Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.639
Filter
1.
World J Gastroenterol ; 30(26): 3257-3260, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39086641

ABSTRACT

Over the years immunotherapy has demonstrably improved the field of cancer treatment. However, achieving long-term survival for colorectal cancer (CRC) patients remains a significant unmet need. Combination immunotherapies incorporating targeted drugs like MEK or multi-kinase inhibitors have offered some palliative benefit. Nevertheless, substantial gaps remain in the current therapeutic armamentarium for CRC. In recent years, there has been a surge of interest in exploring novel treatment strategies, including the application of light-activated drugs in conjunction with optical devices. This approach holds promise for achieving localized and targeted delivery of cytotoxic agents, such as microtubule-targeting drugs, directly to cancerous cells within the colon.


Subject(s)
Colorectal Neoplasms , Microtubules , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Microtubules/drug effects , Microtubules/metabolism , Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy/methods , Drug Delivery Systems/methods , Photosensitizing Agents/therapeutic use , Immunotherapy/methods , Photochemotherapy/methods , Tubulin Modulators/therapeutic use , Tubulin Modulators/pharmacology
2.
J Med Chem ; 67(14): 12118-12142, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38996194

ABSTRACT

Disrupting microtubule dynamics has emerged as a promising strategy for cancer treatment. However, drug resistance remains a challenge hindering the development of microtubule-targeting agents. In this work, a novel class of diaryl substituted fused heterocycles were designed, synthesized, and evaluated, which were demonstrated as effective dual katanin and tubulin regulators with antitumor activity. Following three rounds of stepwise optimization, compound 21b, featuring a 3H-imidazo[4,5-b]pyridine core, displayed excellent targeting capabilities on katanin and tubulin, along with notable antiproliferative and antimetastatic effects. Mechanistic studies revealed that 21b disrupts the microtubule network in tumor cells, leading to G2/M cell cycle arrest and apoptosis induction. Importantly, 21b exhibited significant inhibition of tumor growth in MDA-MB-231 and A549/T xenograft tumor models without evident toxicity and side effects. In conclusion, compound 21b presents a novel mechanism for disrupting microtubule dynamics, warranting further investigation as a dual-targeted antitumor agent with potential antimultidrug resistance properties.


Subject(s)
Antineoplastic Agents , Drug Resistance, Neoplasm , Heterocyclic Compounds , Katanin , Tubulin Modulators , Tubulin , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Tubulin/metabolism , Drug Resistance, Neoplasm/drug effects , Animals , Katanin/metabolism , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Cell Line, Tumor , Tubulin Modulators/pharmacology , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Mice , Apoptosis/drug effects , Cell Proliferation/drug effects , Structure-Activity Relationship , Mice, Nude , Drug Discovery , Microtubules/drug effects , Microtubules/metabolism , Xenograft Model Antitumor Assays , Drug Screening Assays, Antitumor , Mice, Inbred BALB C , Female
3.
Int J Mol Sci ; 25(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891892

ABSTRACT

Recently, a compound derived from recent scientific advances named 34 has emerged as the focus of this research, the aim of which is to explore its potential impact on solid tumor cell lines. Using a combination of bioinformatics and biological assays, this study conducted an in-depth investigation of the effects of 34. The results of this study have substantial implications for cancer research and treatment. 34 has shown remarkable efficacy in inhibiting the growth of several cancer cell lines, including those representing prostate carcinoma (PC3) and cervical carcinoma (HeLa). The high sensitivity of these cells, indicated by low IC50 values, underscores its potential as a promising chemotherapeutic agent. In addition, 34 has revealed the ability to induce cell cycle arrest, particularly in the G2/M phase, a phenomenon with critical implications for tumor initiation and growth. By interfering with DNA replication in cancer cells, 34 has shown the capacity to trigger cell death, offering a new avenue for cancer treatment. In addition, computational analyses have identified key genes affected by 34 treatment, suggesting potential therapeutic targets. These genes are involved in critical biological processes, including cell cycle regulation, DNA replication and microtubule dynamics, all of which are central to cancer development and progression. In conclusion, this study highlights the different mechanisms of 34 that inhibit cancer cell growth and alter the cell cycle. These promising results suggest the potential for more effective and less toxic anticancer therapies. Further in vivo validation and exploration of combination therapies are critical to improve cancer treatment outcomes.


