1
|
Mikesell AR, Isaeva E, Schulte ML, Menzel AD, Sriram A, Prahl MM, Shin SM, Sadler KE, Yu H, Stucky CL. Increased keratinocyte activity and PIEZO1 signaling contribute to paclitaxel-induced mechanical hypersensitivity. Sci Transl Med 2024; 16:eadn5629. [PMID: 39661703 DOI: 10.1126/scitranslmed.adn5629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
Recent work demonstrates that epidermal keratinocytes are critical for normal touch sensation. However, it is unknown whether keratinocytes contribute to touch-evoked pain and hypersensitivity after tissue injury. Here, we used a mouse model of paclitaxel treatment to determine the extent to which keratinocyte activity contributes to the severe neuropathic pain that accompanies chemotherapy. We found that keratinocyte inhibition by either optogenetic or chemogenetic methods largely alleviated paclitaxel-induced mechanical hypersensitivity across acute and persistent time points from 2 days through 3 weeks. Furthermore, we found that paclitaxel exposure sensitized mouse and human keratinocytes to mechanical stimulation and enhanced currents of PIEZO1, a mechanosensitive channel highly expressed in keratinocytes. Deletion of PIEZO1 from keratinocytes alleviated paclitaxel-induced mechanical hypersensitivity in mice. These findings suggest that nonneuronal cutaneous cells contribute substantially to neuropathic pain and pave the way for the development of new pain relief strategies that target epidermal keratinocytes and PIEZO1.
Collapse
Affiliation(s)
- Alexander R Mikesell
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elena Isaeva
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Anthony D Menzel
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anvitha Sriram
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Megan M Prahl
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Katelyn E Sadler
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
2
|
He L, Meng F, Chen R, Qin J, Sun M, Fan Z, Du J. Precise Regulations at the Subcellular Level through Intracellular Polymerization, Assembly, and Transformation. JACS AU 2024; 4:4162-4186. [PMID: 39610726 PMCID: PMC11600172 DOI: 10.1021/jacsau.4c00849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/30/2024]
Abstract
A living cell is an intricate machine that creates subregions to operate cell functions effectively. Subcellular dysfunction has been identified as a potential druggable target for successful drug design and therapy. The treatments based on intracellular polymerization, self-assembly, or transformation offer various advantages, including enhanced blood circulation of monomers, long-term drug delivery pharmacokinetics, low drug resistance, and the ability to target deep tissues and organelles. In this review, we discuss the latest developments of intracellular synthesis applied to precisely control cellular functions. First, we discuss the design and applications of endogenous and exogenous stimuli-triggered intracellular polymerization, self-assembly, and dynamic morphology transformation of biomolecules at the subcellular level. Second, we highlight the benefits of these strategies applied in cancer diagnosis and treatment and modulating cellular states or cell metabolism of living systems. Finally, we conclude the recent progress in this field, discuss future perspectives, analyze the challenges of the intracellular functional reactions for regulation, and find future opportunities.
Collapse
Affiliation(s)
- Le He
- School
of Materials Science and Engineering, East
China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
| | - Fanying Meng
- Department
of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Ran Chen
- Department
of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jinlong Qin
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
| | - Min Sun
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
| | - Zhen Fan
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
- Department
of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jianzhong Du
- School
of Materials Science and Engineering, East
China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- Department
of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology
and Brain Functional Modulation, Clinical Research Center for Anesthesiology
and Perioperative Medicine, Translational Research Institute of Brain
and Brain-Like Intelligence, Shanghai Fourth People’s Hospital,
School of Medicine, Tongji University, Shanghai 200434, China
- Department
of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| |
Collapse
|
3
|
Gonzalez SJ, Heckel JM, Goldblum RR, Reid TA, McClellan M, Gardner MK. Rapid binding to protofilament edge sites facilitates tip tracking of EB1 at growing microtubule plus-ends. eLife 2024; 13:e91719. [PMID: 38385657 PMCID: PMC10883673 DOI: 10.7554/elife.91719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/31/2024] [Indexed: 02/23/2024] Open
Abstract
EB1 is a key cellular protein that delivers regulatory molecules throughout the cell via the tip-tracking of growing microtubule plus-ends. Thus, it is important to understand the mechanism for how EB1 efficiently tracks growing microtubule plus-ends. It is widely accepted that EB1 binds with higher affinity to GTP-tubulin subunits at the growing microtubule tip, relative to GDP-tubulin along the microtubule length. However, it is unclear whether this difference in affinity alone is sufficient to explain the tip-tracking of EB1 at growing microtubule tips. Previously, we found that EB1 binds to exposed microtubule protofilament-edge sites at a ~70 fold faster rate than to closed-lattice sites, due to diffusional steric hindrance to binding. Thus, we asked whether rapid protofilament-edge binding could contribute to efficient EB1 tip tracking. A computational simulation with differential EB1 on-rates based on closed-lattice or protofilament-edge binding, and with EB1 off-rates that were dependent on the tubulin hydrolysis state, robustly recapitulated experimental EB1 tip tracking. To test this model, we used cell-free biophysical assays, as well as live-cell imaging, in combination with a Designed Ankyrin Repeat Protein (DARPin) that binds exclusively to protofilament-edge sites, and whose binding site partially overlaps with the EB1 binding site. We found that DARPin blocked EB1 protofilament-edge binding, which led to a decrease in EB1 tip tracking on dynamic microtubules. We conclude that rapid EB1 binding to microtubule protofilament-edge sites contributes to robust EB1 tip tracking at the growing microtubule plus-end.
Collapse
Affiliation(s)
- Samuel J Gonzalez
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| | - Julia M Heckel
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| | - Rebecca R Goldblum
- Department of Biophysics, Molecular Biology, and Biochemistry, University of MinnesotaMinneapolisUnited States
- Medical Scientist Training Program, University of MinnesotaMinneapolisUnited States
| | - Taylor A Reid
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| | - Mark McClellan
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| | - Melissa K Gardner
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| |
Collapse
|
4
|
Lim S, Jung HR, Lee H, Chu Y, Kim H, Kim E, Lee S. Microtubule-destabilizing agents enhance STING-mediated innate immune response via biased mechanism in human monocyte cells. Biomed Pharmacother 2023; 169:115883. [PMID: 37979373 DOI: 10.1016/j.biopha.2023.115883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023] Open
Abstract
The stimulator of the interferon gene (STING) signaling pathway acts as a primary defense system against DNA pathogens. Because of the crucial role of STING in type I interferon (IFN) response and innate immunity, extensive research has been conducted to elucidate the roles of various effector molecules involved in STING-mediated signal transduction. However, despite the substantial contribution of microtubules to the immune system, the association between the STING signaling pathway and microtubules remains unclear. In this study, we revealed that the modulation of STING via microtubule-destabilizing agents (MDAs) specifically induced type I IFN responses rather than inflammatory responses in human monocytes. Co-treatment of MDAs with STING agonists induced the elevation of phospho-TANK-binding kinase 1 (TBK1), amplifying the innate immune response. However, during the deficiency of TBK1, the non-canonical signaling pathway through nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) contributed to MDA-induced STING activation in type I IFN response which suggested the versatile regulation of MDA in STING-mediated immunity.
Collapse
Affiliation(s)
- Songhyun Lim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Hee Ra Jung
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Hyelim Lee
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yeonjeong Chu
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, South Korea
| | - Hyejin Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea
| | - Eunha Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, South Korea
| | - Sanghee Lee
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; Department of HY-KIST Bio-convergence, Hanyang University, Seoul 04763, South Korea.
| |
Collapse
|
5
|
Gudimchuk NB, Alexandrova VV. Measuring and modeling forces generated by microtubules. Biophys Rev 2023; 15:1095-1110. [PMID: 37974983 PMCID: PMC10643784 DOI: 10.1007/s12551-023-01161-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/25/2023] [Indexed: 11/19/2023] Open
Abstract
Tubulins are essential proteins, which are conserved across all eukaryotic species. They polymerize to form microtubules, cytoskeletal components of paramount importance for cellular mechanics. The microtubules combine an extraordinarily high flexural rigidity and a non-equilibrium behavior, manifested in their intermittent assembly and disassembly. These chemically fueled dynamics allow microtubules to generate significant pushing and pulling forces at their ends to reposition intracellular organelles, remodel membranes, bear compressive forces, and transport chromosomes during cell division. In this article, we review classical and recent studies, which have allowed the quantification of microtubule-generated forces. The measurements, to which we owe most of the quantitative information about microtubule forces, were carried out in biochemically reconstituted systems in vitro. We also discuss how mathematical and computational modeling has contributed to the interpretations of these results and shaped our understanding of the mechanisms of force production by tubulin polymerization and depolymerization.
Collapse
Affiliation(s)
- Nikita B. Gudimchuk
- Department of Physics, Lomonosov Moscow State University, Moscow, Russia
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russia
- Pskov State University, Pskov, Russia
| | - Veronika V. Alexandrova
- Department of Physics, Lomonosov Moscow State University, Moscow, Russia
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russia
| |
Collapse
|
6
|
Chew YM, Cross RA. Taxol acts differently on different tubulin isotypes. Commun Biol 2023; 6:946. [PMID: 37717119 PMCID: PMC10505170 DOI: 10.1038/s42003-023-05306-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/31/2023] [Indexed: 09/18/2023] Open
Abstract
Taxol is a small molecule effector that allosterically locks tubulin into the microtubule lattice. We show here that taxol has different effects on different single-isotype microtubule lattices. Using in vitro reconstitution, we demonstrate that single-isotype α1β4 GDP-tubulin lattices are stabilised and expanded by 10 µM taxol, as reported by accelerated microtubule gliding in kinesin motility assays, whereas single-isotype α1β3 GDP-tubulin lattices are stabilised but not expanded. This isotype-specific action of taxol drives gliding of segmented-isotype GDP-taxol microtubules along convoluted, sinusoidal paths, because their expanded α1β4 segments try to glide faster than their compacted α1β3 segments. In GMPCPP, single-isotype α1β3 and α1β4 lattices both show accelerated gliding, indicating that both can in principle be driven to expand. We therefore propose that taxol-induced lattice expansion requires a higher taxol occupancy than taxol-induced stabilisation, and that higher taxol occupancies are accessible to α1β4 but not α1β3 single-isotype lattices.
