Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Neurobiol Dis ; 198: 106554, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38844243

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder that severely affects the basal ganglia and regions of the cerebral cortex. While astrocytosis and microgliosis both contribute to basal ganglia pathology, the contribution of gliosis and potential factors driving glial activity in the human HD cerebral cortex is less understood. Our study aims to identify nuanced indicators of gliosis in HD which is challenging to identify in the severely degenerated basal ganglia, by investigating the middle temporal gyrus (MTG), a cortical region previously documented to demonstrate milder neuronal loss. Immunohistochemistry was conducted on MTG paraffin-embedded tissue microarrays (TMAs) comprising 29 HD and 35 neurologically normal cases to compare the immunoreactivity patterns of key astrocytic proteins (glial fibrillary acidic protein, GFAP; inwardly rectifying potassium channel 4.1, Kir4.1; glutamate transporter-1, GLT-1; aquaporin-4, AQP4), key microglial proteins (ionised calcium-binding adapter molecule-1, IBA-1; human leukocyte antigen (HLA)-DR; transmembrane protein 119, TMEM119; purinergic receptor P2RY12, P2RY12), and indicators of proliferation (Ki-67; proliferative cell nuclear antigen, PCNA). Our findings demonstrate an upregulation of GFAP+ protein expression attributed to the presence of more GFAP+ expressing cells in HD, which correlated with greater cortical mutant huntingtin (mHTT) deposition. In contrast, Kir4.1, GLT-1, and AQP4 immunoreactivity levels were unchanged in HD. We also demonstrate an increased number of IBA-1+ and TMEM119+ microglia with somal enlargement. IBA-1+, TMEM119+, and P2RY12+ reactive microglia immunophenotypes were also identified in HD, evidenced by the presence of rod-shaped, hypertrophic, and dystrophic microglia. In HD cases, IBA-1+ cells contained either Ki-67 or PCNA, whereas GFAP+ astrocytes were devoid of proliferative nuclei. These findings suggest cortical microgliosis may be driven by proliferation in HD, supporting the hypothesis of microglial proliferation as a feature of HD pathophysiology. In contrast, astrocytes in HD demonstrate an altered GFAP expression profile that is associated with the degree of mHTT deposition.


Subject(s)
Astrocytes , Cell Proliferation , Huntington Disease , Microglia , Humans , Huntington Disease/metabolism , Huntington Disease/pathology , Microglia/metabolism , Microglia/pathology , Astrocytes/metabolism , Astrocytes/pathology , Male , Female , Middle Aged , Cell Proliferation/physiology , Adult , Aged , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Calcium-Binding Proteins/metabolism , Gliosis/metabolism , Gliosis/pathology , Glial Fibrillary Acidic Protein/metabolism , Membrane Proteins , Microfilament Proteins
2.
Cell Commun Signal ; 22(1): 30, 2024 01 11.
Article in English | MEDLINE | ID: mdl-38212807

ABSTRACT

Glioblastoma is the most common and aggressive primary brain tumour in adults. The development of anti-brain cancer agents are challenged by the blood-brain barrier and the resistance conferred by the local tumour microenvironment. Heptamethine cyanine dyes (HMCDs) are a class of near-infrared fluorescence compounds that have recently emerged as promising agents for drug delivery. We conjugated palbociclib, a cyclin-dependent kinase (CDK) 4/6 inhibitor, to an HMCD, MHI-148, and conducted drug activity analysis on primary patient-derived glioblastoma cell lines. In addition to the expected cytostatic activity, our in vitro studies revealed that palbociclib-MHI-148 conjugate resulted in an almost 100-fold increase in cytotoxicity compared to palbociclib alone. This shift of palbociclib from cytostatic to cytotoxic when conjugated to MHI-148 was due to increased DNA damage, as indicated by an increase in γH2AX foci, followed by an increased expression of key extrinsic apoptosis genes, including TP53, TNFR1, TRAIL, FADD and caspase 8. In addition, we observed a time-dependent increase in the cell surface expression of TNFR1, consistent with an observed increase in the secretion TNFα, followed by TNFR1 endocytosis at 48 h. The treatment of patient GBM cells with the palbociclib-MHI-148 conjugate prevented TNFα-induced NFκB translocation, suggesting conjugate-induced TNFR1 signalling favoured the TNFR1-mediated apoptotic response rather than the pro-inflammatory response pathway. Notably, pharmacological inhibition of endocytosis of TNFR1, and siRNA-knockdown of TNFR1 reversed the palbociclib-MHI-148-induced cell death. These results show a novel susceptibility of glioblastoma cells to TNFR1-dependent apoptosis, dependent on inhibition of canonical NFκB signalling using our previously reported palbociclib-HMCD conjugate. Video Abstract.


Subject(s)
Antineoplastic Agents , Carbocyanines , Cytostatic Agents , Glioblastoma , Indoles , Piperazines , Pyridines , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Cytostatic Agents/pharmacology , Cytostatic Agents/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/metabolism , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism
3.
Cytometry A ; 103(6): 518-527, 2023 06.
Article in English | MEDLINE | ID: mdl-36786336

ABSTRACT

Current analysis techniques available for migration assays only provide quantitative measurements for overall migration. However, the potential of regional migration analyses can open further insight into migration patterns and more avenues of experimentation with the same assays. Previously, we developed an analysis pipeline utilizing the finite element (FE) method to show its potential in analyzing glioblastoma (GBM) tumorsphere migration, especially in characterizing regional changes in the migration pattern. This study aims to streamline and further automate the analysis system by integrating the machine-learning-based U-Net segmentation with the FE method. Our U-Net-based segmentation achieved a 98% accuracy in segmenting our tumorspheres. From the segmentations, FE models made up of 3D hexahedral elements were generated, and the migration patterns of the tumorspheres were analyzed under treatments B and C (under non-disclosure agreements). Our results show that our overall migration analysis correlated very strongly (R2 of 0.9611 and 0.9986 for treatments B and C, respectively) with ImageJ's method of migration area analysis, which is the most common method of tumorsphere migration analysis. Additionally, we were able to quantitatively represent the regional migration patterns in our FE models, which the methods purely based on segmentations could not do. Moreover, the new pipeline improved the efficiency and accessibility of the initial pipeline by implementing machine learning-based automated segmentation onto a mainly open-sourced FE analysis platform. In conclusion, our algorithm enables the development of a high-content and high-throughput in vitro screening platform to elucidate anti-migratory molecules that may reduce the invasiveness of these malignant tumors.


Subject(s)
Glioblastoma , Machine Learning , Humans , Glioblastoma/pathology , Algorithms
4.
Chem Biol Drug Des ; 101(3): 696-716, 2023 03.
Article in English | MEDLINE | ID: mdl-36323652

ABSTRACT

The development of chemotherapies for glioblastoma is hindered by their limited bioavailability and toxicity on normal brain function. To overcome these limitations, we investigated the structure-dependent activity of heptamethine cyanine dyes (HMCD), a group of tumour-specific and BBB permeable near-infrared fluorescent dyes, in both commercial (U87MG) and patient-derived GBM cell lines. HMCD analogues with strongly ionisable sulphonic acid groups were not taken up by patient-derived GBM cells, but were taken up by the U87MG cell line. HMCD uptake relies on a combination of transporter uptake through organic anion-transporting polypeptides (OATPs) and endocytosis into GBM cells. The uptake of HMCDs was not affected by p-glycoprotein efflux in GBM cells. Finally, we demonstrate structure-dependent cytotoxic activity at high concentrations (EC50 : 1-100 µM), likely due to mitochondrial damage-induced apoptosis. An in vivo orthotopic glioblastoma model highlights tumour-specific accumulation of our lead HMCD, MHI-148, for up to 7 days following a single intraperitoneal injection. These studies suggest that strongly ionisable groups like sulphonic acids hamper the cellular uptake of HMCDs in patient-derived GBM cell lines, highlighting cell line-specific differences in HMCD uptake. We envisage these findings will help in the design and structural modifications of HMCDs for drug-delivery applications for glioblastoma.


Subject(s)
Antineoplastic Agents , Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Fluorescent Dyes , Brain Neoplasms/drug therapy
5.
Neurooncol Adv ; 4(1): vdac166, 2022.
Article in English | MEDLINE | ID: mdl-36382105

ABSTRACT

Background: Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Whilst the role of the efflux transporters are well established in GBM, the expression and function of uptake transporters, such as the organic anion transporting polypeptide (OATP) family, are not well understood. OATPs possess broad substrate specificity that includes anti-cancer agents; therefore, we sought to investigate the expression of four OATP isoforms in human GBM cell types using patient tumor tissue. Methods: We used fluorescent immunohistochemical labeling of paraffin-embedded surgically resected tissues and single-cell image analysis methods to explore the expression of the OATP isoforms in different tumor cell types through co-labeling with cell-type specific markers, such as IBA1 (pan-myeloid), GFAP (tumor cell), PDGFRß (stromal cell), and UEA-1-lectin (endothelial). Results: We found significant over-expression of all the OATP isoforms (OATP1A2, 2B1, 1C1 and 4A1) in GBM tumor sections when compared to non-neoplastic brain. A single-cell image analysis revealed that OATPs were significantly upregulated throughout the tumor parenchyma, with significantly higher expression found on lectin-positive blood vessels and IBA1-positive myeloid cells in GBM compared to non-tumor brain tissue. Qualitative analysis of the four OATP isoforms demonstrated greater expression of OATP4A1 in peri-necrotic regions of GBM tissue, which correlated with hypoxia-related markers within the Ivy GAP RNAseq dataset. Conclusion: Here, we demonstrate, for the first time, the protein expression of four OATPs in human GBM tissue, including upregulation within the tumor microenvironment by myeloid cells and tumor vasculature, and isoform-specific upregulation within hypoxic niches.

6.
PLoS One ; 17(11): e0277658, 2022.
Article in English | MEDLINE | ID: mdl-36399706

ABSTRACT

Parkinson's disease (PD) is a progressive, neurodegenerative disorder characterised by the abnormal accumulation of α-synuclein (α-syn) aggregates. Central to disease progression is the gradual spread of pathological α-syn. α-syn aggregation is closely linked to progressive neuron loss. As such, clearance of α-syn aggregates may slow the progression of PD and lead to less severe symptoms. Evidence is increasing that non-neuronal cells play a role in PD and other synucleinopathies such as Lewy body dementia and multiple system atrophy. Our previous work has shown that pericytes-vascular mural cells that regulate the blood-brain barrier-contain α-syn aggregates in human PD brains. Here, we demonstrate that pericytes efficiently internalise fibrillar α-syn irrespective of being in a monoculture or mixed neuronal cell culture. Pericytes cleave fibrillar α-syn aggregates (Fibrils, Ribbons, fibrils65, fibrils91 and fibrils110), with cleaved α-syn remaining present for up to 21 days. The number of α-syn aggregates/cell and average aggregate size depends on the type of strain, but differences disappear within 5 five hours of treatment. Our results highlight the role brain vasculature may play in reducing α-syn aggregate burden in PD.


Subject(s)
Lewy Body Disease , Parkinson Disease , Humans , alpha-Synuclein/metabolism , Pericytes/metabolism , Lewy Body Disease/metabolism , Parkinson Disease/pathology , Neurons/metabolism
7.
Front Neurosci ; 16: 994251, 2022.
Article in English | MEDLINE | ID: mdl-36440264

ABSTRACT

Traumatic brain injury (TBI) is defined as brain damage due to an external force that negatively impacts brain function. Up to 90% of all TBI are considered in the mild severity range (mTBI) but there is still no therapeutic solution available. Therefore, further understanding of the mTBI pathology is required. To assist with this understanding, we developed a cell injury device (CID) based on a dielectric elastomer actuator (DEA), which is capable of modeling mTBI via injuring cultured cells with mechanical stretching. Our injury model is the first to use patient-derived brain pericyte cells, which are ubiquitous cells in the brain involved in injury response. Pericytes were cultured in our CIDs and mechanically strained up to 40%, and by at least 20%, prior to gene expression analysis. Our injury model is a platform capable of culturing and stretching primary human brain pericytes. The heterogeneous response in gene expression changes in our result may suggest that the genes implicated in pathological changes after mTBI could be a patient-dependent response, but requires further validation. The results of this study demonstrate that our CID is a suitable tool for simulating mTBI as an in vitro stretch injury model, that is sensitive enough to induce responses from primary human brain pericytes due to mechanical impacts.

8.
Commun Biol ; 5(1): 235, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35301433

ABSTRACT

Platelet-derived growth factor-BB (PDGF-BB):PDGF receptor-ß (PDGFRß) signalling in brain pericytes is critical to the development, maintenance and function of a healthy blood-brain barrier (BBB). Furthermore, BBB impairment and pericyte loss in Alzheimer's disease (AD) is well documented. We found that PDGF-BB:PDGFRß signalling components were altered in human AD brains, with a marked reduction in vascular PDGFB. We hypothesised that reduced PDGF-BB:PDGFRß signalling in pericytes may impact on the BBB. We therefore tested the effects of PDGF-BB on primary human brain pericytes in vitro to define pathways related to BBB function. Using pharmacological inhibitors, we dissected distinct aspects of the PDGF-BB response that are controlled by extracellular signal-regulated kinase (ERK) and Akt pathways. PDGF-BB promotes the proliferation of pericytes and protection from apoptosis through ERK signalling. In contrast, PDGF-BB:PDGFRß signalling through Akt augments pericyte-derived inflammatory secretions. It may therefore be possible to supplement PDGF-BB signalling to stabilise the cerebrovasculature in AD.


Subject(s)
Alzheimer Disease , Pericytes , Alzheimer Disease/metabolism , Becaplermin/metabolism , Becaplermin/pharmacology , Brain/metabolism , Humans , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/pharmacology
9.
Nat Protoc ; 17(2): 190-221, 2022 02.
Article in English | MEDLINE | ID: mdl-35022619

ABSTRACT

When modeling disease in the laboratory, it is important to use clinically relevant models. Patient-derived human brain cells grown in vitro to study and test potential treatments provide such a model. Here, we present simple, highly reproducible coordinated procedures that can be used to routinely culture most cell types found in the human brain from single neurosurgically excised brain specimens. The cell types that can be cultured include dissociated cultures of neurons, astrocytes, microglia, pericytes and brain endothelial and neural precursor cells, as well as explant cultures of the leptomeninges, cortical slice cultures and brain tumor cells. The initial setup of cultures takes ~2 h, and the cells are ready for further experiments within days to weeks. The resulting cells can be studied as purified or mixed population cultures, slice cultures and explant-derived cultures. This protocol therefore enables the investigation of human brain cells to facilitate translation of neuroscience research to the clinic.


Subject(s)
Neural Stem Cells
10.
Methods Mol Biol ; 2389: 125-154, 2022.
Article in English | MEDLINE | ID: mdl-34558008

ABSTRACT

The discovery, in 1998, that the adult human brain contains at least two populations of progenitor cells and that progenitor cells are upregulated in response to a range of degenerative brain diseases has raised hopes for their use in replacing dying brain cells. Since these early findings, the race has been on to understand the biology of progenitor cells in the human brain, and they have now been isolated and studied in many major neurodegenerative diseases. Before these cells can be exploited for cell replacement purposes, it is important to understand how to (1) locate them, (2) label them, (3) determine what receptors they express, (4) isolate them, and (5) examine their electrophysiological properties when differentiated. In this chapter we have described the methods we use for studying progenitor cells in the adult human brain and in particular the tissue processing, immunohistochemistry, autoradiography, progenitor cell culture, and electrophysiology on brain cells. The Neurological Foundation of New Zealand Human Brain Bank has been receiving human tissue for approximately 25 years during which time we have developed a number of unique ways to examine and isolate progenitor cells from resected surgical specimens as well as from postmortem brain tissue. There are ethical and technical considerations that are unique to working with human brain tissue, and these, as well as the processing of this tissue and the culturing of it for the purpose of studying progenitor cells, are the topic of this chapter.


Subject(s)
Neural Stem Cells , Adult , Adult Stem Cells , Brain , Cell Culture Techniques , Cell Differentiation , Humans , Immunohistochemistry
11.
Neurooncol Adv ; 3(1): vdab031, 2021.
Article in English | MEDLINE | ID: mdl-34286275

ABSTRACT

BACKGROUND: Microglia and tumor-associated macrophages (TAMs) constitute up to half of the total tumor mass of glioblastomas. Despite these myeloid populations being ontogenetically distinct, they have been largely conflated. Recent single-cell transcriptomic studies have identified genes that distinguish microglia from TAMs. Here we investigated whether the translated proteins of genes enriched in microglial or TAM populations can be used to differentiate these myeloid cells in immunohistochemically stained human glioblastoma tissue. METHODS: Tissue sections from resected low-grade, meningioma, and glioblastoma (grade IV) tumors and epilepsy tissues were immunofluorescently triple-labeled for Iba1 (pan-myeloid marker), CD14 or CD163 (preferential TAM markers), and either P2RY12 or TMEM119 (microglial-specific markers). Using a single-cell-based image analysis pipeline, we quantified the abundance of each marker within single myeloid cells, allowing the identification and analysis of myeloid populations. RESULTS: P2RY12 and TMEM119 successfully discriminated microglia from TAMs in glioblastoma. In contrast, CD14 and CD163 expression were not restricted to invading TAMs and were upregulated by tumor microglia. Notably, a higher ratio of microglia to TAMs significantly correlated with increased patient survival. CONCLUSIONS: We demonstrate the validity of previously defined microglial-specific genes P2RY12 and TMEM119 as robust discriminators of microglia and TAMs at the protein level in human tissue. Moreover, our data suggest that a higher proportion of microglia may be beneficial for patient survival in glioblastoma. Accordingly, this tissue-based method for myeloid population differentiation could serve as a useful prognostic tool.

12.
Front Oncol ; 11: 654921, 2021.
Article in English | MEDLINE | ID: mdl-34141613

ABSTRACT

Effective cancer therapeutics for brain tumors must be able to cross the blood-brain barrier (BBB) to reach the tumor in adequate quantities and overcome the resistance conferred by the local tumor microenvironment. Clinically approved chemotherapeutic agents have been investigated for brain neoplasms, but despite their effectiveness in peripheral cancers, failed to show therapeutic success in brain tumors. This is largely due to their poor bioavailability and specificity towards brain tumors. A targeted delivery system might improve the efficacy of the candidate compounds by increasing the retention time in the tumor tissue, and minimizing the numerous side effects associated with the non-specific distribution of the chemotherapy agent. Heptamethine cyanine dyes (HMCDs) are a class of near-infrared fluorescence (NIRF) compounds that have recently emerged as promising agents for drug delivery. Initially explored for their use in imaging and monitoring neoplasms, their tumor-targeting properties have recently been investigated for their use as drug carrier systems. This review will explore the recent developments in the tumour-targeting properties of a specific group of NIRF cyanine dyes and the preclinical evidence for their potential as drug-delivery systems in the treatment of primary and metastatic brain tumors.

13.
STAR Protoc ; 2(2): 100518, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34027479

ABSTRACT

Microglia are the primary innate immune effectors of the central nervous system. Although numerous protocols have been developed to isolate fetal mouse microglia, the isolation of adult mouse microglia has proven more difficult. Here, we present a simple, widely accessible protocol to isolate pure microglia cultures from 4- to 14-month-old mouse brains using their adherent properties in vitro. These isolated microglia recapitulate the adherent properties of adult human microglia and present a more suitable model for studying age-related diseases. For complete details on the use and execution of this protocol in adult human microglia, please refer to Rustenhoven et al. (2016).


Subject(s)
Aging/immunology , Brain/immunology , Cell Separation , Microglia/immunology , Animals , Brain/cytology , Mice , Microglia/cytology
14.
Brain Commun ; 2(2): fcaa171, 2020.
Article in English | MEDLINE | ID: mdl-33215086

ABSTRACT

The ability to characterize and study primary neurons isolated directly from the adult human brain would greatly advance neuroscience research. However, significant challenges such as accessibility of human brain tissue and the lack of a robust neuronal cell culture protocol have hampered its progress. Here, we describe a simple and reproducible method for the isolation and culture of functional adult human neurons from neurosurgical brain specimens. In vitro, adult human neurons form a dense network and express a plethora of mature neuronal and synaptic markers. Most importantly, for the first time, we demonstrate the re-establishment of mature neurophysiological properties in vitro, such as repetitive fast-spiking action potentials, and spontaneous and evoked synaptic activity. Together, our dissociated and slice culture systems enable studies of adult human neurophysiology and gene expression under normal and pathological conditions and provide a high-throughput platform for drug testing on brain cells directly isolated from the adult human brain.

15.
J Clin Neurosci ; 80: 242-249, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33099354

ABSTRACT

Glioblastoma (GBM) is a malignant cerebral neoplasm carrying poor prognosis. The importance of extent of resection (EoR) in GBM patient outcomes has been argued in the literature. Previous studies included tumors in eloquent regions of the brain. This confounds the role of EoR by including patients with intrinsically worse outcomes but will be over-represented in the reduced EoR category. In a homogenous group of patients in whom GTR was considered achievable, we investigated the effect of increasing EoR on survival. A retrospective review of 51 patients was undertaken. Quantitative, volumetric analysis of pre-operative and post-operative magnetic resonance image was compared with corresponding clinical details. The primary outcome measured was post-operative overall survival. Median overall survival was 18.3 months for GTR patients compared to 11.6 months for non-GTR (p = 0.025). Median pre-operative contrast-enhancing tumor volume for GTR patients was 54.7 cm3 and 24.9 cm3 for non-GTR. Post-operative median residual tumor volume was 1.1 cm3 in the non-GTR cohort. In multivariate analyses, GTR (HR [95% CI] = 0.973 [0.954-0.994], p = 0.00559) and increasing EoR (HR [95% CI] = 0.964 [0.944-0.985], p = 0.000665) remained predictors of survival. Centile dichotomization of EoR revealed 74% (HR [95% CI] = 0.351 [0.128-0.958], p = 0.0409) as the lowest threshold conferring statistically significant survival benefit. Where technically feasible, both GTR and EoR remained as independent prognostic factors for survival. GTR remains the gold standard for surgical treatment of GBM in patients, 74% being the minimum EoR required to confer survival benefit.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/surgery , Glioblastoma/diagnostic imaging , Glioblastoma/surgery , Neurosurgical Procedures/methods , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Cohort Studies , Female , Humans , Male , Middle Aged , Neurosurgical Procedures/mortality , Prognosis , Retrospective Studies , Survival Rate/trends , Tumor Burden , Young Adult
16.
Bioorg Med Chem Lett ; 30(14): 127252, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32527552

ABSTRACT

We describe the synthesis and in vitro activity of drug-dye conjugate 1, which is a combination of the PARP inhibitor rucaparib and heptamethine cyanine dye IR-786. The drug-dye conjugate 1 was evaluated in three different patient-derived glioblastoma cell lines and showed strong cytotoxic activity with nanomolar potency (EC50: 128 nM), which was a 780 fold improvement over rucaparib itself. We also observe a synergistic effect of 1 with temozolomide (TMZ), the standard drug for treatment for glioblastoma even though these cell lines were resistant to TMZ treatment. We envisage such conjugates to be worth exploring for their utility in the treatment of various brain cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Carbocyanines/pharmacology , Glioblastoma/drug therapy , Indoles/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carbocyanines/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Indoles/chemistry , Molecular Structure , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Structure-Activity Relationship
17.
Bioconjug Chem ; 31(7): 1724-1739, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32530288

ABSTRACT

This review covers the application of heptamethine cyanine dye (HMCD) mediated drug delivery. A relatively small number of HMCDs possess tumor targeting abilities, and this has spurred interest from research groups to explore them as drug delivery systems. Their tumor selectivity is primarily attributed to their uptake by certain isoforms of organic anion transporting polypeptides (OATPs) which are overexpressed in cancer tissues, although there are other possible mechanisms for the observed selectivity still under investigation. This specificity is confirmed using various cancer cell lines and is accompanied by moderate cytotoxicity. Their retention in tumor tissue is facilitated by the formation of albumin adducts as revealed by published mechanistic studies. HMCDs are also organelle selective dyes with specificity toward mitochondria and lysosomes, and with absorption and emission in the near-infrared region. This makes them valuable tools for biomedical imaging, especially in the field of fluorescence-guided tumor surgery. Furthermore, conjugating antitumor agents to HMCDs is providing novel drugs that await clinical testing. HMCD development as theranostic agents with dual tumor targeting and treatment capability signals a new approach to overcome drug resistance (mediated through evasion of efflux pumps) and systemic toxicity, the two parameters which have long plagued drug discovery.


Subject(s)
Antineoplastic Agents/administration & dosage , Carbocyanines/administration & dosage , Coloring Agents/administration & dosage , Drug Delivery Systems/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Burkitt Lymphoma/drug therapy , Carbocyanines/pharmacology , Carbocyanines/therapeutic use , Drug Discovery , Drug Resistance, Neoplasm/drug effects , Female , Humans , Kidney Neoplasms/drug therapy , Male , Precision Medicine , Prostatic Neoplasms/drug therapy
18.
J Neurophysiol ; 123(3): 945-965, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31995449

ABSTRACT

The human brain shows remarkable complexity in its cellular makeup and function, which are distinct from nonhuman species, signifying the need for human-based research platforms for the study of human cellular neurophysiology and neuropathology. However, the use of adult human brain tissue for research purposes is hampered by technical, methodological, and accessibility challenges. One of the major problems is the limited number of in vitro systems that, in contrast, are readily available from rodent brain tissue. With recent advances in the optimization of protocols for adult human brain preparations, there is a significant opportunity for neuroscientists to validate their findings in human-based systems. This review addresses the methodological aspects, advantages, and disadvantages of human neuron in vitro systems, focusing on the unique properties of human neurons and synapses in neocortical microcircuits. These in vitro models provide the incomparable advantage of being a direct representation of the neurons that have formed part of the human brain until the point of recording, which cannot be replicated by animal models nor human stem-cell systems. Important distinct cellular mechanisms are observed in human neurons that may underlie the higher order cognitive abilities of the human brain. The use of human brain tissue in neuroscience research also raises important ethical, diversity, and control tissue limitations that need to be considered. Undoubtedly however, these human neuron systems provide critical information to increase the potential of translation of treatments from the laboratory to the clinic in a way animal models are failing to provide.


Subject(s)
Neocortex/physiology , Nerve Net/physiology , Neuronal Plasticity/physiology , Organ Culture Techniques , Synapses/physiology , Humans
19.
Bioorg Med Chem Lett ; 29(18): 2617-2621, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31378572

ABSTRACT

We describe the synthesis of drug-dye conjugate 1 between anaplastic lymphoma kinase inhibitor Crizotinib and heptamethine cyanine dye IR-786. The drug-dye conjugate 1 was evaluated in three different patient-derived glioblastoma cell lines and showed potent cytotoxic activity with nanomolar potency (EC50: 50.9 nM). We also demonstrate evidence for antiproliferative activity of 1 with single digit nanomolar potency (IC50: 4.7 nM). Furthermore, the cytotoxic effects conveyed a dramatic, 110-fold improvement over Crizotinib. This improvement was even more pronounced (492-fold) when 1 was combined with Temozolomide, the standard drug for treatment for glioblastoma. This work lays the foundation for future exploration of similar tyrosine kinase inhibitor drug-dye conjugates for the treatment of glioblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Carbocyanines/pharmacology , Crizotinib/pharmacology , Cytostatic Agents/pharmacology , Fluorescent Dyes/pharmacology , Glioblastoma/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Carbocyanines/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Crizotinib/chemistry , Cytostatic Agents/chemical synthesis , Cytostatic Agents/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Glioblastoma/diagnostic imaging , Glioblastoma/metabolism , Humans , Molecular Structure , Optical Imaging , Structure-Activity Relationship
20.
Mol Neurodegener ; 13(1): 44, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30124174

ABSTRACT

BACKGROUND: Microglia play critical roles in the brain during homeostasis and pathological conditions. Understanding the molecular events underpinning microglial functions and activation states will further enable us to target these cells for the treatment of neurological disorders. The transcription factor PU.1 is critical in the development of myeloid cells and a major regulator of microglial gene expression. In the brain, PU.1 is specifically expressed in microglia and recent evidence from genome-wide association studies suggests that reductions in PU.1 contribute to a delayed onset of Alzheimer's disease (AD), possibly through limiting neuroinflammatory responses. METHODS: To investigate how PU.1 contributes to immune activation in human microglia, microarray analysis was performed on primary human mixed glial cultures subjected to siRNA-mediated knockdown of PU.1. Microarray hits were confirmed by qRT-PCR and immunocytochemistry in both mixed glial cultures and isolated microglia following PU.1 knockdown. To identify attenuators of PU.1 expression in microglia, high throughput drug screening was undertaken using a compound library containing FDA-approved drugs. NanoString and immunohistochemistry was utilised to investigate the expression of PU.1 itself and PU.1-regulated mediators in primary human brain tissue derived from neurologically normal and clinically and pathologically confirmed cases of AD. RESULTS: Bioinformatic analysis of gene expression upon PU.1 silencing in mixed glial cultures revealed a network of modified AD-associated microglial genes involved in the innate and adaptive immune systems, particularly those involved in antigen presentation and phagocytosis. These gene changes were confirmed using isolated microglial cultures. Utilising high throughput screening of FDA-approved compounds in mixed glial cultures we identified the histone deacetylase inhibitor vorinostat as an effective attenuator of PU.1 expression in human microglia. Further characterisation of vorinostat in isolated microglial cultures revealed gene and protein changes partially recapitulating those seen following siRNA-mediated PU.1 knockdown. Lastly, we demonstrate that several of these PU.1-regulated genes are expressed by microglia in the human AD brain in situ. CONCLUSIONS: Collectively, these results suggest that attenuating PU.1 may be a valid therapeutic approach to limit microglial-mediated inflammatory responses in AD and demonstrate utility of vorinostat for this purpose.


Subject(s)
Alzheimer Disease/metabolism , Gene Expression Regulation/physiology , Microglia/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Microglia/drug effects , Vorinostat/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...