Subject(s)
Acrylonitrile , Antineoplastic Agents , Microtubules , Humans , Microtubules/drug effects , Microtubules/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Acrylonitrile/analogs & derivatives , Acrylonitrile/pharmacology , Acrylonitrile/therapeutic use , Cell Proliferation/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , HeLa Cells , Apoptosis/drug effects , Triazoles/pharmacology , Cell Cycle Checkpoints/drug effects , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , PC-3 Cells
4.
J Integr Neurosci ; 23(6): 118, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38940085

ABSTRACT

BACKGROUND: Alcohol abuse, a prevalent global health issue, is associated with the onset of cognitive impairment and neurodegeneration. Actin filaments (F-actin) and microtubules (MTs) polymerized from monomeric globular actin (G-actin) and tubulin form the structural basis of the neuronal cytoskeleton. Precise regulation of the assembly and disassembly of these cytoskeletal proteins, and their dynamic balance, play a pivotal role in regulating neuronal morphology and function. Nevertheless, the effect of prolonged alcohol exposure on cytoskeleton dynamics is not fully understood. This study investigates the chronic effects of alcohol on cognitive ability, neuronal morphology and cytoskeleton dynamics in the mouse hippocampus. METHODS: Mice were provided ad libitum access to 5% (v/v) alcohol in drinking water and were intragastrically administered 30% (v/v, 6.0 g/kg/day) alcohol for six weeks during adulthood. Cognitive functions were then evaluated using the Y maze, novel object recognition and Morris water maze tests. Hippocampal histomorphology was assessed through hematoxylin-eosin (HE) and Nissl staining. The polymerized and depolymerized states of actin cytoskeleton and microtubules were separated using two commercial assay kits and quantified by Western blot analysis. RESULTS: Mice chronically exposed to alcohol exhibited significant deficits in spatial and recognition memory as evidenced by behavioral tests. Histological analysis revealed notable hippocampal damage and neuronal loss. Decreased ratios of F-actin/G-actin and MT/tubulin, along with reduced levels of polymerized F-actin and MTs, were found in the hippocampus of alcohol-treated mice. CONCLUSIONS: Our findings suggest that chronic alcohol consumption disrupted the assembly of the actin cytoskeleton and MTs in the hippocampus, potentially contributing to the cognitive deficits and pathological injury induced by chronic alcohol intoxication.


Subject(s)
Actin Cytoskeleton , Ethanol , Hippocampus , Microtubules , Animals , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Microtubules/drug effects , Microtubules/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Male , Ethanol/pharmacology , Ethanol/administration & dosage , Mice , Mice, Inbred C57BL , Central Nervous System Depressants/pharmacology , Central Nervous System Depressants/administration & dosage , Disease Models, Animal , Behavior, Animal/drug effects
5.
Alzheimers Dement ; 20(7): 4540-4558, 2024 07.
Article in English | MEDLINE | ID: mdl-38884283

ABSTRACT

INTRODUCTION: Intraneuronal inclusions composed of tau protein are found in Alzheimer's disease (AD) and other tauopathies. Tau normally binds microtubules (MTs), and its disengagement from MTs and misfolding in AD is thought to result in MT abnormalities. We previously identified triazolopyrimidine-containing MT-stabilizing compounds that provided benefit in AD mouse models and herein describe the characterization and efficacy testing of an optimized candidate, CNDR-51997. METHODS: CNDR-51997 underwent pharmacokinetic, pharmacodynamic, safety pharmacology, and mouse tolerability testing. In addition, the compound was examined for efficacy in 5XFAD amyloid beta (Aß) plaque mice and PS19 tauopathy mice. RESULTS: CNDR-51997 significantly reduced Aß plaques in 5XFAD mice and tau pathology in PS19 mice, with the latter also showing attenuated axonal dystrophy and gliosis. CNDR-51997 was well tolerated at doses that exceeded efficacy doses, with a good safety pharmacology profile. DISCUSSION: CNDR-51997 may be a candidate for advancement as a potential therapeutic agent for AD and/or other tauopathies. Highlights There is evidence of microtubule alterations (MT) in Alzheimer's disease (AD) brain and in mouse models of AD pathology. Intermittent dosing with an optimized, brain-penetrant MT-stabilizing small-molecule, CNDR-51997, reduced both Aß plaque and tau inclusion pathology in established mouse models of AD. CNDR-51997 attenuated axonal dystrophy and gliosis in a tauopathy mouse model, with a strong trend toward reduced hippocampal neuron loss. CNDR-51997 is well tolerated in mice at doses that are meaningfully greater than required for efficacy in AD mouse models, and the compound has a good safety pharmacology profile.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Mice, Transgenic , Microtubules , Plaque, Amyloid , tau Proteins , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Mice , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , tau Proteins/metabolism , Microtubules/drug effects , Microtubules/metabolism , Brain/drug effects , Brain/pathology , Brain/metabolism , Tauopathies/drug therapy , Tauopathies/pathology , Humans , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , Amyloid beta-Peptides/metabolism
6.
Life Sci ; 351: 122836, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38879159

