Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 132
Filtrer
1.
iScience ; 27(8): 110463, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39129829

RÉSUMÉ

During malaria infection, Plasmodium sporozoites, the fast-moving stage of the parasite, are injected by a mosquito into the skin of the mammalian host. In the skin, sporozoites need to migrate through the dermal tissue to enter the blood vessel. Sporozoite motility is critical for infection but not well understood. Here, we used collagen hydrogels with tunable fiber structures, as an in vitro model for the skin. After injecting sporozoites into the hydrogel, we analyzed their motility in three-dimension (3D). We found that sporozoites demonstrated chiral motility, in that they mostly follow right-handed helical trajectories. In high-concentration collagen gel, sporozoites have lower instantaneous speed, but exhibit straighter tracks compared to low-concentration collagen gel, which leads to longer net displacement and faster dissemination. Taken together, our study indicates an inner mechanism for sporozoites to adapt to the environment, which could help with their successful exit from the skin tissue.

2.
Adv Funct Mater ; 34(30)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39131199

RÉSUMÉ

Plasmonic nanomaterials bearing targeting ligands are of great interest for surface-enhanced Raman scattering (SERS)-based bioimaging applications. However, the practical utility of SERS-based imaging strategies has been hindered by the lack of a straightforward method to synthesize highly sensitive SERS-active nanostructures with high yield and efficiency. In this work, leveraging DNA origami principles, we report the first-in-class design of a SERS-based plasmonically coupled nanoprobe for targeted cancer imaging (SPECTRA). The nanoprobe harnesses a cancer cell targeting DNA aptamer sequence and vibrational tag with stretching frequency in the cell-silent Raman window. Through the integration of aptamer sequence specific for DU145 cells, we show the unique capabilities of SPECTRA for targeted imaging of DU145 cells. Our results demonstrate that the scalability, cost-effectiveness, and reproducibility of this method of fabrication of SERS nanoprobes can serve as a versatile platform for creating nanoprobes with broad applications in the fields of cancer biology and biomedical imaging.

3.
ACS Sens ; 9(7): 3794-3804, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-38976969

RÉSUMÉ

Articular cartilage is a complex tissue, and early detection of osteoarthritis (OA) is crucial for effective treatment. However, current imaging modalities lack molecular specificity and primarily detect late-stage changes. In this study, we propose the use of spatially offset Raman spectroscopy (SORS) for noninvasive, depth-dependent, and molecular-specific diagnostics of articular cartilage. We demonstrate the potential of SORS to penetrate deep layers of cartilage, providing a comprehensive understanding of disease progression. Our SORS measurements were characterized and validated through mechanical and histological techniques, revealing strong correlations between spectroscopic measurements and both Young's modulus and depth of cartilage damage. By longitudinally monitoring enzymatically degraded condyles, we further developed a depth-dependent damage-tracking method. Our analysis revealed distinct components related to sample depth and glycosaminoglycan (GAG) changes, offering a comprehensive picture of cartilage health. Collectively, these findings highlight the potential of SORS as a valuable tool for enhancing OA management and improving patient outcomes.


Sujet(s)
Cartilage articulaire , Arthrose , Analyse spectrale Raman , Analyse spectrale Raman/méthodes , Cartilage articulaire/imagerie diagnostique , Cartilage articulaire/anatomopathologie , Arthrose/diagnostic , Animaux , Glycosaminoglycanes/analyse , Glycosaminoglycanes/composition chimique , Humains , Bovins
4.
bioRxiv ; 2024 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-38712120

RÉSUMÉ

Cellular morphology, shaped by various genetic and environmental influences, is pivotal to studying experimental cell biology, necessitating precise measurement and analysis techniques. Traditional approaches, which rely on geometric metrics derived from stained images, encounter obstacles stemming from both the imaging and analytical domains. Staining processes can disrupt the cell's natural state and diminish accuracy due to photobleaching, while conventional analysis techniques, which categorize cells based on shape to discern pathophysiological conditions, often fail to capture the continuous and asynchronous nature of biological processes such as cell differentiation, immune responses, and cancer progression. In this work, we propose the use of quantitative phase imaging for morphological assessment due to its label-free nature. For analysis, we repurposed the genomic analysis toolbox to perform trajectory inference analysis purely based on morphology information. We applied the developed framework to study the progression of leukemia and breast cancer metastasis. Our approach revealed a clear pattern of morphological evolution tied to the diseases' advancement, highlighting the efficacy of our method in identifying functionally significant shape changes where conventional techniques falter. This advancement offers a fresh perspective on analyzing cellular morphology and holds significant potential for the broader research community, enabling a deeper understanding of complex biological dynamics.

5.
Spectrochim Acta A Mol Biomol Spectrosc ; 317: 124387, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-38704999

RÉSUMÉ

The development of tools that can provide a holistic picture of the evolution of the tumor microenvironment in response to intermittent fasting on the prevention of breast cancer is highly desirable. Here, we show, for the first time, the use of label-free Raman spectroscopy to reveal biomolecular alterations induced by intermittent fasting in the tumor microenvironment of breast cancer using a dimethyl-benzanthracene induced rat model. To quantify biomolecular alterations in the tumor microenvironment, chemometric analysis of Raman spectra obtained from untreated and treated tumors was performed using multivariate curve resolution-alternative least squares and support vector machines. Raman measurements revealed remarkable and robust differences in lipid, protein, and glycogen content prior to morphological manifestations in a dynamically changing tumor microenvironment, consistent with the proteomic changes observed by quantitative mass spectrometry. Taken together with its non-invasive nature, this research provides prospective evidence for the clinical translation of Raman spectroscopy to identify biomolecular variations in the microenvironment induced by intermittent fasting for the prevention of breast cancer, providing new perspectives on the specific molecular effects in the tumorigenesis of breast cancer.


Sujet(s)
Tumeurs du sein , Jeûne , Analyse spectrale Raman , Microenvironnement tumoral , Analyse spectrale Raman/méthodes , Animaux , Femelle , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tumeurs du sein/prévention et contrôle , Tumeurs du sein/anatomopathologie , Rats , Modèles animaux de maladie humaine , 7,12-Diméthyl-benzo[a]anthracène/toxicité , Tumeurs expérimentales de la mamelle/prévention et contrôle , Tumeurs expérimentales de la mamelle/induit chimiquement , Tumeurs expérimentales de la mamelle/anatomopathologie , Rat Sprague-Dawley , Jeûne intermittent
6.
J Biomed Opt ; 29(Suppl 2): S22706, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38638450

RÉSUMÉ

Significance: Three-dimensional quantitative phase imaging (QPI) has rapidly emerged as a complementary tool to fluorescence imaging, as it provides an objective measure of cell morphology and dynamics, free of variability due to contrast agents. It has opened up new directions of investigation by providing systematic and correlative analysis of various cellular parameters without limitations of photobleaching and phototoxicity. While current QPI systems allow the rapid acquisition of tomographic images, the pipeline to analyze these raw three-dimensional (3D) tomograms is not well-developed. We focus on a critical, yet often underappreciated, step of the analysis pipeline that of 3D cell segmentation from the acquired tomograms. Aim: We report the CellSNAP (Cell Segmentation via Novel Algorithm for Phase Imaging) algorithm for the 3D segmentation of QPI images. Approach: The cell segmentation algorithm mimics the gemstone extraction process, initiating with a coarse 3D extrusion from a two-dimensional (2D) segmented mask to outline the cell structure. A 2D image is generated, and a segmentation algorithm identifies the boundary in the x-y plane. Leveraging cell continuity in consecutive z-stacks, a refined 3D segmentation, akin to fine chiseling in gemstone carving, completes the process. Results: The CellSNAP algorithm outstrips the current gold standard in terms of speed, robustness, and implementation, achieving cell segmentation under 2 s per cell on a single-core processor. The implementation of CellSNAP can easily be parallelized on a multi-core system for further speed improvements. For the cases where segmentation is possible with the existing standard method, our algorithm displays an average difference of 5% for dry mass and 8% for volume measurements. We also show that CellSNAP can handle challenging image datasets where cells are clumped and marred by interferogram drifts, which pose major difficulties for all QPI-focused AI-based segmentation tools. Conclusion: Our proposed method is less memory intensive and significantly faster than existing methods. The method can be easily implemented on a student laptop. Since the approach is rule-based, there is no need to collect a lot of imaging data and manually annotate them to perform machine learning based training of the model. We envision our work will lead to broader adoption of QPI imaging for high-throughput analysis, which has, in part, been stymied by a lack of suitable image segmentation tools.


Sujet(s)
Traitement d'image par ordinateur , Imagerie tridimensionnelle , Humains , Traitement d'image par ordinateur/méthodes , Imagerie tridimensionnelle/méthodes , , Algorithmes , Imagerie optique
7.
Biosens Bioelectron ; 254: 116199, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38492362

RÉSUMÉ

Genetic and epigenetic modifications are linked to the activation of oncogenes and inactivation of tumor suppressor genes. Likewise, the associated molecular alternations can best inform precision medicine for personalized tumor treatment. Therefore, performing characterization of genetic and epigenetic alternations at the molecular level represents a crucial step in early diagnosis and/or therapeutics of cancer. However, the prevailing methods for DNA analysis involve a series of tedious and complicated steps, in which important genetic and epigenetic information could be lost or altered. To provide a potential approach for non-invasive, direct, and efficient DNA analysis, herein, we present a promising strategy for label-free molecular profiling of serum DNA in its pristine form by fusing surface-enhanced Raman spectroscopy with machine learning on a superior plasmonic nanostructured platform. Using DNA methylation and single-point mutation as two case studies, the presented strategy allows a well-balanced sensitive and specific detection of epigenetic and genetic changes at the single-nucleotide level in serum. We envision the presented label-free strategy could serve as a versatile tool for direct molecular profiling in pristine forms of a wide range of biological markers and aid biomedical diagnostics as well as therapeutics.


Sujet(s)
Techniques de biocapteur , Nanoparticules métalliques , Tumeurs , Humains , ADN/composition chimique , Épigenèse génétique , Méthylation de l'ADN , Analyse spectrale Raman/méthodes , Tumeurs/génétique , Nanoparticules métalliques/composition chimique
8.
Adv Biol (Weinh) ; 8(5): e2400020, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38548657

RÉSUMÉ

Understanding the intricate processes of neuronal growth, degeneration, and neurotoxicity is paramount for unraveling nervous system function and holds significant promise in improving patient outcomes, especially in the context of chemotherapy-induced peripheral neuropathy (CIPN). These processes are influenced by a broad range of entwined events facilitated by chemical, electrical, and mechanical signals. The progress of each process is inherently linked to phenotypic changes in cells. Currently, the primary means of demonstrating morphological changes rely on measurements of neurite outgrowth and axon length. However, conventional techniques for monitoring these processes often require extensive preparation to enable manual or semi-automated measurements. Here, a label-free and non-invasive approach is employed for monitoring neuronal differentiation and degeneration using quantitative phase imaging (QPI). Operating on unlabeled specimens and offering little to no phototoxicity and photobleaching, QPI delivers quantitative maps of optical path length delays that provide an objective measure of cellular morphology and dynamics. This approach enables the visualization and quantification of axon length and other physical properties of dorsal root ganglion (DRG) neuronal cells, allowing greater understanding of neuronal responses to stimuli simulating CIPN conditions. This research paves new avenues for the development of more effective strategies in the clinical management of neurotoxicity.


Sujet(s)
Axones , Différenciation cellulaire , Ganglions sensitifs des nerfs spinaux , Animaux , Ganglions sensitifs des nerfs spinaux/anatomopathologie , Ganglions sensitifs des nerfs spinaux/cytologie , Axones/anatomopathologie , Neurones/anatomopathologie , Humains , Souris , Neuropathies périphériques/anatomopathologie , Neuropathies périphériques/induit chimiquement , Neuropathies périphériques/physiopathologie ,
9.
Sens Actuators B Chem ; 4042024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38524639

RÉSUMÉ

Recent advances in Raman spectroscopy have shown great potential for non-invasive analyte sensing, but the lack of a standardized optical phantom for these measurements has hindered further progress. While many research groups have developed optical phantoms that mimic bulk optical absorption and scattering, these materials typically have strong Raman scattering, making it difficult to distinguish metabolite signals. As a result, solid tissue phantoms for spectroscopy have been limited to highly scattering tissues such as bones and calcifications, and metabolite sensing has been primarily performed using liquid tissue phantoms. To address this issue, we have developed a layered skin-mimetic phantom that can support metabolite sensing through Raman spectroscopy. Our approach incorporates millifluidic vasculature that mimics blood vessels to allow for diffusion akin to metabolite diffusion in the skin. Furthermore, our skin phantoms are mechanically mimetic, providing an ideal model for development of minimally invasive optical techniques. By providing a standardized platform for measuring metabolites, our approach has the potential to facilitate critical developments in spectroscopic techniques and improve our understanding of metabolite dynamics in vivo.

10.
ACS Appl Mater Interfaces ; 16(9): 11159-11171, 2024 Mar 06.
Article de Anglais | MEDLINE | ID: mdl-38385360

RÉSUMÉ

For the improved delivery of cancer therapeutics and imaging agents, the conjugation of cell-penetrating peptides (CPPs) increases the cellular uptake and water solubility of agents. Among the various CPPs, arginine-rich peptides have been the most widely used. Combining CPPs with enzyme-responsive peptides presents an innovative strategy to target specific intracellular enzymes in cancer cells and when combined with the appropriate click chemistry can enhance theranostic drug delivery through the formation of intracellular self-assembled nanostructures. However, one drawback of CPPs is their high positive charge which can cause nonspecific binding, leading to off-target accumulation and potential toxicity. Hence, balancing cell-specific penetration, toxicity, and biocompatibility is essential for future clinical efficacy. We synthesized six cancer-specific, legumain-responsive RnAANCK peptides containing one to six arginine residues, with legumain being an asparaginyl endopeptidase that is overexpressed in aggressive prostate tumors. When conjugated to Alexa Fluor 488, R1-R6AANCK peptides exhibited a concentration- and time-dependent cell penetration in prostate cancer cells, which was higher for peptides with higher R values, reaching a plateau after approximately 120 min. Highly aggressive DU145 prostate tumor cells, but not less aggressive LNCaP cells, self-assembled nanoparticles in the cytosol after the cleavage of the legumain-specific peptide. The in vivo biocompatibility was assessed in mice after the intravenous injection of R1-R6AANCK peptides, with concentrations ranging from 0.0125 to 0.4 mmol/kg. The higher arginine content in R4-6 peptides showed blood and urine indicators for the impairment of bone marrow, liver, and kidney function in a dose-dependent manner, with instant hemolysis and morbidity in extreme cases. These findings underscore the importance of designing peptides with the optimal arginine residue length for a proper balance of cell-specific penetration, toxicity, and in vivo biocompatibility.


Sujet(s)
Peptides de pénétration cellulaire , Tumeurs , Animaux , Souris , Arginine/composition chimique , Peptides de pénétration cellulaire/composition chimique , Tumeurs/traitement médicamenteux
11.
Small ; 20(6): e2305110, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37752776

RÉSUMÉ

Functional disorders of the thyroid remain a global challenge and have profound impacts on human health. Serving as the barometer for thyroid function, thyroid-stimulating hormone (TSH) is considered the single most useful test of thyroid function. However, the prevailing TSH immunoassays rely on two types of antibodies in a sandwich format. The requirement of repeated incubation and washing further complicates the issue, making it unable to meet the requirements of the shifting public health landscape that demands rapid, sensitive, and low-cost TSH tests. Herein, a systematic study is performed to investigate the clinical translational potential of a single antibody-based biosensing platform for the TSH test. The biosensing platform leverages Raman spectral variations induced by the interaction between a TSH antigen and a Raman molecule-conjugated TSH antibody. In conjunction with machine learning, it allows TSH concentrations in various patient samples to be predicted with high accuracy and precision, which is robust against substrate-to-substrate, intra-substrate, and day-to-day variations. It is envisioned that the simplicity and generalizability of this single-antibody immunoassay coupled with the demonstrated performance in patient samples pave the way for it to be widely applied in clinical settings for low-cost detection of hormones, other molecular biomarkers, DNA, RNA, and pathogens.


Sujet(s)
Anticorps , Thyréostimuline , Humains , Dosage immunologique
12.
Adv Sci (Weinh) ; 10(35): e2304343, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37908150

RÉSUMÉ

Here, the authors report that co-crystallization of fluorophores with matrix-assisted laser desorption/ionization (MALDI) imaging matrices significantly enhances fluorophore brightness up to 79-fold, enabling the amplification of innate tissue autofluorescence. This discovery facilitates FluoMALDI, the imaging of the same biological sample by both fluorescence microscopy and MALDI imaging. The approach combines the high spatial resolution and specific labeling capabilities of fluorescence microscopy with the inherently multiplexed, versatile imaging capabilities of MALDI imaging. This new paradigm simplifies registration by avoiding physical changes between fluorescence and MALDI imaging, allowing to image the exact same cells in tissues with both modalities. Matrix-fluorophore co-crystallization also facilitates applications with insufficient fluorescence brightness. The authors demonstrate  feasibility of FluoMALDI imaging with endogenous and exogenous fluorophores and autofluorescence-based FluoMALDI of brain and kidney tissue sections. FluoMALDI will advance structural-functional microscopic imaging in cell biology, biomedicine, and pathology.


Sujet(s)
Encéphale , Rein , Spectrométrie de masse MALDI/méthodes , Cristallisation , Microscopie de fluorescence , Rein/imagerie diagnostique
13.
Adv Opt Mater ; 11(6)2023 Mar 17.
Article de Anglais | MEDLINE | ID: mdl-37920689

RÉSUMÉ

Nitrogen-vacancy (NV) centers in nanodiamond hold great promise for creating superior biological labels and quantum sensing methods. Yet, inefficient photon generation and extraction from excited NV centers restricts the achievable sensitivity and temporal resolution. Herein, we report an entirely complementary route featuring pyramidal hyperbolic metasurface to modify the spontaneous emission of NV centers. Fabricated using nanosphere lithography, the metasurface consists of alternatively stacked silica-silver thin films configured in a pyramidal fashion, and supports both spectrally broadband Purcell enhancement and spatially extended intense local fields owing to the hyperbolic dispersion and plasmonic coupling. The enhanced photophysical properties are manifested as a simultaneous amplification to the spontaneous decay rate and emission intensity of NV centers. We envision the reported pyramidal metasurface could serve as a versatile platform for creating chip-based ultrafast single-photon sources and spin-enhanced quantum biosensing strategies, as well as aiding in further fundamental understanding of photoexcited species in condensed phases.

14.
Sci Rep ; 13(1): 18566, 2023 10 30.
Article de Anglais | MEDLINE | ID: mdl-37903851

RÉSUMÉ

The premetastatic niche hypothesis proposes an active priming of the metastatic site by factors secreted from the primary tumor prior to the arrival of the first cancer cells. We investigated several extracellular matrix (ECM) structural proteins, ECM degrading enzymes, and ECM processing proteins involved in the ECM remodeling of the premetastatic niche. Our in vitro model consisted of lung fibroblasts, which were exposed to factors secreted by nonmalignant breast epithelial cells, nonmetastatic breast cancer cells, or metastatic breast cancer cells. We assessed ECM remodeling in vivo in premetastatic lungs of female mice growing orthotopic primary breast tumor xenografts, as compared to lungs of control mice without tumors. Premetastatic lungs contained significantly upregulated Collagen (Col) Col4A5, matrix metalloproteinases (MMPs) MMP9 and MMP14, and decreased levels of MMP13 and lysyl oxidase (LOX) as compared to control lungs. These in vivo findings were consistent with several of our in vitro cell culture findings, which showed elevated Col14A1, Col4A5, glypican-1 (GPC1) and decreased Col5A1 and Col15A1 for ECM structural proteins, increased MMP2, MMP3, and MMP14 for ECM degrading enzymes, and decreased LOX, LOXL2, and prolyl 4-hydroxylase alpha-1 (P4HA1) for ECM processing proteins in lung fibroblasts conditioned with metastatic breast cancer cell media as compared to control. Taken together, our data show that premetastatic priming of lungs by primary breast tumors resulted in significant ECM remodeling which could facilitate metastasis by increasing interstitial fibrillar collagens and ECM stiffness (Col14A1), disruptions of basement membranes (Col4A5), and formation of leaky blood vessels (MMP2, MMP3, MMP9, and MMP14) to promote metastasis.


Sujet(s)
Tumeurs du sein , Tumeurs mammaires de l'animal , Humains , Femelle , Souris , Animaux , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 9/métabolisme , Matrix metalloproteinase 14/métabolisme , Matrix metalloproteinase 3/métabolisme , Poumon/anatomopathologie , Matrice extracellulaire/métabolisme , Tumeurs mammaires de l'animal/métabolisme , Protéines de la matrice extracellulaire/métabolisme , Tumeurs du sein/anatomopathologie
15.
Nano Lett ; 23(20): 9529-9537, 2023 Oct 25.
Article de Anglais | MEDLINE | ID: mdl-37819891

RÉSUMÉ

While fundamental to quantum sensing, quantum state control has been traditionally limited to extreme conditions. This restricts the impact of the practical implementation of quantum sensing on a broad range of physical measurements. Plexcitons, however, provide a promising path under ambient conditions toward quantum state control and thus quantum sensing, owing to their origin from strong plasmon-exciton coupling. Herein, we harness plexcitons to demonstrate quantum plexcitonic sensing by strongly coupling excitonic particles to a plasmonic hyperbolic metasurface. As compared to classical sensing in the weak-coupling regime, our model of quantum plexcitonic sensing performs at a level that is ∼40 times more sensitive. Noise-modulated sensitivity studies reinforce the quantum advantage over classical sensing, featuring better sensitivity, smaller sensitivity uncertainty, and higher resilience against optical noise. The successful demonstration of quantum plexcitonic sensing opens the door for a variety of physical, chemical, and biological measurements by leveraging strongly coupled plasmon-exciton systems.

16.
J Histochem Cytochem ; 71(11): 643-652, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37833851

RÉSUMÉ

In the clinical setting, routine identification of the main types of tissue amyloid deposits, light-chain amyloid (AL) and serum amyloid A (AA), is based on histochemical staining; rarer types of amyloid require mass spectrometry analysis. Raman spectroscopic imaging is an analytical tool, which can be used to chemically map, and thus characterize, the molecular composition of fluid and solid tissue. In this proof-of-concept study, we tested the feasibility of applying Raman spectroscopy combined with artificial intelligence to detect and characterize amyloid deposits in unstained frozen tissue sections from kidney biopsies with pathologic diagnosis of AL and AA amyloidosis and control biopsies with no amyloidosis (NA). Raman hyperspectral images, mapped in a 2D grid-like fashion over the tissue sections, were obtained. Three machine learning-assisted analysis models of the hyperspectral images could accurately distinguish AL (types λ and κ), AA, and NA 93-100% of the time. Although very preliminary, these findings illustrate the potential of Raman spectroscopy as a technique to identify, and possibly, subtype renal amyloidosis.


Sujet(s)
Amyloïdose , Plaque amyloïde , Humains , Plaque amyloïde/anatomopathologie , Intelligence artificielle , Amyloïde/analyse , Amyloïdose/imagerie diagnostique , Amyloïdose/anatomopathologie , Rein/anatomopathologie
17.
Sci Adv ; 9(37): eadg6231, 2023 09 15.
Article de Anglais | MEDLINE | ID: mdl-37703362

RÉSUMÉ

Anticancer therapy screening in vitro identifies additional treatments and improves clinical outcomes. Systematically, although most tested cells respond to cues with apoptosis, an appreciable portion enters a senescent state, a critical condition potentially driving tumor resistance and relapse. Conventional screening protocols would strongly benefit from prompt identification and monitoring of therapy-induced senescent (TIS) cells in their native form. We combined complementary all-optical, label-free, and quantitative microscopy techniques, based on coherent Raman scattering, multiphoton absorption, and interferometry, to explore the early onset and progression of this phenotype, which has been understudied in unperturbed conditions. We identified TIS manifestations as early as 24 hours following treatment, consisting of substantial mitochondrial rearrangement and increase of volume and dry mass, followed by accumulation of lipid vesicles starting at 72 hours. This work holds the potential to affect anticancer treatment research, by offering a label-free, rapid, and accurate method to identify initial TIS in tumor cells.


Sujet(s)
Tumeurs , Humains , Prévention secondaire , Apoptose , Signaux , Imagerie moléculaire
18.
Adv Sci (Weinh) ; 10(34): e2304164, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37715297

RÉSUMÉ

Inspired by the principle of in situ self-assembly, the development of enzyme-activated molecular nanoprobes can have a profound impact on targeted tumor detection. However, despite their intrinsic promise, obtaining an optical readout of enzyme activity with high specificity in native milieu has proven to be challenging. Here, a fundamentally new class of Raman-active self-assembling bioorthogonal enzyme recognition (nanoSABER) probes for targeted tumor imaging is reported. This class of Raman probes presents narrow spectral bands reflecting their vibrational fingerprints and offers an attractive solution for optical imaging at different bio-organization levels. The optical beacon harnesses an enzyme-responsive peptide sequence, unique tumor-penetrating properties, and vibrational tags with stretching frequencies in the cell-silent Raman window. The design of nanoSABER is tailored and engineered to transform into a supramolecular structure exhibiting distinct vibrational signatures in presence of target enzyme, creating a direct causality between enzyme activity and Raman signal. Through the integration of substrate-specific for tumor-associated enzyme legumain, unique capabilities of nanoSABER for imaging enzyme activity at molecular, cellular, and tissue levels in combination with machine learning models are shown. These results demonstrate that the nanoSABER probe may serve as a versatile platform for Raman-based recognition of tumor aggressiveness, drug accumulation, and therapeutic response.


Sujet(s)
Tumeurs , Humains , Tumeurs/imagerie diagnostique , Imagerie optique
19.
Biosens Bioelectron ; 239: 115597, 2023 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-37597501

RÉSUMÉ

Multimodal tissue imaging techniques that integrate two complementary modalities are powerful discovery tools for unraveling biological processes and identifying biomarkers of disease. Combining Raman spectroscopic imaging (RSI) and matrix-assisted laser-desorption/ionization (MALDI) mass spectrometry imaging (MSI) to obtain fused images with the advantages of both modalities has the potential of providing spatially resolved, sensitive, specific biomolecular information, but has so far involved two separate sample preparations, or even consecutive tissue sections for RSI and MALDI MSI, resulting in images with inherent disparities. We have developed RaMALDI, a streamlined, integrated, multimodal imaging workflow of RSI and MALDI MSI, performed on a single tissue section with one sample preparation protocol. We show that RaMALDI imaging of various tissues effectively integrates molecular information acquired from both RSI and MALDI MSI of the same sample, which will drive discoveries in cell biology, biomedicine, and pathology, and advance tissue diagnostics.


Sujet(s)
Techniques de biocapteur , Spectrométrie de masse MALDI , Imagerie multimodale , Sérogroupe , Manipulation d'échantillons
20.
bioRxiv ; 2023 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-37546926

RÉSUMÉ

Quantitative phase imaging (QPI) has rapidly emerged as a complementary tool to fluorescence imaging, as it provides an objective measure of cell morphology and dynamics, free of variability due to contrast agents. In particular, three-dimensional (3D) tomographic imaging of live cells has opened up new directions of investigation by providing systematic and correlative analysis of various cellular parameters without limitations of photobleaching and phototoxicity. While current QPI systems allow the rapid acquisition of tomographic images, the pipeline to analyze these raw 3D tomograms is not well-developed. This work focuses on a critical, yet often underappreciated, step of the analysis pipeline, that of 3D cell segmentation from the acquired tomograms. The current method employed for such tasks is the Otsu-based 3D watershed algorithm, which works well for isolated cells; however, it is very challenging to draw boundaries when the cells are clumped. This process is also memory intensive since the processing requires computation on a 3D stack of images. We report the CellSNAP (Cell Segmentation via Novel Algorithm for Phase Imaging) algorithm for the segmentation of QPI images, which outstrips the current gold standard in terms of speed, robustness, and implementation, achieving cell segmentation under 2 seconds per cell on a single-core processor. The implementation of CellSNAP can easily be parallelized on a multi-core system for further speed improvements. For the cases where segmentation is possible with the existing standard method, our algorithm displays an average difference of 5% for dry mass and 8% for volume measurements. We also show that CellSNAP can handle challenging image datasets where cells are clumped and marred by interferogram drifts, which pose major difficulties for all QPI-focused segmentation tools. We envision our work will lead to the broader adoption of QPI imaging for high-throughput analysis, which has, in part, been stymied by a lack of suitable image segmentation tools.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE