterozygous, missense mutations in α- or β-tubulin genes are associated with a wide range of human brain malformations, known as tubulinopathies. We seek to understand whether the impact of a mutation at the molecular and cellular levels scale with the severity of brain malformation. Here we focus on two mutations at the valine 409 residue of TUBA1A, V409I and V409A, identified in patients with pachygyria or lissencephaly, respectively. We find that ectopic expression of TUBA1A-V409I/A mutants disrupt neuronal migration in mice and promote excessive neurite branching and delayed retraction in primary neuronal cultures, accompanied by increased microtubule acetylation. To determine the molecular mechanisms, we modeled the V409I/A mutants in budding yeast and found that they promote intrinsically faster microtubule polymerization rates in cells and in reconstitution experiments with purified tubulin. In addition, V409I/A mutants decrease the recruitment of XMAP215/Stu2 to plus ends and ablate tubulin binding to TOG domains. In each assay tested, the TUBA1A-V409I mutant exhibits an intermediate phenotype between wild type and the more severe TUBA1A-V409A, reflecting the severity observed in brain malformations. Together, our data support a model in which the V409I/A mutations may limit tubulin conformational states and thereby disrupt microtubule regulation during neuronal morphogenesis and migration.


Kinetically Stabilizing Mutations in Beta Tubulins Create Isotype-Specific Brain Malformations

November 2021

·

68 Reads

·

15 Citations

Frontiers in Cell and Developmental Biology

Frontiers in Cell and Developmental Biology

Mutations in the family of genes encoding the tubulin subunits of microtubules are associated with a spectrum of human brain malformations known as tubulinopathies. How these mutations impact tubulin activity to give rise to distinct developmental consequences is poorly understood. Here we report two patients exhibiting brain malformations characteristic of tubulinopathies and heterozygous T178M missense mutations in different β-tubulin genes, TUBB2A or TUBB3. RNAseq analysis indicates that both TUBB2A and TUBB3 are expressed in the brain during development, but only TUBB2A maintains high expression in neurons into adulthood. The T178 residue is highly conserved in β-tubulins and located in the exchangeable GTP-binding pocket of β-tubulin. To determine the impact of T178M on β-tubulin function we created an analogous mutation in the β-tubulin of budding yeast and show that the substitution acts dominantly to produce kinetically stabilized microtubules that assemble and disassemble slowly, with fewer transitions between these states. In vitro experiments with purified mutant tubulin demonstrate that T178M decreases the intrinsic assembly activity of β-tubulin and forms microtubules that rarely transition to disassembly. We provide evidence that the T178M substitution disrupts GTPase-dependent conformational changes in tubulin, providing a mechanistic explanation for kinetic stabilization. Our findings demonstrate the importance of tubulin’s GTPase activity during brain development, and indicate that tubulin isotypes play different, important roles during brain development.


Inhibition of the CCL2 receptor, CCR2, enhances tumor response to immune checkpoint therapy

November 2020

·

86 Reads

·

98 Citations

Communications Biology

Immunotherapies targeting the PD-1/PD-L1 axis are now a mainstay in the clinical management of multiple cancer types, however, many tumors still fail to respond. CCL2 is highly expressed in various cancer types and has been shown to be associated with poor prognosis. Inhibition or blockade of the CCL2/CCR2 signaling axis has thus been an area of interest for cancer therapy. Here we show across multiple murine tumor and metastasis models that CCR2 antagonism in combination with anti-PD-1 therapy leads to sensitization and enhanced tumor response over anti-PD-1 monotherapy. We show that enhanced treatment response correlates with enhanced CD8⁺ T cell recruitment and activation and a concomitant decrease in CD4⁺ regulatory T cell. These results provide strong preclinical rationale for further clinical exploration of combining CCR2 antagonism with PD-1/PD-L1-directed immunotherapies across multiple tumor types especially given the availability of small molecule CCR2 inhibitors and antibodies.


Figure 4 TUBA1A ND α-tubulin does not incorporate into neuronal microtubule polymers. A. Schematic of cor tical neur on isolation and tr ansfection. B. Cor tical r at neur ons at DIV 2 tr ansfected with TUBA1A-GFP. Top panel shows neur ons with intr acellular envir onment intact (unextr acted), containing soluble tubulin dimer s and polymerized micr otubules tr ansfected to expr ess a TUBA1A-GFP fusion pr otein. Bottom panel shows neur ons with soluble tubulin dimer s extr acted, showing only GFP-labeled TUBA1A that is incor por ated into micr otubule polymer. C. Rat cortical neur ons at DIV 2 tr ansfected with wild-type (WT) TUBA1A-His6, TUBA1A ND -His6 LOF mutation, and TUBA1A TE -His6 polymerization-incompetent mutant as a negative contr ol. Top panels show composite image containing membrane-bound GFP (gr een) for confir mation of tr ansfection, α-His6 (Magenta) and DAPI (blue) immunolabeling. Middle panels show unextr acted and bottom panels show tubulin extracted DIV 2 neur ons (descr ibed in B.), labeled with α-His6 antibodies to visualize ectopic TUBA1A-His6 proteins. All images were taken at 63X magnification, scale bar is 25µm.
Figure 7 Tuba1a ND neurons do not correctly localize Map1b to the developing growth cone. A. Wester n blots showing Map1b pr otein associate with a tubulin-enr iched fr action fr om br ain (top panel) and total Map1b protein in whole br ain lysate (bottom panel) fr om wild-type (WT) and Tuba1a ND/+ mice. Due to the amount of pr otein that was loaded for Map1b western blots, antibody-stained bands for α-tubulin wer e over satur ated and could not be quantified, thus Map1b was nor malized to the 50kDa band (pr esumed to be primar ily tubulin) on a UV-activated stain-fr ee blot. B. Scatter plot quantifying Map1b associated with the tubulin-enr iched br ain lysate, nor malized to the 50kDa pr esumed tubulin band using stain-fr ee western blotting. p=0.03 C. Scatter plot r epr esenting total Map1b pr otein in br ain lysate by wester n blot, nor malized to the total protein on a stain-fr ee blot. p=0.98 D. Scatter plot showing the subcellular distr ibution of Map1b pr otein in WT and Tuba1a ND/+ cor tical neurons at DIV 3. Data ar e r epr esented as Map1b fluorescent signal in gr owth cone r egion divided by a r egion proximal to the cell body. p=0.009. E. Repr esentative images showing altered subcellular distribution of Map1b in Tuba1a ND/+ (bottom) cor tical neur ons compared to WT (top) at DIV 3. Composite and individual channel gr ayscale images of MAP2 and Map1b immunocytochemistr y are shown, i and ii indicate enlarged regions shown in insets. Scale bar s are 10µm. Differ ences between gr oups wer e evaluated by t test. * p<0.05; ** p<0.01.
Tuba1a is uniquely important for axon guidance through midline commissural structures

May 2020

·

118 Reads

·

3 Citations

Developing neurons undergo dramatic morphological changes to appropriately migrate and extend axons to make synaptic connections. The microtubule cytoskeleton, made of α/β-tubulin dimers, drives neurite outgrowth, promotes neuronal growth cone responses, and facilitates intracellular transport of critical cargoes during neurodevelopment. TUBA1A constitutes the majority of α-tubulin in the developing brain and mutations to TUBA1A in humans cause severe brain malformations accompanied by varying neurological defects, collectively termed tubulinopathies. Studies of TUBA1A function in vivo have been limited by the presence of multiple genes encoding highly similar tubulin proteins, which prevents TUBA1A-specific antibody generation and makes genetic manipulation challenging. Here we present a novel tagging method for studying and manipulating TUBA1A in cells without impairing tubulin function. Using this tool, we show that a TUBA1A loss-of-function mutation TUBA1AN102D (TUBA1AND), reduced the amount of TUBA1A protein and prevented incorporation of TUBA1A into microtubule polymers. Reduced Tuba1a α-tubulin in heterozygous Tuba1aND/+ mice significantly impacted axon extension and impaired formation of forebrain commissures. Neurons with reduced Tuba1a caused by Tuba1aND had altered microtubule dynamics and slower neuron outgrowth compared to controls. Neurons deficient in Tuba1a failed to localize microtubule associated protein-1b (Map1b) to the developing growth cone, likely impacting reception of developmental guidance cues. Overall, we show that reduced Tuba1a is sufficient to support neuronal migration, but not axon guidance, and provide mechanistic insight as to how TUBA1A tunes microtubule function to support neurodevelopment.

Citations (7)


... Another decreased EciRNA-circBIRC6 might facilitate the progression of PTC by regulating miR-24-3p/BCL2L11 axis. BCL2L11, belonged to BCL2 family, functioned as a tumor suppressor gene by regulating cell apoptosis [44] . BCL2L11 expression was usually reduced in PTC patients, which was reported to activate apoptosis after treatment with Src and MEK1/2 inhibitors [45] . ...

Reference:

Expression profiling and bioinformatics analysis of serum exosomal circular RNAs in lymph node metastasis of papillary thyroid carcinoma
BCL2L11 Induction Mediates Sensitivity to Src and MEK1/2 Inhibition in Thyroid Cancer
Cancers

Cancers

... Commercially available genetic panels may suggest classical Mendelian or complex patterns of inheritance. Tubulinopathies exemplify one of the multiple complex neuronal disease pathways that represent genetic defects involving cell cycle abnormalities within the cytoskeleton of different neuroprogenitor populations (145,146). Interdisciplinary efforts are now underway to establish prenatal diagnoses of tubulinopathies (147). Post-translational modifications (PTMs) are comprised of a complicated sequence of spatially and temporally regulated processes required for healthy neurodevelopment (148). ...

The molecular biology of tubulinopathies: Understanding the impact of variants on tubulin structure and microtubule regulation

Frontiers in Cellular Neuroscience

... Tubulinopathies are a collection of clinical conditions caused by de novo tubulin mutations that impair neuronal migration during development to cause severe brain malformations [103,108,109]. Expression of αR402, αG436 and αV409 missense mutations in budding yeast and mouse model organisms implicate impaired dynein or XMAP215 interactions as the defect underlying these tubulinopathies [110][111][112]. Although αR402 substituted tubulins assemble into microtubules, these do not support dynein motor activity and ectopic expression of R402C or R402H substituted α-tubulin disrupts cortical neuronal migration in the developing mouse brain [110]. ...

Tubulinopathy mutations in TUBA1A that disrupt neuronal morphogenesis and migration override XMAP215/Stu2 regulation of microtubule dynamics

eLife

... As described previously 63 , for the separation of soluble tubulin and microtubules, cells were washed once with PBS and PHEM buffer, respectively, and resuspended in PHEM buffer containing 0.1% Triton X-100 and 0.1% DMSO at RT for 10 min. The cells were then centrifugated at 12,000 rpm for 10 min. ...

Bridging the Gap: The Importance of TUBA1A α-Tubulin in Forming Midline Commissures
Frontiers in Cell and Developmental Biology

Frontiers in Cell and Developmental Biology

... Humans have 9 α and 10 β tubulin isotypes 29,30 . It is unclear why humans need so many tubulins, but we do: single point mutations in specific human tubulin isotypes are documented to cause specific human diseases (tubulinopathies) 31 , often linked to developmental abnormalities 32,33 . Taxol resistance in patients can be accompanied by an increase in β3 tubulin expression 34 , suggesting that β3 tubulin expression can protect against taxol 26,35 . ...

Kinetically Stabilizing Mutations in Beta Tubulins Create Isotype-Specific Brain Malformations
Frontiers in Cell and Developmental Biology

Frontiers in Cell and Developmental Biology

... CCL2 can mediate the invasiveness of metastatic melanoma cells [54]. Ligand-receptor interactions lead to the recruitment of protumoral stromal cells as well as myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (M2 phenotype) [55,56]. NF-κB and STAT pathways can lead to CCL2 production [57]. ...

Inhibition of the CCL2 receptor, CCR2, enhances tumor response to immune checkpoint therapy

Communications Biology

... We expressed a hexahistidine (6X-His)-tagged TUBA1A construct in cortical neuron cultures harvested from P0 rats. The 6X-His tag was inserted between residues I42 and G43 in a flexible loop of TUBA1A that allows for the addition of amino acids without perturbing tubulin function (Buscaglia et al., 2020a). After 2 days in culture (days in vitro 2 [DIV2]), we extracted soluble tubulin from the neurons, fixed the cells, and stained for the 6X-His tag. ...

Tuba1a is uniquely important for axon guidance through midline commissural structures