Collapse
Affiliation(s)
- Yean Ming Chew
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, CV4 7LA, UK
| | - Robert A Cross
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, CV4 7LA, UK.
| |
Collapse
|
7
|
Radwitz J, Hausrat TJ, Heisler FF, Janiesch PC, Pechmann Y, Rübhausen M, Kneussel M. Tubb3 expression levels are sensitive to neuronal activity changes and determine microtubule growth and kinesin-mediated transport. Cell Mol Life Sci 2022; 79:575. [DOI: 10.1007/s00018-022-04607-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/03/2022]
Abstract
AbstractMicrotubules are dynamic polymers of α/β-tubulin. They regulate cell structure, cell division, cell migration, and intracellular transport. However, functional contributions of individual tubulin isotypes are incompletely understood. The neuron-specific β-tubulin Tubb3 displays highest expression around early postnatal periods characterized by exuberant synaptogenesis. Although Tubb3 mutations are associated with neuronal disease, including abnormal inhibitory transmission and seizure activity in patients, molecular consequences of altered Tubb3 levels are largely unknown. Likewise, it is unclear whether neuronal activity triggers Tubb3 expression changes in neurons. In this study, we initially asked whether chemical protocols to induce long-term potentiation (cLTP) affect microtubule growth and the expression of individual tubulin isotypes. We found that growing microtubules and Tubb3 expression are sensitive to changes in neuronal activity and asked for consequences of Tubb3 downregulation in neurons. Our data revealed that reduced Tubb3 levels accelerated microtubule growth in axons and dendrites. Remarkably, Tubb3 knockdown induced a specific upregulation of Tubb4 gene expression, without changing other tubulin isotypes. We further found that Tubb3 downregulation reduces tubulin polyglutamylation, increases KIF5C motility and boosts the transport of its synaptic cargo N-Cadherin, which is known to regulate synaptogenesis and long-term potentiation. Due to the large number of tubulin isotypes, we developed and applied a computational model based on a Monte Carlo simulation to understand consequences of tubulin expression changes in silico. Together, our data suggest a feedback mechanism with neuronal activity regulating tubulin expression and consequently microtubule dynamics underlying the delivery of synaptic cargoes.
Collapse
|
8
|
Dandugudumula R, Fischer-Weinberger R, Zilberstein D. Morphogenesis Dynamics in Leishmania Differentiation. Pathogens 2022; 11:952. [PMID: 36145385 PMCID: PMC9505065 DOI: 10.3390/pathogens11090952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Leishmania, the causative agent of leishmaniasis, is an obligatory intracellular parasite that cycles between phagolysosome of mammalian macrophages, where it resides as round intracellular amastigotes, and the midgut of female sandflies, where it resides as extracellular elongated promastigotes. This protozoan parasite cytoskeleton is composed of stable and abundant subpellicular microtubules (SPMT). This study aims to determine the kinetics of developmental morphogenesis and assess whether microtubules remodelling is involved in this process. Using image-streaming technology, we observed that rounding of promastigotes during differentiation into amastigotes was initiated promptly after exposure to the differentiation signal. Stabilizing microtubules with taxol sped rounding, but later killed differentiating parasites if taxol was not removed. Microtubule destabilizers such as vinblastine had no effect on the rate of rounding, nor on the viability of differentiating parasites. In the reverse process, elongation is initiated after a delay of 7.5 and completed 72 h after exposure to the amastigote to the promastigote differentiation signal. During the delay, parasites became highly sensitive to treatment with microtubule destabilizers. The addition of vinblastine during the first 7.5 h halted differentiation and killed parasites. Between 8 and 24 h, parasites gradually became resistant to vinblastine and, in parallel, started to elongate. In contrast, taxol had no effect on parasite elongation, nor on the viability of these cells. In a parallel study, we showed that the Leishmania-specific protein kinase A (PKA) holoenzyme containing the LdPKAR3-C3 complex is essential for promastigote elongation. Mutant promastigotes lacking either of these proteins are round, but maintain their flagella. Here, we observed that during differentiation into amastigotes, these mutants round at the same rate as the wild type, but never exceed the WT density of round amastigotes. In the reverse process, these mutants undergo the same initial delay and then elongate at the same rate as the WT. They stop elongating when they reach 20% of elongated cells in mature promastigotes. Our analysis indicates that while promastigote rounding into amastigotes did not require microtubule remodelling, morphogenesis of round amastigotes into elongated promastigotes required microtubule rearrangement before elongation was initiated. This is the first study that investigates the dynamics of microtubules during parasite development.
Collapse
Affiliation(s)
| | | | - Dan Zilberstein
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
9
|
Naghsh F, Monajjemi M. Thermodynamic Study of Assembling ↔ Disassembling of Microtubules via the Monte Carlo Simulation. RUSSIAN JOURNAL OF PHYSICAL CHEMISTRY A 2022. [DOI: 10.1134/s0036024422070111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
10
|
Abstract
Melanoma is a relentless type of skin cancer which involves myriad signaling pathways which regulate many cellular processes. This makes melanoma difficult to treat, especially when identified late. At present, therapeutics include chemotherapy, surgical resection, biochemotherapy, immunotherapy, photodynamic and targeted approaches. These interventions are usually administered as either a single-drug or in combination, based on tumor location, stage, and patients' overall health condition. However, treatment efficacy generally decreases as patients develop treatment resistance. Genetic profiling of melanocytes and the discovery of novel molecular factors involved in the pathogenesis of melanoma have helped to identify new therapeutic targets. In this literature review, we examine several newly approved therapies, and briefly describe several therapies being assessed for melanoma. The goal is to provide a comprehensive overview of recent developments and to consider future directions in the field of melanoma.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Pavan Kumar Dhanyamraju, Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA17033, USA. Tel: +1-6096474712, E-mail:
| | - Trupti N. Patel
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
11
|
Park K, Hoff KJ, Wethekam L, Stence N, Saenz M, Moore JK. Kinetically Stabilizing Mutations in Beta Tubulins Create Isotype-Specific Brain Malformations. Front Cell Dev Biol 2021; 9:765992. [PMID: 34869359 PMCID: PMC8637541 DOI: 10.3389/fcell.2021.765992] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/29/2021] [Indexed: 11/13/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Kristen Park
- Department of Pediatrics and Neurology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Katelyn J Hoff
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Linnea Wethekam
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nicholas Stence
- Section of Pediatric Radiology, Department of Radiology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Margarita Saenz
- Section of Genetics, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeffrey K Moore
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
12
|
Regulation of microtubule dynamics, mechanics and function through the growing tip. Nat Rev Mol Cell Biol 2021; 22:777-795. [PMID: 34408299 DOI: 10.1038/s41580-021-00399-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Microtubule dynamics and their control are essential for the normal function and division of all eukaryotic cells. This plethora of functions is, in large part, supported by dynamic microtubule tips, which can bind to various intracellular targets, generate mechanical forces and couple with actin microfilaments. Here, we review progress in the understanding of microtubule assembly and dynamics, focusing on new information about the structure of microtubule tips. First, we discuss evidence for the widely accepted GTP cap model of microtubule dynamics. Next, we address microtubule dynamic instability in the context of structural information about assembly intermediates at microtubule tips. Three currently discussed models of microtubule assembly and dynamics are reviewed. These are considered in the context of established facts and recent data, which suggest that some long-held views must be re-evaluated. Finally, we review structural observations about the tips of microtubules in cells and describe their implications for understanding the mechanisms of microtubule regulation by associated proteins, by mechanical forces and by microtubule-targeting drugs, prominently including cancer chemotherapeutics.
Collapse
|
13
|
Gangapuram M, Mazzio EA, Redda KK, Soliman KFA. Transcriptome Profile Analysis of Triple-Negative Breast Cancer Cells in Response to a Novel Cytostatic Tetrahydroisoquinoline Compared to Paclitaxel. Int J Mol Sci 2021; 22:ijms22147694. [PMID: 34299315 PMCID: PMC8306781 DOI: 10.3390/ijms22147694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
The absence of chemotherapeutic target hormone receptors in breast cancer is descriptive of the commonly known triple-negative breast cancer (TNBC) subtype. TNBC remains one of the most aggressive invasive breast cancers, with the highest mortality rates in African American women. Therefore, new drug therapies are continually being explored. Microtubule-targeting agents such as paclitaxel (Taxol) interfere with microtubules dynamics, induce mitotic arrest, and remain a first-in-class adjunct drug to treat TNBC. Recently, we synthesized a series of small molecules of substituted tetrahydroisoquinolines (THIQs). The lead compound of this series, with the most potent cytostatic effect, was identified as 4-Ethyl-N-(7-hydroxy-3,4-dihydroisoquinolin-2(1H)-yl) benzamide (GM-4-53). In our previous work, GM-4-53 was similar to paclitaxel in its capacity to completely abrogate cell cycle in MDA-MB-231 TNBC cells, with the former not impairing tubulin depolymerization. Given that GM-4-53 is a cytostatic agent, and little is known about its mechanism of action, here, we elucidate differences and similarities to paclitaxel by evaluating whole-transcriptome microarray data in MDA-MB-231 cells. The data obtained show that both drugs were cytostatic at non-toxic concentrations and caused deformed morphological cytoskeletal enlargement in 2D cultures. In 3D cultures, the data show greater core penetration, observed by GM-4-53, than paclitaxel. In concentrations where the drugs entirely blocked the cell cycle, the transcriptome profile of the 48,226 genes analyzed (selection criteria: (p-value, FDR p-value < 0.05, fold change −2< and >2)), paclitaxel evoked 153 differentially expressed genes (DEGs), GM-4-53 evoked 243 DEGs, and, of these changes, 52/153 paclitaxel DEGs were also observed by GM-4-53, constituting a 34% overlap. The 52 DEGS analysis by String database indicates that these changes involve transcripts that influence microtubule spindle formation, chromosome segregation, mitosis/cell cycle, and transforming growth factor-β (TGF-β) signaling. Of interest, both drugs effectively downregulated “inhibitor of DNA binding, dominant negative helix-loop-helix” (ID) transcripts; ID1, ID3 and ID4, and amphiregulin (AREG) and epiregulin (EREG) transcripts, which play a formidable role in cell division. Given the efficient solubility of GM-4-53, its low molecular weight (MW; 296), and capacity to penetrate a small solid tumor mass and effectively block the cell cycle, this drug may have future therapeutic value in treating TNBC or other cancers. Future studies will be required to evaluate this drug in preclinical models.
Collapse
|
14
|
Wang J, Yang K, Yao L, Ma Z, Li C, Si E, Li B, Meng Y, Ma X, Shang X, Wang H. Metabolomics Analyses Provide Insights Into Nutritional Value and Abiotic Stress Tolerance in Halophyte Halogeton glomeratus. FRONTIERS IN PLANT SCIENCE 2021; 12:703255. [PMID: 34290730 PMCID: PMC8287573 DOI: 10.3389/fpls.2021.703255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/08/2021] [Indexed: 06/13/2023]
Abstract
Halogeton glomeratus is a succulent annual herbaceous halophyte belonging to the Chenopodiaceae family, has attracted wide attention as a promising candidate for phytoremediation and as an oilseed crop and noodle-improver. More importantly, H. glomeratus has important medicinal value in traditional Chinese medicine. However, there are few comprehensive studies on the nutrients, particularly secondary metabolites. Here, we adopted untargeted metabolomics to compare the differences in metabolites of different tissues (root, stem, leaf, and seed) and identify the compounds related to pharmacological effects and response to abiotic stress in H. glomeratus. A total of 2,152 metabolites were identified, and the metabolic profiles of root, stem, leaf, and seed samples were clearly separated. More than 50% of the metabolites showed significant differences among root, stem, leaf, and seed. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of differential metabolites suggested an extensive alteration in the metabolome among the different organs. Furthermore, the identified metabolites related to pharmacological effects and response to abiotic stress included flavones, flavonols, flavandiols, glucosinolates, isoquinolines, pyridines, indoles, amino acids, lipids, carbohydrates, and ATP-binding cassette transporters. These metabolites have application in treating human cardiovascular diseases, cancers, diabetes, and heart disease, induce sleeping and have nutritive value. In plants, they are related to osmotic adjustment, alleviating cell damage, adjusting membrane lipid action and avoiding toxins. To the best of our knowledge, this is the first metabolomics-based report to overview the metabolite compounds in H. glomeratus and provide a reference for future development and utilization of H. glomeratus.
Collapse
Affiliation(s)
- Juncheng Wang
- Gansu Provincial Key Lab of Aridland Crop Science/Gansu Key Lab of Crop Improvement and Germplasm Enhancement, Lanzhou, China
- Department of Crop Genetics and Breeding, College of Agronomy, Gansu Agricultural University, Lanzhou, China
| | - Ke Yang
- Gansu Provincial Key Lab of Aridland Crop Science/Gansu Key Lab of Crop Improvement and Germplasm Enhancement, Lanzhou, China
- Department of Crop Genetics and Breeding, College of Agronomy, Gansu Agricultural University, Lanzhou, China
| | - Lirong Yao
- Gansu Provincial Key Lab of Aridland Crop Science/Gansu Key Lab of Crop Improvement and Germplasm Enhancement, Lanzhou, China
- Department of Crop Genetics and Breeding, College of Agronomy, Gansu Agricultural University, Lanzhou, China
| | - Zengke Ma
- Gansu Provincial Key Lab of Aridland Crop Science/Gansu Key Lab of Crop Improvement and Germplasm Enhancement, Lanzhou, China
- Department of Crop Genetics and Breeding, College of Agronomy, Gansu Agricultural University, Lanzhou, China
| | - Chengdao Li
- Western Barley Genetics Alliance, College of Science, Health, Engineering and Education, Murdoch University, Murdoch, WA, Australia
| | - Erjing Si
- Gansu Provincial Key Lab of Aridland Crop Science/Gansu Key Lab of Crop Improvement and Germplasm Enhancement, Lanzhou, China
- Department of Crop Genetics and Breeding, College of Agronomy, Gansu Agricultural University, Lanzhou, China
| | - Baochun Li
- Gansu Provincial Key Lab of Aridland Crop Science/Gansu Key Lab of Crop Improvement and Germplasm Enhancement, Lanzhou, China
- Department of Botany, College of Life Sciences and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yaxiong Meng
- Gansu Provincial Key Lab of Aridland Crop Science/Gansu Key Lab of Crop Improvement and Germplasm Enhancement, Lanzhou, China
- Department of Crop Genetics and Breeding, College of Agronomy, Gansu Agricultural University, Lanzhou, China
| | - Xiaole Ma
- Gansu Provincial Key Lab of Aridland Crop Science/Gansu Key Lab of Crop Improvement and Germplasm Enhancement, Lanzhou, China
- Department of Crop Genetics and Breeding, College of Agronomy, Gansu Agricultural University, Lanzhou, China
| | - Xunwu Shang
- Department of Crop Genetics and Breeding, College of Agronomy, Gansu Agricultural University, Lanzhou, China
| | - Huajun Wang
- Gansu Provincial Key Lab of Aridland Crop Science/Gansu Key Lab of Crop Improvement and Germplasm Enhancement, Lanzhou, China
- Department of Crop Genetics and Breeding, College of Agronomy, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
15
|
Best RL, LaPointe NE, Azarenko O, Miller H, Genualdi C, Chih S, Shen BQ, Jordan MA, Wilson L, Feinstein SC, Stagg NJ. Microtubule and tubulin binding and regulation of microtubule dynamics by the antibody drug conjugate (ADC) payload, monomethyl auristatin E (MMAE): Mechanistic insights into MMAE ADC peripheral neuropathy. Toxicol Appl Pharmacol 2021; 421:115534. [PMID: 33852878 DOI: 10.1016/j.taap.2021.115534] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 11/25/2022]
Abstract
Monomethyl auristatin E (MMAE) is a potent anti-cancer microtubule-targeting agent (MTA) used as a payload in three approved MMAE-containing antibody drug conjugates (ADCs) and multiple ADCs in clinical development to treat different types of cancers. Unfortunately, MMAE-ADCs can induce peripheral neuropathy, a frequent adverse event leading to treatment dose reduction or discontinuation and subsequent clinical termination of many MMAE-ADCs. MMAE-ADC-induced peripheral neuropathy is attributed to non-specific uptake of the ADC in peripheral nerves and release of MMAE, disrupting microtubules (MTs) and causing neurodegeneration. However, molecular mechanisms underlying MMAE and MMAE-ADC effects on MTs remain unclear. Here, we characterized MMAE-tubulin/MT interactions in reconstituted in vitro soluble tubulin or MT systems and evaluated MMAE and vcMMAE-ADCs in cultured human MCF7 cells. MMAE bound to soluble tubulin heterodimers with a maximum stoichiometry of ~1:1, bound abundantly along the length of pre-assembled MTs and with high affinity at MT ends, introduced structural defects, suppressed MT dynamics, and reduced the kinetics and extent of MT assembly while promoting tubulin ring formation. In cells, MMAE and MMAE-ADC (via nonspecific uptake) suppressed proliferation, mitosis and MT dynamics, and disrupted the MT network. Comparing MMAE action to other MTAs supports the hypothesis that peripheral neuropathy severity is determined by the precise mechanism(s) of each individual drug-MT interaction (location of binding, affinity, effects on morphology and dynamics). This work demonstrates that MMAE binds extensively to tubulin and MTs and causes severe MT dysregulation, providing convincing evidence that MMAE-mediated inhibition of MT-dependent axonal transport leads to severe peripheral neuropathy.
Collapse
Affiliation(s)
- Rebecca L Best
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Nichole E LaPointe
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Olga Azarenko
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Herb Miller
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Christine Genualdi
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Stephen Chih
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Ben-Quan Shen
- Preclinical and Translational PK, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Mary Ann Jordan
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Leslie Wilson
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Stuart C Feinstein
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA.
| | - Nicola J Stagg
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
16
|
Atomistic Basis of Microtubule Dynamic Instability Assessed Via Multiscale Modeling. Ann Biomed Eng 2021; 49:1716-1734. [PMID: 33537926 PMCID: PMC8302526 DOI: 10.1007/s10439-020-02715-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023]
Abstract
Microtubule “dynamic instability,” the abrupt switching from assembly to disassembly caused by the hydrolysis of GTP to GDP within the β subunit of the αβ-tubulin heterodimer, is necessary for vital cellular processes such as mitosis and migration. Despite existing high-resolution structural data, the key mechanochemical differences between the GTP and GDP states that mediate dynamic instability behavior remain unclear. Starting with a published atomic-level structure as an input, we used multiscale modeling to find that GTP hydrolysis results in both longitudinal bond weakening (~ 4 kBT) and an outward bending preference (~ 1.5 kBT) to both drive dynamic instability and give rise to the microtubule tip structures previously observed by light and electron microscopy. More generally, our study provides an example where atomic level structural information is used as the sole input to predict cellular level dynamics without parameter adjustment.
Collapse
|
17
|
Castle BT, McKibben KM, Rhoades E, Odde DJ. Tau Avoids the GTP Cap at Growing Microtubule Plus-Ends. iScience 2020; 23:101782. [PMID: 33294790 PMCID: PMC7691178 DOI: 10.1016/j.isci.2020.101782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 09/15/2020] [Accepted: 11/04/2020] [Indexed: 10/26/2022] Open
Abstract
Plus-end tracking proteins (+TIPs) associate with the growing end of microtubules and mediate important cellular functions. The majority of +TIPs are directed to the plus-end through a family of end-binding proteins (EBs), which preferentially bind the stabilizing cap of GTP-tubulin present during microtubule growth. One outstanding question is whether there may exist other microtubule-associated proteins (MAPs) that preferentially bind specific nucleotide states of tubulin. Here, we report that the neuronal MAP tau preferentially binds GDP-tubulin (K D = 0.26 μM) over GMPCPP-tubulin (K D = 1.1 μM) in vitro, as well as GTP-tubulin at the tips of growing microtubules, causing tau binding to lag behind the plus-end both in vitro and in live cells. Thus, tau is a microtubule tip avoiding protein, establishing the framework for a possible new class of tip avoiding MAPs. We speculate that disease-relevant tau mutations may exert their phenotype by their failure to properly recognize GDP-tubulin.
Collapse
Affiliation(s)
- Brian T. Castle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kristen M. McKibben
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth Rhoades
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David J. Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
18
|
Boiarska Z, Passarella D. Microtubule-targeting agents and neurodegeneration. Drug Discov Today 2020; 26:604-615. [PMID: 33279455 DOI: 10.1016/j.drudis.2020.11.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/17/2020] [Accepted: 11/28/2020] [Indexed: 11/25/2022]
Abstract
The association of microtubule (MT) breakdown with neurodegeneration and neurotoxicity has provided an emerging therapeutic approach for neurodegenerative diseases. Tubulin binders are able to modulate MT dynamics and, as a result, are of particular interest both as potential therapeutics and experimental tools used to validate this strategy. Here, we provide a comprehensive overview of current knowledge and recent advancements regarding MT-targeting approaches for neurodegeneration and evaluate the potential application of MT-targeting agents (MTAs) based on available preclinical and clinical data.
Collapse
Affiliation(s)
- Zlata Boiarska
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | - Daniele Passarella
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy.
| |
Collapse
|
19
|
Čermák V, Dostál V, Jelínek M, Libusová L, Kovář J, Rösel D, Brábek J. Microtubule-targeting agents and their impact on cancer treatment. Eur J Cell Biol 2020; 99:151075. [PMID: 32414588 DOI: 10.1016/j.ejcb.2020.151075] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/25/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
Microtubule-targeting agents (MTAs) constitute a diverse group of chemical compounds that bind to microtubules and affect their properties and function. Disruption of microtubules induces various cellular responses often leading to cell cycle arrest or cell death, the most common effect of MTAs. MTAs have found a plethora of practical applications in weed control, as fungicides and antiparasitics, and particularly in cancer treatment. Here we summarize the current knowledge of MTAs, the mechanisms of action and their role in cancer treatment. We further outline the potential use of MTAs in anti-metastatic therapy based on inhibition of cancer cell migration and invasiveness. The two main problems associated with cancer therapy by MTAs are high systemic toxicity and development of resistance. Toxic side effects of MTAs can be, at least partly, eliminated by conjugation of the drugs with various carriers. Moreover, some of the novel MTAs overcome the resistance mediated by both multidrug resistance transporters as well as overexpression of specific β-tubulin types. In anti-metastatic therapy, MTAs should be combined with other drugs to target all modes of cancer cell invasion.
Collapse
Affiliation(s)
- Vladimír Čermák
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Vojtěch Dostál
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic
| | - Michael Jelínek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lenka Libusová
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic
| | - Jan Kovář
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic.
| |
Collapse
|
20
|
Prahl LS, Stanslaski MR, Vargas P, Piel M, Odde DJ. Predicting Confined 1D Cell Migration from Parameters Calibrated to a 2D Motor-Clutch Model. Biophys J 2020; 118:1709-1720. [PMID: 32145191 PMCID: PMC7136340 DOI: 10.1016/j.bpj.2020.01.048] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/22/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Biological tissues contain micrometer-scale gaps and pores, including those found within extracellular matrix fiber networks, between tightly packed cells, and between blood vessels or nerve bundles and their associated basement membranes. These spaces restrict cell motion to a single-spatial dimension (1D), a feature that is not captured in traditional in vitro cell migration assays performed on flat, unconfined two-dimensional (2D) substrates. Mechanical confinement can variably influence cell migration behaviors, and it is presently unclear whether the mechanisms used for migration in 2D unconfined environments are relevant in 1D confined environments. Here, we assessed whether a cell migration simulator and associated parameters previously measured for cells on 2D unconfined compliant hydrogels could predict 1D confined cell migration in microfluidic channels. We manufactured microfluidic devices with narrow channels (60-μm2 rectangular cross-sectional area) and tracked human glioma cells that spontaneously migrated within channels. Cell velocities (vexp = 0.51 ± 0.02 μm min-1) were comparable to brain tumor expansion rates measured in the clinic. Using motor-clutch model parameters estimated from cells on unconfined 2D planar hydrogel substrates, simulations predicted similar migration velocities (vsim = 0.37 ± 0.04 μm min-1) and also predicted the effects of drugs targeting the motor-clutch system or cytoskeletal assembly. These results are consistent with glioma cells utilizing a motor-clutch system to migrate in confined environments.
Collapse
Affiliation(s)
- Louis S Prahl
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Maria R Stanslaski
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Pablo Vargas
- Institut Curie, PSL Research University, CNRS UMR 144 and Institut Pierre-Gilles de Gennes, PSL Research University, Paris, France; INSERM U932 Immunité et Cancer, Institut Curie, PSL Research University, Paris, France
| | - Matthieu Piel
- Institut Curie, PSL Research University, CNRS UMR 144 and Institut Pierre-Gilles de Gennes, PSL Research University, Paris, France
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Physical Sciences-Oncology Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
21
|
Rai A, Liu T, Glauser S, Katrukha EA, Estévez-Gallego J, Rodríguez-García R, Fang WS, Díaz JF, Steinmetz MO, Altmann KH, Kapitein LC, Moores CA, Akhmanova A. Taxanes convert regions of perturbed microtubule growth into rescue sites. NATURE MATERIALS 2020; 19:355-365. [PMID: 31819210 DOI: 10.1038/s41563-019-0546-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 10/24/2019] [Indexed: 06/10/2023]
Abstract
Microtubules are polymers of tubulin dimers, and conformational transitions in the microtubule lattice drive microtubule dynamic instability and affect various aspects of microtubule function. The exact nature of these transitions and their modulation by anticancer drugs such as Taxol and epothilone, which can stabilize microtubules but also perturb their growth, are poorly understood. Here, we directly visualize the action of fluorescent Taxol and epothilone derivatives and show that microtubules can transition to a state that triggers cooperative drug binding to form regions with altered lattice conformation. Such regions emerge at growing microtubule ends that are in a pre-catastrophe state, and inhibit microtubule growth and shortening. Electron microscopy and in vitro dynamics data indicate that taxane accumulation zones represent incomplete tubes that can persist, incorporate tubulin dimers and repeatedly induce microtubule rescues. Thus, taxanes modulate the material properties of microtubules by converting destabilized growing microtubule ends into regions resistant to depolymerization.
Collapse
Affiliation(s)
- Ankit Rai
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Tianyang Liu
- Institute of Structural and Molecular Biology, Birkbeck, University of London, London, UK
| | - Simon Glauser
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| | - Eugene A Katrukha
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Juan Estévez-Gallego
- Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Ruddi Rodríguez-García
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Wei-Shuo Fang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Beijing, China
| | - J Fernando Díaz
- Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
- University of Basel, Biozentrum, Basel, Switzerland
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, Birkbeck, University of London, London, UK
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
22
|
Numerical Parameter Space Compression and Its Application to Biophysical Models. Biophys J 2020; 118:1455-1465. [PMID: 32070477 DOI: 10.1016/j.bpj.2020.01.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/07/2019] [Accepted: 01/14/2020] [Indexed: 11/20/2022] Open
Abstract
Physical models of biological systems can become difficult to interpret when they have a large number of parameters. But the models themselves actually depend on (i.e., are sensitive to) only a subset of those parameters. This phenomenon is due to parameter space compression (PSC), in which a subset of parameters emerges as "stiff" as a function of time or space. PSC has only been used to explain analytically solvable physics models. We have generalized this result by developing a numerical approach to PSC that can be applied to any computational model. We validated our method against analytically solvable models of a random walk with drift and protein production and degradation. We then applied our method to a simple computational model of microtubule dynamic instability. We propose that numerical PSC has the potential to identify the low-dimensional structure of many computational models in biophysics. The low-dimensional structure of a model is easier to interpret and identifies the mechanisms and experiments that best characterize the system.
Collapse
|
23
|
Genualdi C, Feinstein S, Wilson L, Jordan M, Stagg N. Assessing the utility of in vitro microtubule assays for studying mechanisms of peripheral neuropathy with the microtubule inhibitor class of cancer chemotherapy. Chem Biol Interact 2020; 315:108906. [DOI: 10.1016/j.cbi.2019.108906] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/16/2019] [Accepted: 11/18/2019] [Indexed: 01/28/2023]
|
24
|
Prahl LS, Bangasser PF, Stopfer LE, Hemmat M, White FM, Rosenfeld SS, Odde DJ. Microtubule-Based Control of Motor-Clutch System Mechanics in Glioma Cell Migration. Cell Rep 2019; 25:2591-2604.e8. [PMID: 30485822 PMCID: PMC6345402 DOI: 10.1016/j.celrep.2018.10.101] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 09/25/2018] [Accepted: 10/26/2018] [Indexed: 11/30/2022] Open
Abstract
Microtubule-targeting agents (MTAs) are widely used chemotherapy drugs capable of disrupting microtubule-dependent cellular functions, such as division and migration. We show that two clinically approved MTAs, paclitaxel and vinblastine, each suppress stiffness-sensitive migration and polarization characteristic of human glioma cells on compliant hydrogels. MTAs influence microtubule dynamics and cell traction forces by nearly opposite mechanisms, the latter of which can be explained by a combination of changes in myosin motor and adhesion clutch number. Our results support a microtubule-dependent signaling-based model for controlling traction forces through a motor-clutch mechanism, rather than microtubules directly relieving tension within F-actin and adhesions. Computational simulations of cell migration suggest that increasing protrusion number also impairs stiffness-sensitive migration, consistent with experimental MTA effects. These results provide a theoretical basis for the role of microtubules and mechanisms of MTAs in controlling cell migration. Prahl et al. examine the mechanisms by which microtubule-targeting drugs inhibit glioma cell migration. They find that dynamic microtubules regulate actin-based protrusion dynamics that facilitate cell polarity and migration. Changes in net microtubule assembly alter cell traction forces via signaling-based regulation of a motor-clutch system.
Collapse
Affiliation(s)
- Louis S Prahl
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Physical Sciences-Oncology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Patrick F Bangasser
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Physical Sciences-Oncology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lauren E Stopfer
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research and Physical Sciences-Oncology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mahya Hemmat
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Forest M White
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research and Physical Sciences-Oncology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Steven S Rosenfeld
- Physical Sciences-Oncology Center, University of Minnesota, Minneapolis, MN 55455, USA; Brain Tumor and Neuro-Oncology Center and Department of Cancer Biology, Cleveland Clinic, Cleveland, OH 44195, USA
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Physical Sciences-Oncology Center, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
25
|
Hahn I, Voelzmann A, Liew YT, Costa-Gomes B, Prokop A. The model of local axon homeostasis - explaining the role and regulation of microtubule bundles in axon maintenance and pathology. Neural Dev 2019; 14:11. [PMID: 31706327 PMCID: PMC6842214 DOI: 10.1186/s13064-019-0134-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Axons are the slender, cable-like, up to meter-long projections of neurons that electrically wire our brains and bodies. In spite of their challenging morphology, they usually need to be maintained for an organism's lifetime. This makes them key lesion sites in pathological processes of ageing, injury and neurodegeneration. The morphology and physiology of axons crucially depends on the parallel bundles of microtubules (MTs), running all along to serve as their structural backbones and highways for life-sustaining cargo transport and organelle dynamics. Understanding how these bundles are formed and then maintained will provide important explanations for axon biology and pathology. Currently, much is known about MTs and the proteins that bind and regulate them, but very little about how these factors functionally integrate to regulate axon biology. As an attempt to bridge between molecular mechanisms and their cellular relevance, we explain here the model of local axon homeostasis, based on our own experiments in Drosophila and published data primarily from vertebrates/mammals as well as C. elegans. The model proposes that (1) the physical forces imposed by motor protein-driven transport and dynamics in the confined axonal space, are a life-sustaining necessity, but pose a strong bias for MT bundles to become disorganised. (2) To counterbalance this risk, MT-binding and -regulating proteins of different classes work together to maintain and protect MT bundles as necessary transport highways. Loss of balance between these two fundamental processes can explain the development of axonopathies, in particular those linking to MT-regulating proteins, motors and transport defects. With this perspective in mind, we hope that more researchers incorporate MTs into their work, thus enhancing our chances of deciphering the complex regulatory networks that underpin axon biology and pathology.
Collapse
Affiliation(s)
- Ines Hahn
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - André Voelzmann
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Yu-Ting Liew
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Beatriz Costa-Gomes
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Andreas Prokop
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK.
| |
Collapse
|
26
|
Reid TA, Coombes C, Mukherjee S, Goldblum RR, White K, Parmar S, McClellan M, Zanic M, Courtemanche N, Gardner MK. Structural state recognition facilitates tip tracking of EB1 at growing microtubule ends. eLife 2019; 8:48117. [PMID: 31478831 PMCID: PMC6742484 DOI: 10.7554/elife.48117] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/23/2019] [Indexed: 11/13/2022] Open
Abstract
The microtubule binding protein EB1 specifically targets the growing ends of microtubules in cells, where EB1 facilitates the interactions of cellular proteins with microtubule plus-ends. Microtubule end targeting of EB1 has been attributed to high-affinity binding of EB1 to GTP-tubulin that is present at growing microtubule ends. However, our 3D single-molecule diffusion simulations predicted a ~ 6000% increase in EB1 arrivals to open, tapered microtubule tip structures relative to closed lattice conformations. Using quantitative fluorescence, single-molecule, and electron microscopy experiments, we found that the binding of EB1 onto opened, structurally disrupted microtubules was dramatically increased relative to closed, intact microtubules, regardless of hydrolysis state. Correspondingly, in cells, the blunting of growing microtubule plus-ends by Vinblastine was correlated with reduced EB1 targeting. Together, our results suggest that microtubule structural recognition, based on a fundamental diffusion-limited binding model, facilitates the tip tracking of EB1 at growing microtubule ends.
Collapse
Affiliation(s)
- Taylor A Reid
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Courtney Coombes
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Soumya Mukherjee
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Rebecca R Goldblum
- Medical Scientist Training Program, University of Minnesota, Minneapolis, United States.,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, United States
| | - Kyle White
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Sneha Parmar
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Mark McClellan
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Marija Zanic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Naomi Courtemanche
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Melissa K Gardner
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| |
Collapse
|
27
|
Hemmat M, Castle BT, Sachs JN, Odde DJ. Multiscale Computational Modeling of Tubulin-Tubulin Lateral Interaction. Biophys J 2019; 117:1234-1249. [PMID: 31493861 DOI: 10.1016/j.bpj.2019.08.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/26/2019] [Accepted: 08/13/2019] [Indexed: 10/26/2022] Open
Abstract
Microtubules are multistranded polymers in eukaryotic cells that support key cellular functions such as chromosome segregation, motor-based cargo transport, and maintenance of cell polarity. Microtubules self-assemble via "dynamic instability," in which the dynamic plus ends switch stochastically between alternating phases of polymerization and depolymerization. A key question in the field is what are the atomistic origins of this switching, i.e., what is different between the GTP- and GDP-tubulin states that enables microtubule growth and shortening, respectively? More generally, a major challenge in biology is how to connect theoretical frameworks across length- and timescales, from atoms to cellular behavior. In this study, we describe a multiscale model by linking atomistic molecular dynamics (MD), molecular Brownian dynamics (BD), and cellular-level thermokinetic modeling of microtubules. Here, we investigated the underlying interaction energy when tubulin dimers associate laterally by performing all-atom MD simulations. We found that the lateral potential energy is not significantly different among three nucleotide states of tubulin, GTP, GDP, and GMPCPP and is estimated to be ≅ -11 kBT. Furthermore, using MD potential energy in our BD simulations of tubulin dimers confirms that the lateral bond is weak on its own, with a mean lifetime of ∼0.1 μs, implying that the longitudinal bond is required for microtubule assembly. We conclude that nucleotide-dependent lateral-bond strength is not the key mediator microtubule dynamic instability, implying that GTP acts elsewhere to exert its stabilizing influence on microtubule polymer. Furthermore, the estimated lateral-bond strength (ΔGlat0≅ -5 kBT) is well-aligned with earlier estimates based on thermokinetic modeling and light microscopy measurements. Thus, we have computationally connected atomistic-level structural information, obtained by cryo-electron microscopy, to cellular-scale microtubule assembly dynamics using a combination of MD, BD, and thermokinetic models to bridge from Ångstroms to micrometers and from femtoseconds to minutes.
Collapse
Affiliation(s)
- Mahya Hemmat
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Brian T Castle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Jonathan N Sachs
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
28
|
Mustyatsa VV, Kostarev AV, Tvorogova AV, Ataullakhanov FI, Gudimchuk NB, Vorobjev IA. Fine structure and dynamics of EB3 binding zones on microtubules in fibroblast cells. Mol Biol Cell 2019; 30:2105-2114. [PMID: 31141458 PMCID: PMC6743451 DOI: 10.1091/mbc.e18-11-0723] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/14/2019] [Accepted: 05/21/2019] [Indexed: 11/26/2022] Open
Abstract
End-binding (EB) proteins associate with the growing tips of microtubules (MTs)and modulate their dynamics directly and indirectly, by recruiting essential factors to fine-tune MTs for their many essential roles in cells. Previously EB proteins have been shown to recognize a stabilizing GTP/GDP-Pi cap at the tip of growing MTs, but information about additional EB-binding zones on MTs has been limited. In this work, we studied fluorescence intensity profiles of one of the three mammalian EB-proteins, EB3, fused with red fluorescent protein (RFP). The distribution of EB3 on MTs in mouse fibroblasts frequently deviated from single exponential decay and exhibited secondary peaks. Those secondary peaks, which we refer to as EB3-islands, were detected on 56% comets of growing MTs and were encountered once per 44 s of EB3-RFP comet growth time with about 5 s half-lifetime. The majority of EB3-islands in the vicinity of MT tips was stationary and originated from EB3 comets moving with the growing MT tips. Computational modeling of the decoration of dynamic MT tips by EB3 suggested that the EB3-islands could not be explained simply by a stochastic first-order GTP hydrolysis/phosphate release. We speculate that additional protein factors contribute to EB3 residence time on MTs in cells, likely affecting MT dynamics.
Collapse
Affiliation(s)
- V. V. Mustyatsa
- Lomonosov Moscow State University, 119991 Moscow, Russia
- Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, 117198 Moscow, Russia
| | - A. V. Kostarev
- Lomonosov Moscow State University, 119991 Moscow, Russia
| | | | - F. I. Ataullakhanov
- Lomonosov Moscow State University, 119991 Moscow, Russia
- Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, 117198 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - N. B. Gudimchuk
- Lomonosov Moscow State University, 119991 Moscow, Russia
- Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, 117198 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - I. A. Vorobjev
- Lomonosov Moscow State University, 119991 Moscow, Russia
- Nazarbayev University, 010000 Nur-Sultan, Kazakhstan
| |
Collapse
|
29
|
Leung JC, Cassimeris L. Reorganization of paclitaxel-stabilized microtubule arrays at mitotic entry: roles of depolymerizing kinesins and severing proteins. Cancer Biol Ther 2019; 20:1337-1347. [PMID: 31345098 PMCID: PMC6783116 DOI: 10.1080/15384047.2019.1638678] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Paclitaxel is a widely used anti-cancer treatment that disrupts cell cycle progression by blocking cells in mitosis. The block at mitosis, with spindles assembled from short microtubules, is surprising given paclitaxel’s microtubule stabilizing activity and the need to depolymerize long interphase microtubules prior to spindle formation. Cells must antagonize paclitaxel’s microtubule stabilizing activity during a brief window of time at the transition from interphase to mitosis, allowing microtubule reorganization into a mitotic spindle, although the mechanism underlying microtubule depolymerization in the presence of paclitaxel has not been examined. Here we test the hypothesis that microtubule severing and/or depolymerizing proteins active at mitotic entry are necessary to clear the interphase array in paclitaxel-treated cells and allow subsequent formation of mitotic spindles formed of short microtubules. A549 and LLC-PK1 cells treated with 30nM paclitaxel approximately 4 h prior to mitotic entry successfully progress through the G2/M transition by clearing the interphase microtubule array from the cell interior outward to the cell periphery, a spatial pattern of reorganization that differs from that of cells possessing dynamic microtubules. Depletion of kinesin-8s, KIF18A and/or KIF18B obstructed interphase microtubule clearing at mitotic entry in paclitaxel-treated cells, with KIF18B making the larger contribution. Of the severing proteins, depletion of spastin, but not katanin, reduced microtubule loss as cells entered mitosis in the presence of paclitaxel. These results support a model in which KIF18A, KIF18B, and spastin promote interphase microtubule array disassembly at mitotic entry and can overcome paclitaxel-induced microtubule stability specifically at the G2/M transition.
Collapse
Affiliation(s)
- Jessica C Leung
- Department of Biological Sciences, 111 Research Dr. Lehigh University , Bethlehem , PA , USA
| | - Lynne Cassimeris
- Department of Biological Sciences, 111 Research Dr. Lehigh University , Bethlehem , PA , USA
| |
Collapse
|
30
|
Qiang L, Piermarini E, Muralidharan H, Yu W, Leo L, Hennessy LE, Fernandes S, Connors T, Yates PL, Swift M, Zholudeva LV, Lane MA, Morfini G, Alexander GM, Heiman-Patterson TD, Baas PW. Hereditary spastic paraplegia: gain-of-function mechanisms revealed by new transgenic mouse. Hum Mol Genet 2019; 28:1136-1152. [PMID: 30520996 DOI: 10.1093/hmg/ddy419] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 10/31/2018] [Accepted: 12/02/2018] [Indexed: 12/17/2022] Open
Abstract
Mutations of the SPAST gene, which encodes the microtubule-severing protein spastin, are the most common cause of hereditary spastic paraplegia (HSP). Haploinsufficiency is the prevalent opinion as to the mechanism of the disease, but gain-of-function toxicity of the mutant proteins is another possibility. Here, we report a new transgenic mouse (termed SPASTC448Y mouse) that is not haploinsufficient but expresses human spastin bearing the HSP pathogenic C448Y mutation. Expression of the mutant spastin was documented from fetus to adult, but gait defects reminiscent of HSP (not observed in spastin knockout mice) were adult onset, as is typical of human patients. Results of histological and tracer studies on the mouse are consistent with progressive dying back of corticospinal axons, which is characteristic of the disease. The C448Y-mutated spastin alters microtubule stability in a manner that is opposite to the expectations of haploinsufficiency. Neurons cultured from the mouse display deficits in organelle transport typical of axonal degenerative diseases, and these deficits were worsened by depletion of endogenous mouse spastin. These results on the SPASTC448Y mouse are consistent with a gain-of-function mechanism underlying HSP, with spastin haploinsufficiency exacerbating the toxicity of the mutant spastin proteins. These findings reveal the need for a different therapeutic approach than indicated by haploinsufficiency alone.
Collapse
Affiliation(s)
| | | | | | | | | | - Laura E Hennessy
- Department of Neurology, Drexel University College of Medicine, Queen Lane, Philadelphia, PA, USA
| | | | | | | | | | | | | | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Guillermo M Alexander
- Department of Neurology, Drexel University College of Medicine, Queen Lane, Philadelphia, PA, USA
| | - Terry D Heiman-Patterson
- Department of Neurology, Drexel University College of Medicine, Queen Lane, Philadelphia, PA, USA
| | | |
Collapse
|
31
|
Hafner AE, Krausser J, Šarić A. Minimal coarse-grained models for molecular self-organisation in biology. Curr Opin Struct Biol 2019; 58:43-52. [PMID: 31226513 DOI: 10.1016/j.sbi.2019.05.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/13/2019] [Accepted: 05/19/2019] [Indexed: 01/19/2023]
Abstract
The molecular machinery of life is largely created via self-organisation of individual molecules into functional assemblies. Minimal coarse-grained models, in which a whole macromolecule is represented by a small number of particles, can be of great value in identifying the main driving forces behind self-organisation in cell biology. Such models can incorporate data from both molecular and continuum scales, and their results can be directly compared to experiments. Here we review the state of the art of models for studying the formation and biological function of macromolecular assemblies in living organisms. We outline the key ingredients of each model and their main findings. We illustrate the contribution of this class of simulations to identifying the physical mechanisms behind life and diseases, and discuss their future developments.
Collapse
Affiliation(s)
- Anne E Hafner
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London WC1E 6BT, UK
| | - Johannes Krausser
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London WC1E 6BT, UK
| | - Anđela Šarić
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London WC1E 6BT, UK.
| |
Collapse
|
32
|
Microtubule Polymerization and Cross-Link Dynamics Explain Axonal Stiffness and Damage. Biophys J 2019; 114:201-212. [PMID: 29320687 DOI: 10.1016/j.bpj.2017.11.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/27/2017] [Accepted: 11/10/2017] [Indexed: 01/02/2023] Open
Abstract
Axonal damage is a critical indicator for traumatic effects of physical impact to the brain. However, the precise mechanisms of axonal damage are still unclear. Here, we establish a mechanistic and highly dynamic model of the axon to explore the evolution of damage in response to physical forces. Our axon model consists of a bundle of dynamically polymerizing and depolymerizing microtubules connected by dynamically detaching and reattaching cross-links. Although the probability of cross-link attachment depends exclusively on thermal fluctuations, the probability of detachment increases in the presence of physical forces. We systematically probe the landscape of axonal stretch and stretch rate and characterize the overall axonal force, stiffness, and damage as a direct result of the interplay between microtubule and cross-link dynamics. Our simulations reveal that slow loading is dominated by cross-link dynamics, a net reduction of cross-links, and a gradual accumulation of damage, whereas fast loading is dominated by cross-link deformations, a rapid increase in stretch, and an immediate risk of rupture. Microtubule polymerization and depolymerization decrease the overall axonal stiffness, but do not affect the evolution of damage at timescales relevant to axonal failure. Our study explains different failure mechanisms in the axon as emergent properties of microtubule polymerization, cross-link dynamics, and physical forces. We anticipate that our model will provide insight into causal relations by which molecular mechanisms determine the timeline and severity of axon damage after a physical impact to the brain.
Collapse
|
33
|
Qiu K, Liu C, Shi Y, Yu X, Chen G, Wu J, Li G, Lv L. An LC-MS/MS Method for Synchronous Determination of Paclitaxel and Curcumin: Development, Validation, and Application to a Pharmacokinetic Study. CURR PHARM ANAL 2019. [DOI: 10.2174/1573412914666180222140839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background:
A chromatography tandem mass spectrometry method was first established and
validated for the synchronous determination of curcumin(CUR) and paclitaxel (PTX) in this study.
</P><P>
Objective: An LC-MS/MS Method for Determination of Paclitaxel and Curcumin.
Methods:
The analytes were extracted with methanol, and docetaxel was used as the internal standard
(IS). The analytes and the IS were separated on a C18 (4.6 mm × 50 mm, 3.5 µm) column with a mobile
phase of 0.1% formic acid solution and methanol (80:20, v/v). The flow velocity of the mobile phase
was 0.5 mL/min. And then, the method was applied to study the pharmacokinetic behavior of CUR and
PTX in rats.
Results:
The calibration curves were linear within the concentration ranges of 2–1000 ng/mL for PTX
and 5–500 ng/mL for CUR, the mean extraction recoveries and matrix effects of PTX, CUR, and the IS
were within an acceptable range. The apparent volume of distribution of PTX was different between the
group of administration of PTX and the group of co-administration with CUR and PTX.
Conclusion:
A sensitive and simple liquid chromatography-tandem mass spectrometry method was
established and validated for the synchronous determination of PTX and CUR in rat plasma, CUR increased
the apparent volume of distribution of PTX when CUR and PTX were co-administered.
Collapse
Affiliation(s)
- Kaifeng Qiu
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| | - Chunxia Liu
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| | - Yonghui Shi
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| | - Xiaoxia Yu
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| | - Guanghui Chen
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| | - Junyan Wu
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| | - Guocheng Li
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| | - Li Lv
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, China
| |
Collapse
|
34
|
Castle BT, Odde DJ, Wood DK. Rapid and inefficient kinetics of sickle hemoglobin fiber growth. SCIENCE ADVANCES 2019; 5:eaau1086. [PMID: 30891490 PMCID: PMC6415962 DOI: 10.1126/sciadv.aau1086] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 01/29/2019] [Indexed: 05/22/2023]
Abstract
In sickle cell disease, the aberrant assembly of hemoglobin fibers induces changes in red blood cell morphology and stiffness, which leads to downstream symptoms of the disease. Therefore, understanding of this assembly process will be important for the treatment of sickle cell disease. By performing the highest spatiotemporal resolution measurements (55 nm at 1 Hz) of single sickle hemoglobin fiber assembly to date and combining them with a model that accounts for the multistranded structure of the fibers, we show that the rates of sickle hemoglobin addition and loss have been underestimated in the literature by at least an order of magnitude. These results reveal that the sickle hemoglobin self-assembly process is very rapid and inefficient (4% efficient versus 96% efficient based on previous analyses), where net growth is the small difference between over a million addition-loss events occurring every second.
Collapse
|
35
|
Serikbaeva A, Tvorogova A, Kauanova S, Vorobjev IA. Analysis of Microtubule Dynamics Heterogeneity in Cell Culture. Methods Mol Biol 2019; 1745:181-204. [PMID: 29476470 DOI: 10.1007/978-1-4939-7680-5_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Microtubules (MTs) are dynamic components of the cytoskeleton playing an important role in a large number of cell functions. Individual MTs in living cells undergo stochastic switching between alternate states of growth, shortening and attenuated phase, a phenomenon known as tempered dynamic instability. Dynamic instability of MTs is usually analyzed by labeling MTs with +TIPs, namely, EB proteins. Tracking of +TIP trajectories allows analyzing MT growth in cells with a different density of MTs. Numerous labs now use +TIP to track growing MTs in a variety of cell cultures. However, heterogeneity of MT dynamics is usually underestimated, and rather small sampling for the description of dynamic instability parameters is often used. The strategy described in this chapter is the method for repetitive quantitative analysis of MT growth rate within the same cell that allows minimization of the variation in MT dynamics measurement. We show that variability in MT dynamics within a cell when using repeated measurements is significantly less than between different cells in the same chamber. This approach allows better estimation of the heterogeneity of cells' responses to different treatments. To compare the effects of different MT inhibitors, the protocol using normalized values for MT dynamics and repetitive measurements for each cell is employed. This chapter provides detailed methods for analysis of MT dynamics in tissue cultures. We describe protocols for imaging MT dynamics by fluorescent microscopy, contrast enhancement technique, and MT dynamics analysis using triple color-coded display based on sequential subtraction analysis.
Collapse
Affiliation(s)
- Anara Serikbaeva
- Department of Biology, School of Science and Technology, Nazarbayev University, Astana, Kazakhstan
- Department of Biology, School of Sciences and Technology, Nazarbayev University, Astana, Kazakhstan
| | - Anna Tvorogova
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Sholpan Kauanova
- School of Engineering, Nazarbayev University, Astana, Kazakhstan
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Ivan A Vorobjev
- Department of Biology, School of Sciences and Technology, Nazarbayev University, Astana, Kazakhstan.
| |
Collapse
|
36
|
Basic principles of drug delivery systems - the case of paclitaxel. Adv Colloid Interface Sci 2019; 263:95-130. [PMID: 30530177 DOI: 10.1016/j.cis.2018.11.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 01/15/2023]
Abstract
Cancer is the second cause of death worldwide, exceeded only by cardiovascular diseases. The prevalent treatment currently used against metastatic cancer is chemotherapy. Among the most studied drugs that inhibit neoplastic cells from acquiring unlimited replicative ability (a hallmark of cancer) are the taxanes. They operate via a unique molecular mechanism affecting mitosis. In this review, we show this mechanism for one of them, paclitaxel, and for other (non-taxanes) anti-mitotic drugs. However, the use of paclitaxel is seriously limited (its bioavailability is <10%) due to several long-standing challenges: its poor water solubility (0.3 μg/mL), its being a substrate for the efflux multidrug transporter P-gp, and, in the case of oral delivery, its first-pass metabolism by certain enzymes. Adequate delivery methods are therefore required to enhance the anti-tumor activity of paclitaxel. Thus, we have also reviewed drug delivery strategies in light of the various physical, chemical, and enzymatic obstacles facing the (especially oral) delivery of drugs in general and paclitaxel in particular. Among the powerful and versatile platforms that have been developed and achieved unprecedented opportunities as drug carriers, microemulsions might have great potential for this aim. This is due to properties such as thermodynamic stability (leading to long shelf-life), increased drug solubilization, and ease of preparation and administration. In this review, we define microemulsions and nanoemulsions, analyze their pertinent properties, and review the results of several drug delivery carriers based on these systems.
Collapse
|
37
|
Porcelli L, Stolfa D, Stefanachi A, Di Fonte R, Garofoli M, Iacobazzi RM, Silvestris N, Guarini A, Cellamare S, Azzariti A. Synthesis and biological evaluation of N-biphenyl-nicotinic based moiety compounds: A new class of antimitotic agents for the treatment of Hodgkin Lymphoma. Cancer Lett 2018; 445:1-10. [PMID: 30583077 DOI: 10.1016/j.canlet.2018.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022]
Abstract
We previously demonstrated that some N-biphenylanilides caused cell-cycle arrest at G2/M transition in breast cancer cells. Among them we choose three derivatives, namely PTA34, PTA73 and RS35 for experimentation in solid tumor cell lines, classical Hodgkin Lymphoma (cHL) cell lines and bona fide normal cell lines. Almost all tumor cells were sensitive to compounds in the nanomolar range whereas, they were not cytotoxic to normal ones. Interestingly the compounds caused a strong G2/M phase arrest in cHL cell lines, thus, here we investigated whether they affected the integrity of microtubules in such cells. We found that they induced a long prometaphase arrest, followed by induction of apoptosis which involved mitochondria. PTA73 and RS35 induced the mitotic arrest through the fragmentation of microtubules which prevented the kinethocore-mitotic spindle interaction and the exit from mitosis. PTA34 is instead a tubulin-targeting agent because it inhibited the tubulin polymerization as vinblastine. As such, PTA34 maintained the Cyclin B1-CDK1 regulatory complex activated during the G2/M arrest while inducing the inactivation of Bcl-2 through phosphorylation in Ser70, the degradation of Mcl-1 and a strong activation of BIML and BIMS proapoptotic isoforms. In addition PTA34 exerted an antiangiogenic effect by suppressing microvascular formation.
Collapse
Affiliation(s)
- L Porcelli
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - D Stolfa
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy; Dipartimento di Farmacia-Scienza del Farmaco, Università di Bari, Bari, Italy
| | - A Stefanachi
- Dipartimento di Farmacia-Scienza del Farmaco, Università di Bari, Bari, Italy
| | - R Di Fonte
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - M Garofoli
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - R M Iacobazzi
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - N Silvestris
- Medical Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - A Guarini
- Haematology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - S Cellamare
- Dipartimento di Farmacia-Scienza del Farmaco, Università di Bari, Bari, Italy
| | - A Azzariti
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy.
| |
Collapse
|
38
|
Microtubule lattice plasticity. Curr Opin Cell Biol 2018; 56:88-93. [PMID: 30415187 DOI: 10.1016/j.ceb.2018.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/21/2018] [Indexed: 01/06/2023]
Abstract
In classical microtubule dynamic instability, the dynamics of the built polymer depend only on the nucleotide state of its individual tubulin molecules. Recent work is overturning this view, pointing instead towards lattice plasticity, in which the fine-structure and mechanics of the microtubule lattice are emergent properties that depend not only on the nucleotide state of each tubulin, but also on the nucleotide states of its neighbours, on its and their isotypes, and on interacting proteins, drugs, local mechanical strain, post translational modifications, packing defects and solvent conditions. In lattice plasticity models, the microtubule is an allosteric molecular collective that integrates multiple mechanochemical inputs and responds adaptively by adjusting its conformation, stiffness and dynamics.
Collapse
|
39
|
Chaaban S, Jariwala S, Hsu CT, Redemann S, Kollman JM, Müller-Reichert T, Sept D, Bui KH, Brouhard GJ. The Structure and Dynamics of C. elegans Tubulin Reveals the Mechanistic Basis of Microtubule Growth. Dev Cell 2018; 47:191-204.e8. [PMID: 30245157 DOI: 10.1016/j.devcel.2018.08.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/06/2018] [Accepted: 08/23/2018] [Indexed: 01/04/2023]
Abstract
The dynamic instability of microtubules is a conserved and fundamental mechanism in eukaryotes. Yet microtubules from different species diverge in their growth rates, lattice structures, and responses to GTP hydrolysis. Therefore, we do not know what limits microtubule growth, what determines microtubule structure, or whether the mechanisms of dynamic instability are universal. Here, we studied microtubules from the nematode C. elegans, which have strikingly fast growth rates and non-canonical lattices in vivo. Using a reconstitution approach, we discovered that C. elegans microtubules combine intrinsically fast growth with very frequent catastrophes. We solved the structure of C. elegans microtubules to 4.8 Å and discovered sequence divergence in the lateral contact loops, one of which is ordered in C. elegans but unresolved in other species. We provide direct evidence that C. elegans tubulin has a higher free energy in solution and propose a model wherein the ordering of lateral contact loops activates tubulin for growth.
Collapse
Affiliation(s)
- Sami Chaaban
- Department of Biology, 1205 Avenue Docteur Penfield, Montréal, QC H3A 1B1, Canada
| | - Shashank Jariwala
- Department of Computational Medicine and Bioinformatics, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA
| | - Chieh-Ting Hsu
- Department of Biology, 1205 Avenue Docteur Penfield, Montréal, QC H3A 1B1, Canada
| | - Stefanie Redemann
- Experimental Center, Technische Universität Dresden, Faculty of Medicine, Fiedlerstraße 42, 01307 Dresden, Germany; Center for Membrane & Cell Physiology, University of Virginia and Department of Molecular Physiology & Biological Physics, 480 Ray C. Hung Drive, Charlottesville, VA 22903, USA
| | - Justin M Kollman
- Department of Biochemistry, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Thomas Müller-Reichert
- Experimental Center, Technische Universität Dresden, Faculty of Medicine, Fiedlerstraße 42, 01307 Dresden, Germany
| | - David Sept
- Department of Biomedical Engineering, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109, USA
| | - Khanh Huy Bui
- Department of Anatomy and Cell Biology, 3640 Rue University, Montréal, QC H3A 0C7, Canada
| | - Gary J Brouhard
- Department of Biology, 1205 Avenue Docteur Penfield, Montréal, QC H3A 1B1, Canada.
| |
Collapse
|
40
|
Cong H, Zhao X, Castle BT, Pomeroy EJ, Zhou B, Lee J, Wang Y, Bian T, Miao Z, Zhang W, Sham YY, Odde DJ, Eckfeldt CE, Xing C, Zhuang C. An Indole-Chalcone Inhibits Multidrug-Resistant Cancer Cell Growth by Targeting Microtubules. Mol Pharm 2018; 15:3892-3900. [PMID: 30048137 DOI: 10.1021/acs.molpharmaceut.8b00359] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Multidrug resistance and toxic side effects are the major challenges in cancer treatment with microtubule-targeting agents (MTAs), and thus, there is an urgent clinical need for new therapies. Chalcone, a common simple scaffold found in many natural products, is widely used as a privileged structure in medicinal chemistry. We have previously validated tubulin as the anticancer target for chalcone derivatives. In this study, an α-methyl-substituted indole-chalcone (FC77) was synthesized and found to exhibit an excellent cytotoxicity against the NCI-60 cell lines (average concentration causing 50% growth inhibition = 6 nM). More importantly, several multidrug-resistant cancer cell lines showed no resistance to FC77, and the compound demonstrated good selective toxicity against cancer cells versus normal CD34+ blood progenitor cells. A further mechanistic study demonstrated that FC77 could arrest cells that relate to the binding to tubulin and inhibit the microtubule dynamics. The National Cancer Institute COMPARE analysis and molecular modeling indicated that FC77 had a mechanism of action similar to that of colchicine. Overall, our data demonstrate that this indole-chalcone represents a novel MTA template for further development of potential drug candidates for the treatment of multidrug-resistant cancers.
Collapse
Affiliation(s)
- Hui Cong
- School of Pharmacy , Ningxia Medical University , Yinchuan , China
| | - Xinghua Zhao
- Department of Medicinal Chemistry , University of Minnesota , Minneapolis , Minnesota 55455 , United States.,College of Veterinary Medicine , Hebei Agricultural University , Baoding , China
| | - Brian T Castle
- Department of Biomedical Engineering , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Emily J Pomeroy
- Department of Medicine, Division of Hematology, Oncology, and Transplantation , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Bo Zhou
- Department of Medicinal Chemistry , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - John Lee
- Department of Biochemistry, Molecular Biology and Biophysics , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Yi Wang
- Department of Medicinal Chemistry , University of Florida , Gainesville , Florida 32611 , United States
| | - Tengfei Bian
- Department of Medicinal Chemistry , University of Florida , Gainesville , Florida 32611 , United States
| | - Zhenyuan Miao
- School of Pharmacy , Second Military Medical University , Shanghai , China
| | - Wannian Zhang
- School of Pharmacy , Ningxia Medical University , Yinchuan , China.,School of Pharmacy , Second Military Medical University , Shanghai , China
| | - Yuk Yin Sham
- Department of Integrative Biology and Physiology , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - David J Odde
- Department of Biomedical Engineering , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Craig E Eckfeldt
- Department of Medicine, Division of Hematology, Oncology, and Transplantation , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Chengguo Xing
- School of Pharmacy , Ningxia Medical University , Yinchuan , China.,Department of Medicinal Chemistry , University of Minnesota , Minneapolis , Minnesota 55455 , United States.,Department of Medicinal Chemistry , University of Florida , Gainesville , Florida 32611 , United States
| | - Chunlin Zhuang
- School of Pharmacy , Ningxia Medical University , Yinchuan , China.,School of Pharmacy , Second Military Medical University , Shanghai , China
| |
Collapse
|
41
|
Abstract
Melanoma represents the most aggressive and the deadliest form of skin cancer. Current therapeutic approaches include surgical resection, chemotherapy, photodynamic therapy, immunotherapy, biochemotherapy, and targeted therapy. The therapeutic strategy can include single agents or combined therapies, depending on the patient’s health, stage, and location of the tumor. The efficiency of these treatments can be decreased due to the development of diverse resistance mechanisms. New therapeutic targets have emerged from studies of the genetic profile of melanocytes and from the identification of molecular factors involved in the pathogenesis of the malignant transformation. In this review, we aim to survey therapies approved and under evaluation for melanoma treatment and relevant research on the molecular mechanisms underlying melanomagenesis.
Collapse
Affiliation(s)
- Beatriz Domingues
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Faculty of Sciences, University of Porto, Porto, Portugal
| | - José Manuel Lopes
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Hospital S João, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Paula Soares
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Helena Pópulo
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
42
|
Cassimeris L, Leung JC, Odde DJ. Monte Carlo simulations of microtubule arrays: The critical roles of rescue transitions, the cell boundary, and tubulin concentration in shaping microtubule distributions. PLoS One 2018; 13:e0197538. [PMID: 29782540 PMCID: PMC5962052 DOI: 10.1371/journal.pone.0197538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/03/2018] [Indexed: 01/12/2023] Open
Abstract
Microtubules are dynamic polymers required for a number of processes, including chromosome movement in mitosis. While regulators of microtubule dynamics have been well characterized, we lack a convenient way to predict how the measured dynamic parameters shape the entire microtubule system within a cell, or how the system responds when specific parameters change in response to internal or external signals. Here we describe a Monte Carlo model to simulate an array of dynamic microtubules from parameters including the cell radius, total tubulin concentration, microtubule nucleation rate from the centrosome, and plus end dynamic instability. The algorithm also allows dynamic instability or position of the cell edge to vary during the simulation. Outputs from simulations include free tubulin concentration, average microtubule lengths, length distributions, and individual length changes over time. Using this platform and reported parameters measured in interphase LLCPK1 epithelial cells, we predict that sequestering ~ 15-20% of total tubulin results in fewer microtubules, but promotes dynamic instability of those remaining. Simulations also predict that lowering nucleation rate will increase the stability and average length of the remaining microtubules. Allowing the position of the cell's edge to vary over time changed the average length but not the number of microtubules and generated length distributions consistent with experimental measurements. Simulating the switch from interphase to prophase demonstrated that decreased rescue frequency at prophase is the critical factor needed to rapidly clear the cell of interphase microtubules prior to mitotic spindle assembly. Finally, consistent with several previous simulations, our results demonstrate that microtubule nucleation and dynamic instability in a confined space determines the partitioning of tubulin between monomer and polymer pools. The model and simulations will be useful for predicting changes to the entire microtubule array after modification to one or more parameters, including predicting the effects of tubulin-targeted chemotherapies.
Collapse
Affiliation(s)
- Lynne Cassimeris
- Dept. of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Jessica C Leung
- Dept. of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - David J Odde
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
43
|
Hemmat M, Castle BT, Odde DJ. Microtubule dynamics: moving toward a multi-scale approach. Curr Opin Cell Biol 2018; 50:8-13. [PMID: 29351860 PMCID: PMC5911414 DOI: 10.1016/j.ceb.2017.12.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/12/2017] [Accepted: 12/23/2017] [Indexed: 12/17/2022]
Abstract
Microtubule self-assembly dynamics serve to facilitate many vital cellular functions, such as chromosome segregation during mitosis and synaptic plasticity. However, the detailed atomistic basis of assembly dynamics has remained an unresolved puzzle. A key challenge is connecting together the vast range of relevant length-time scales, events happening at time scales ranging from nanoseconds, such as tubulin molecular interactions (Å-nm), to minutes-hours, such as the cellular response to microtubule dynamics during mitotic progression (μm). At the same time, microtubule interactions with associated proteins and binding agents, such as anti-cancer drugs, can strongly affect this dynamic process through atomic-level mechanisms that remain to be elucidated. New high-resolution technologies for investigating these interactions, including cryo-electron microscopy (EM) techniques and total internal reflection fluorescence (TIRF) microscopy, are yielding important new insights. Here, we focus on recent studies of microtubule dynamics, both theoretical and experimental, and how these findings shed new light on this complex phenomenon across length-time scales, from Å to μm and from nanoseconds to minutes.
Collapse
Affiliation(s)
- Mahya Hemmat
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Brian T Castle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
44
|
Majellaro M, Stefanachi A, Tardia P, Vicenti C, Boccarelli A, Pannunzio A, Campanella F, Coluccia M, Denora N, Leonetti F, de Candia M, Altomare CD, Cellamare S. Investigating Structural Requirements for the Antiproliferative Activity of Biphenyl Nicotinamides. ChemMedChem 2017; 12:1380-1389. [PMID: 28665505 DOI: 10.1002/cmdc.201700365] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Indexed: 11/09/2022]
Abstract
A number of trimethoxybenzoic acid anilides, previously studied as permeability glycoprotein (P-gp) modulators, were screened with the aim of identifying new anticancer agents. One of these compounds, which showed antiproliferative activity against resistant MCF-7 cell line, was selected as the hit structure. Replacement of the trimethoxybenzoyl moiety with a nicotinoyl group, in order to overcome solubility issues, led to a new series of N-biphenyl nicotinoyl anilides, among which a nitro derivative, N-(3',5'-difluoro-3-nitro-[1,1'-biphenyl]-4-yl)nicotinamide (3), displayed antiproliferative activity against MCF-7 and MDA-MB-231 cells in the nanomolar range. The search for a bioisostere of the nitro group led to nitrile analogue N-(3-cyano-4'-fluoro-[1,1'-biphenyl]-4-yl)nicotinamide (36), which shows a strong increase in activity against MCF-7 and MDA-MB-231 cells. Compound 36 induced a dose-dependent accumulation of G2 - and M-phase MCF-7 cell populations, and a decrease in S-phase cells. Relative to vinblastine, a well-known potent antimitotic agent, compound 36 also induced G1 -phase arrest at low doses (20-40 nm), but did not inhibit in vitro tubulin polymerization.
Collapse
Affiliation(s)
- Maria Majellaro
- Department of Pharmacy-Drug Sciences, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Angela Stefanachi
- Department of Pharmacy-Drug Sciences, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Piero Tardia
- D3-Drug Discovery and Development Department, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genova, Italy
| | - Chiara Vicenti
- Department of Emergency and Organ Transplantation, Section of Pathological Anatomy, University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Angelina Boccarelli
- Department of Biomedical Sciences and Human Oncology, University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Alessandra Pannunzio
- Department of Pharmacy-Drug Sciences, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Federica Campanella
- Department of Pharmacy-Drug Sciences, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Mauro Coluccia
- Department of Pharmacy-Drug Sciences, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Nunzio Denora
- Department of Pharmacy-Drug Sciences, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Francesco Leonetti
- Department of Pharmacy-Drug Sciences, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Modesto de Candia
- Department of Pharmacy-Drug Sciences, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | | | - Saverio Cellamare
- Department of Pharmacy-Drug Sciences, University of Bari, Via Orabona 4, 70125, Bari, Italy
| |
Collapse
|