ABSTRACT

AIM: Exploring the efficacy of ß-carboline-based molecular inhibitors in targeting microtubules for the development of novel anticancer therapeutics. MATERIALS AND METHODS: We synthesized a series of 1-Aryl-N-substituted-ß-carboline-3-carboxamide compounds and evaluated their cytotoxicity against human lung carcinoma (A549) cells using the MTT assay. Normal lung fibroblast cells (WI-38) were used to assess compound selectivity. The mechanism of action of MJ-211 was elucidated through Western blot analysis of key pro-apoptotic and cell cycle regulatory proteins. Additionally, the inhibitory effect of MJ-211 on multicellular 3D spheroid growth of A549 cells was evaluated. KEY FINDINGS: Lead compound MJ-211 exhibited remarkable cytotoxicity against A549 cells with an IC50 of 4.075 µM at 24 h treatment and IC50 of 1.7 nM after 72 h of treatment, while demonstrating selectivity towards normal WI-38 cells. MJ-211 activated pro-apoptotic factors Bim and p53, and suppressed Cyclin B1, Phospho HSP 27, BubR1, Mad 2, ERK1/2, and NF-κB, indicating its potent antimitotic and pro-apoptotic effects. MJ-211 significantly suppressed the migration of cells and inhibited the growth of A549 cell-derived multicellular 3D spheroids, highlighting its efficacy in a more physiologically relevant model. SIGNIFICANCE: Cytotoxic effect of MJ-211 against cancer cells, selectivity towards normal cells, and ability to modulate key regulatory proteins involved in apoptosis and cell cycle progression underscore its potential as a promising template for further anticancer lead optimization. Moreover, the inhibitory effect of MJ-211 on multicellular spheroid growth suggests its efficacy in combating tumor heterogeneity and resistance mechanisms, thereby offering a promising avenue for future anticancer drug development.


Subject(s)
Carbolines , Microtubules , NF-kappa B , Humans , Carbolines/pharmacology , NF-kappa B/metabolism , Microtubules/drug effects , Microtubules/metabolism , A549 Cells , Antimitotic Agents/pharmacology , Down-Regulation/drug effects , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects
7.
J Med Chem ; 67(11): 9227-9259, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38833507

ABSTRACT

The marine metabolite diazonamide A exerts low nanomolar cytotoxicity against a range of tumor cell lines; however, its highly complex molecular architecture undermines the therapeutic potential of the natural product. We demonstrate that truncation of heteroaromatic macrocycle in natural diazonamide A, combined with the replacement of the challenging-to-synthesize tetracyclic hemiaminal subunit by oxindole moiety leads to considerably less complex analogues with improved drug-like properties and nanomolar antiproliferative potency. The structurally simplified macrocycles are accessible in 12 steps from readily available indolin-2-one and tert-leucine with excellent diastereoselectivity (99:1 dr) in the key macrocyclization step. The most potent macrocycle acts as a tubulin assembly inhibitor and exerts similar effects on A2058 cell cycle progression and induction of apoptosis as does marketed microtubule-targeting agent vinorelbine.


Subject(s)
Antineoplastic Agents , Apoptosis , Microtubules , Tubulin Modulators , Humans , Tubulin Modulators/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/chemical synthesis , Cell Line, Tumor , Microtubules/drug effects , Microtubules/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Structure-Activity Relationship , Cell Proliferation/drug effects , Cell Cycle/drug effects , Biological Products/pharmacology , Biological Products/chemistry , Biological Products/chemical synthesis , Drug Screening Assays, Antitumor , Stereoisomerism , Tubulin/metabolism , Tubulin/chemistry , Indoles/chemistry , Indoles/pharmacology , Indoles/chemical synthesis , Heterocyclic Compounds, 4 or More Rings , Oxazoles
8.
Exp Cell Res ; 440(1): 114126, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38857838

ABSTRACT

Microtubules are components of the cytoskeleton that perform essential functions in eukaryotes, such as those related to shape change, motility and cell division. In this context some characteristics of these filaments are essential, such as polarity and dynamic instability. In trypanosomatids, microtubules are integral to ultrastructure organization, intracellular transport and mitotic processes. Some species of trypanosomatids co-evolve with a symbiotic bacterium in a mutualistic association that is marked by extensive metabolic exchanges and a coordinated division of the symbiont with other cellular structures, such as the nucleus and the kinetoplast. It is already established that the bacterium division is microtubule-dependent, so in this work, it was investigated whether the dynamism and remodeling of these filaments is capable of affecting the prokaryote division. To this purpose, Angomonas deanei was treated with Trichostatin A (TSA), a deacetylase inhibitor, and mutant cells for histone deacetylase 6 (HDAC6) were obtained by CRISPR-Cas9. A decrease in proliferation, an enhancement in tubulin acetylation, as well as morphological and ultrastructural changes, were observed in TSA-treated protozoa and mutant cells. In both cases, symbiont filamentation occurred, indicating that prokaryote cell division is dependent on microtubule dynamism.


Subject(s)
Cell Division , Microtubules , Symbiosis , Microtubules/metabolism , Microtubules/ultrastructure , Microtubules/drug effects , Trypanosomatina/genetics , Trypanosomatina/metabolism , Trypanosomatina/ultrastructure , Trypanosomatina/physiology , Hydroxamic Acids/pharmacology , Tubulin/metabolism , Tubulin/genetics , Bacteria/metabolism , Bacteria/genetics , Acetylation , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase 6/metabolism , Histone Deacetylase 6/genetics , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure
9.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38892342

ABSTRACT

The synthesis of biphenylmethoxydibenzo[b,f]oxepine or photoswitchable fluorinated dibenzo[b,f]oxepine derivatives with one or three azo bonds, potential microtubule inhibitors, is described. Our studies provide a concise method for constructing derivatives containing the dibenzo[b,f]oxepine skeleton. An analysis of products was run using experimental and theoretical methods. Next, we evaluated the E/Z isomerization of azo-dibenzo[b,f]oxepine derivatives, which could be photochemically controlled using visible-wavelength light.


Subject(s)
Tubulin Modulators , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology , Tubulin Modulators/chemistry , Microtubules/drug effects , Microtubules/metabolism , Molecular Structure
10.
Adv Sci (Weinh) ; 11(29): e2305593, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38873820

ABSTRACT

Centromere protein A (CENP-A), a centromere-specific histone H3 variant, is crucial for kinetochore positioning and chromosome segregation. However, its regulatory mechanism in human cells remains incompletely understood. A structure-activity relationship (SAR) study of the cell-cycle-arresting indole terpenoid mimic JP18 leads to the discovery of two more potent analogs, (+)-6-Br-JP18 and (+)-6-Cl-JP18. Tubulin is identified as a potential cellular target of these halogenated analogs by using the drug affinity responsive target stability (DARTS) based method. X-ray crystallography analysis reveals that both molecules bind to the colchicine-binding site of ß-tubulin. Treatment of human cells with microtubule-targeting agents (MTAs), including these two compounds, results in CENP-A accumulation by destabilizing Cdh1, a co-activator of the anaphase-promoting complex/cyclosome (APC/C) E3 ubiquitin ligase. This study establishes a link between microtubule dynamics and CENP-A accumulation using small-molecule tools and highlights the role of Cdh1 in CENP-A proteolysis.


Subject(s)
Centromere Protein A , Microtubules , Proteolysis , Humans , Microtubules/metabolism , Microtubules/drug effects , Centromere Protein A/metabolism , Centromere Protein A/genetics , Proteolysis/drug effects , Structure-Activity Relationship , Indoles/metabolism , Indoles/pharmacology , Indoles/chemistry , Crystallography, X-Ray/methods , Spindle Apparatus/metabolism , Spindle Apparatus/drug effects , Cdh1 Proteins/metabolism , Cdh1 Proteins/genetics , Antigens, CD , Cadherins
11.
Behav Brain Res ; 471: 115068, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38830386

ABSTRACT

Alcohol-Related Brain Damage (ARBD) manifests predominantly as cognitive impairment and brain atrophy with the hippocampus showing particular vulnerability. Fasudil, a Rho kinase (ROCK) inhibitor, has established neuroprotective properties; however, its impact on alcohol-induced cognitive dysfunction and hippocampal structural damage remains unelucidated. This study probes Fasudil's neuroprotective potential and identifies its mechanism of action in an in vivo context. Male C57BL/6 J mice were exposed to 30% (v/v, 6.0 g/kg) ethanol by intragastric administration for four weeks. Concurrently, these mice received a co-treatment with Fasudil through intraperitoneal injections at a dosage of 10 mg/kg/day. Fasudil was found to mitigate alcohol-induced spatial and recognition memory deficits, which were quantified using Y maze, Morris water maze, and novel object recognition tests. Concurrently, Fasudil attenuated hippocampal structural damage prompted by chronic alcohol exposure. Notably, Fasudil moderated alcohol-induced disassembly of the actin cytoskeleton and microtubules-mechanisms central to the maintenance of hippocampal synaptic integrity. Collectively, our findings indicate that Fasudil partially reverses alcohol-induced cognitive and morphological detriments by modulating cytoskeletal dynamics, offering insights into potential therapeutic strategies for ARBD.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Cognitive Dysfunction , Ethanol , Hippocampus , Mice, Inbred C57BL , Microtubules , Neuroprotective Agents , Animals , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/metabolism , Male , Mice , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/chemically induced , Ethanol/pharmacology , Neuroprotective Agents/pharmacology , Microtubules/drug effects , Microtubules/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Disease Models, Animal , Recognition, Psychology/drug effects
12.
Adv Exp Med Biol ; 1452: 1-19, 2024.
Article in English | MEDLINE | ID: mdl-38805122

ABSTRACT

Microtubules are dynamic polymers composed of α- and ß-tubulin heterodimers. Microtubules are universally conserved among eukaryotes and participate in nearly every cellular process, including intracellular trafficking, replication, polarity, cytoskeletal shape, and motility. Due to their fundamental role in mitosis, they represent a classic target of anti-cancer therapy. Microtubule-stabilizing agents currently constitute a component of the most effective regimens for ovarian cancer therapy in both primary and recurrent settings. Unfortunately, the development of resistance continues to present a therapeutic challenge. An understanding of the underlying mechanisms of resistance to microtubule-active agents may facilitate the development of novel and improved approaches to this disease.


Subject(s)
Cytoskeleton , Microtubules , Ovarian Neoplasms , Tubulin Modulators , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Female , Microtubules/drug effects , Microtubules/metabolism , Tubulin Modulators/therapeutic use , Tubulin Modulators/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Drug Resistance, Neoplasm/drug effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Animals
13.
J Transl Med ; 22(1): 441, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730481

ABSTRACT

Microtubule targeting agents (MTAs) are commonly prescribed to treat cancers and predominantly kill cancer cells in mitosis. Significantly, some MTA-treated cancer cells escape death in mitosis, exit mitosis and become malignant polyploid giant cancer cells (PGCC). Considering the low number of cancer cells undergoing mitosis in tumor tissues, killing them in interphase may represent a favored antitumor approach. We discovered that ST-401, a mild inhibitor of microtubule (MT) assembly, preferentially kills cancer cells in interphase as opposed to mitosis, a cell death mechanism that avoids the development of PGCC. Single cell RNA sequencing identified mRNA transcripts regulated by ST-401, including mRNAs involved in ribosome and mitochondrial functions. Accordingly, ST-401 induces a transient integrated stress response, reduces energy metabolism, and promotes mitochondria fission. This cell response may underly death in interphase and avoid the development of PGCC. Considering that ST-401 is a brain-penetrant MTA, we validated these results in glioblastoma cell lines and found that ST-401 also reduces energy metabolism and promotes mitochondria fission in GBM sensitive lines. Thus, brain-penetrant mild inhibitors of MT assembly, such as ST-401, that induce death in interphase through a previously unanticipated antitumor mechanism represent a potentially transformative new class of therapeutics for the treatment of GBM.


Subject(s)
Cell Death , Giant Cells , Interphase , Microtubules , Polyploidy , Humans , Interphase/drug effects , Microtubules/metabolism , Microtubules/drug effects , Cell Line, Tumor , Cell Death/drug effects , Giant Cells/drug effects , Giant Cells/metabolism , Giant Cells/pathology , Mitochondrial Dynamics/drug effects , Energy Metabolism/drug effects , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/genetics , Neoplasms/pathology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Gene Expression Regulation, Neoplastic/drug effects
14.
Curr Opin Struct Biol ; 87: 102845, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38805950

ABSTRACT

Microtubule-targeting agents (MTAs) have demonstrated remarkable efficacy as antitumor, antifungal, antiparasitic, and herbicidal agents, finding applications in the clinical, veterinary, and agrochemical industry. Recent advances in tubulin and microtubule structural biology have provided powerful tools that pave the way for the rational design of innovative small-molecule MTAs for future basic and applied life science applications. In this mini-review, we present the current status of the tubulin and microtubule structural biology field, the recent impact it had on the discovery and rational design of MTAs, and exciting avenues for future MTA research.


Subject(s)
Microtubules , Tubulin Modulators , Tubulin , Microtubules/metabolism , Microtubules/chemistry , Microtubules/drug effects , Humans , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Drug Discovery/methods , Drug Design , Animals , Structure-Activity Relationship , Models, Molecular
15.
J Mech Behav Biomed Mater ; 156: 106586, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38805872

ABSTRACT

Both mechanical and adhesion properties of cancer cells are complex and reciprocally related to migration, invasion, and metastasis with large cell deformation. Therefore, we evaluated these properties for human cervical cancer cells (HeLa) simultaneously using our previously developed micro tensile tester system. For efficient evaluation, we developed image analysis software to modify the system. The software can analyze the tensile force in real time. The modified system can evaluate the tensile stiffness of cells to which a large deformation is applied, also evaluate the adhesion strength of cancer cells that adhered to a culture substrate and were cultured for several days with their adhesion maturation. We used the modified system to simultaneously evaluate the stiffness of the cancer cells to which a large deformation was applied and their adhesion strength. The obtained results revealed that the middle phase of tensile stiffness and adhesion force of the microtubule-depolymerized group treated with colchicine (an anti-cancer drug) (stiffness, 13.4 ± 7.5 nN/%; adhesion force, 460.6 ± 258.2 nN) were over two times larger than those of the control group (stiffness, 5.0 ± 3.5 nN/%; adhesion force, 168.2 ± 98.0 nN). Additionally, the same trend was confirmed with the detailed evaluation of cell surface stiffness using an atomic force microscope. Confocal fluorescence microscope observations showed that the stress fibers (SFs) of colchicine-treated cells were aligned in the same direction, and focal adhesions (FAs) of the cells developed around both ends of the SFs and aligned parallel to the developed direction of the SFs. There was a possibility that the microtubule depolymerization by the colchicine treatment induced the development of SFs and FAs and subsequently caused an increment of cell stiffness and adhesion force. From the above results, we concluded the modified system would be applicable to cancer detection and anti-cancer drug efficacy tests.


Subject(s)
Cell Adhesion , Microtubules , Tensile Strength , Humans , Microtubules/drug effects , Cell Adhesion/drug effects , Biomechanical Phenomena/drug effects , HeLa Cells , Polymerization/drug effects , Materials Testing , Mechanical Phenomena , Colchicine/pharmacology
16.
Toxicol In Vitro ; 99: 105856, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38821378

ABSTRACT

Acute leukemias present therapeutic challenges despite advances in treatments. Microtubule inhibitors have played a pivotal role in cancer therapy, inspiring exploration into novel compounds like C2E1 from the cyclopenta[b]indole class. In the present study, we investigated C2E1's potential as a therapeutic agent for acute leukemia at molecular, cellular, and genetic levels. C2E1 demonstrated tubulin depolarization activity, significantly reducing leukemia cell viability. Its impact involved multifaceted mechanisms: inducing apoptosis, arrest of cell cycle progression, and inhibition of clonogenicity and migration in leukemia cells. At a molecular level, C2E1 triggered DNA damage, antiproliferative, and apoptosis markers and altered gene expression related to cytoskeletal regulation, disrupting essential cellular processes crucial for leukemia cell survival and proliferation. These findings highlight C2E1's promise as a potential candidate for novel anti-cancer therapies. Notably, its distinct mode of action from conventional microtubule-targeting drugs suggests the potential to bypass common resistance mechanisms encountered with existing treatments. In summary, C2E1 emerges as a compelling compound with diverse effects on leukemia cells, showcasing promising antineoplastic properties. Its ability to disrupt critical cellular functions selective to leukemia cells positions it as a candidate for future therapeutic development.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Survival , Indoles , Leukemia , Tubulin Modulators , Humans , Leukemia/drug therapy , Tubulin Modulators/pharmacology , Indoles/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Proliferation/drug effects , Tubulin/metabolism , DNA Damage/drug effects , Cell Movement/drug effects , Microtubules/drug effects
17.
Brain Res ; 1838: 148987, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38718851

ABSTRACT

Dynamin is a microtubule (MT) binding protein playing a key role in vesicle endocytosis. In a brain slice model, tau loaded in presynaptic terminals assembles MTs, thereby impairing vesicle endocytosis via depletion of cytosolic dynamin. The peptide PHDP5, derived from the pleckstrin homology domain of dynamin 1, inhibits dynamin-MT interaction and rescues endocytosis and synaptic transmission impaired by tau when co-loaded in presynaptic terminals. We tested whether in vivo administration of PHDP5 could rescue the learning/memory deficits observed in Alzheimer's disease (AD) model mice. A modified PHDP5 incorporating a cell-penetrating peptide (CPP) and a FITC fluorescent marker was delivered intranasally to Tau609 transgenic (Tg) and 3xTg-AD mice. FITC-positive puncta were observed in the hippocampus of mice infused with PHDP5 or scrambled (SPHDP5) peptide, but not in saline-infused controls. In the Morris water maze (MWM) test for spatial learning/memory, AD model mice treated with FITC-PHDP5-CPP showed prominent improvements in learning and memory, performing close to the level of saline-infused WT mice control. In contrast, mice treated with a scrambled construct (FITC-SPHDP5-CPP) showed no significant improvement. We conclude that PHDP5 can be a candidate for human AD therapy.


Subject(s)
Alzheimer Disease , Memory Disorders , Spatial Learning , Animals , Male , Mice , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/drug effects , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/drug therapy , Memory Disorders/metabolism , Mice, Transgenic , Microtubules/metabolism , Microtubules/drug effects , Spatial Learning/drug effects , tau Proteins/metabolism , Dynamin I/metabolism
18.
Biochem Pharmacol ; 225: 116329, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38821375

ABSTRACT

Calcium signaling abnormality in cardiomyocytes, as a key mechanism, is closely associated with developing heart failure. Fibroblast growth factor 13 (FGF13) demonstrates important regulatory roles in the heart, but its association with cardiac calcium signaling in heart failure remains unknown. This study aimed to investigate the role and mechanism of FGF13 on calcium mishandling in heart failure. Mice underwent transaortic constriction to establish a heart failure model, which showed decreased ejection fraction, fractional shortening, and contractility. FGF13 deficiency alleviated cardiac dysfunction. Heart failure reduces calcium transients in cardiomyocytes, which were alleviated by FGF13 deficiency. Meanwhile, FGF13 deficiency restored decreased Cav1.2 and Serca2α expression and activity in heart failure. Furthermore, FGF13 interacted with microtubules in the heart, and FGF13 deficiency inhibited the increase of microtubule stability during heart failure. Finally, in isoproterenol-stimulated FGF13 knockdown neonatal rat ventricular myocytes (NRVMs), wildtype FGF13 overexpression, but not FGF13 mutant, which lost the binding site of microtubules, promoted calcium transient abnormality aggravation and Cav1.2 downregulation compared with FGF13 knockdown group. Generally, FGF13 deficiency improves abnormal calcium signaling by inhibiting the increased microtubule stability in heart failure, indicating the important role of FGF13 in cardiac calcium homeostasis and providing new avenues for heart failure prevention and treatment.


Subject(s)
Calcium Signaling , Fibroblast Growth Factors , Heart Failure , Microtubules , Myocytes, Cardiac , Animals , Male , Mice , Rats , Cells, Cultured , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Heart Failure/metabolism , Heart Failure/genetics , Mice, Inbred C57BL , Mice, Knockout , Microtubules/metabolism , Microtubules/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Rats, Sprague-Dawley
19.
Nanoscale ; 16(16): 7976-7987, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38567463

ABSTRACT

Collective functionalization of the phytochemicals of medicinal herbs on nanoparticles is emerging as a potential cancer therapeutic strategy. This study presents the facile synthesis of surface-functionalized gold nanoparticles using Bacopa monnieri (Brahmi; Bm) phytochemicals and their therapeutically relevant mechanism of action in the colorectal cancer cell line, HT29. The nanoparticles were characterized using UV-visible spectroscopy, TEM-EDAX, zeta potential analysis, TGA, FTIR and 1H NMR spectroscopy, and HR-LC-MS. The particles (Bm-GNPs) were of polygonal shape and were stable against aggregation. They entered the target cells and inhibited the viability and clonogenicity of the cells with eight times more antiproliferative efficacy (25 ± 1.5 µg mL-1) than Bm extract (Bm-EX). In vitro studies revealed that Bm-GNPs bind tubulin (a protein crucial in cell division and a target of anticancer drugs) and disrupt its helical structure without grossly altering its tertiary conformation. Like other antitubulin agents, Bm-GNPs induced G2/M arrest and ultimately killed the cells, as confirmed using flow cytometry analyses. ZVAD-FMK-mediated global pan-caspase inhibition and the apparent absence of cleaved caspase-3 in treated cells indicated that the death did not involve the classic apoptosis pathway. Cellular ultrastructure analyses, western immunoblots, and in situ immunofluorescence visualization of cellular microtubules revealed microtubule-acetylation-independent induction of autophagy as the facilitator of cell death. Together, the data indicate strong antiproliferative efficacy and a possible mechanism of action for these designer nanoparticles. Bm-GNPs, therefore, merit further investigations, including preclinical evaluations, for their therapeutic potential as inducers of non-apoptotic cell death.


Subject(s)
Autophagy , Colorectal Neoplasms , Gold , Metal Nanoparticles , Humans , Gold/chemistry , Gold/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Metal Nanoparticles/chemistry , Autophagy/drug effects , Acetylation , Microtubules/metabolism , Microtubules/drug effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/drug therapy , HT29 Cells , Caspases/metabolism , Phytochemicals/pharmacology , Phytochemicals/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Tubulin/metabolism , Tubulin/chemistry
20.
World J Gastroenterol ; 30(13): 1780-1790, 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38659489

ABSTRACT

Colorectal cancer (CRC) has remained the second and the third leading cause of cancer-related death worldwide and in the United States, respectively. Although significant improvement in overall survival has been achieved, death in adult populations under the age of 55 appears to have increased in the past decades. Although new classes of therapeutic strategies such as immunotherapy have emerged, their application is very limited in CRC so far. Microtubule (MT) inhibitors such as taxanes, are not generally successful in CRC. There may be some way to make MT inhibitors work effectively in CRC. One potential advantage that we can take to treat CRC may be the combination of optical techniques coupled to an endoscope or other fiber optics-based devices. A combination of optical devices and photo-activatable drugs may allow us to locally target advanced CRC cells with highly potent MT-targeting drugs. In this Editorial review, we would like to discuss the potential of optogenetic approaches in CRC management.


Subject(s)
Colorectal Neoplasms , Microtubules , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/therapy , Humans , Microtubules/drug effects , Microtubules/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Optogenetics/methods , Tubulin Modulators/therapeutic use , Tubulin Modulators/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL