Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Theranostics ; 14(6): 2526-2543, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38646640

RESUMEN

Rationale: A mature tissue resident macrophage (TRM) population residing in the peritoneal cavity has been known for its unique ability to migrate to peritoneally located injured tissues and impart wound healing properties. Here, we sought to expand on this unique ability of large peritoneal macrophages (LPMs) by investigating whether these GATA6+ LPMs could also intravasate into systemic circulation and migrate to extra-peritoneally located lungs upon ablating lung-resident alveolar macrophages (AMs) by intranasally administered clodronate liposomes in mice. Methods: C12-200 cationic lipidoid-based nanoparticles were employed to selectively deliver a small interfering RNA (siRNA)-targeting CD-45 labeled with a cyanine 5.5 (Cy5.5) dye to LPMs in vivo via intraperitoneal injection. We utilized a non-invasive optical technique called Diffuse In Vivo Flow Cytometry (DiFC) to then systemically track these LPMs in real time and paired it with more conventional techniques like flow cytometry and immunocytochemistry to initially confirm uptake of C12-200 encapsulated siRNA-Cy5.5 (siRNA-Cy5.5 (C12-200)) into LPMs, and further track them from the peritoneal cavity to the lungs in a mouse model of AM depletion incited by intranasally administered clodronate liposomes. Also, we stained for LPM-specific marker zinc-finger transcription factor GATA6 in harvested cells from biofluids like broncho-alveolar lavage as well as whole blood to probe for Cy5.5-labeled LPMs in the lungs as well as in systemic circulation. Results: siRNA-Cy5.5 (C12-200) was robustly taken up by LPMs. Upon depletion of lung-resident AMs, these siRNA-Cy5.5 (C12-200) labeled LPMs rapidly migrated to the lungs via systemic circulation within 12-24 h. DiFC results showed that these LPMs intravasated from the peritoneal cavity and utilized a systemic route of migration. Moreover, immunocytochemical staining of zinc-finger transcription factor GATA6 further confirmed results from DiFC and flow cytometry, confirming the presence of siRNA-Cy5.5 (C12-200)-labeled LPMs in the peritoneum, whole blood and BALF only upon clodronate-administration. Conclusion: Our results indicate for the very first time that selective tropism, migration, and infiltration of LPMs into extra-peritoneally located lungs was dependent on clodronate-mediated AM depletion. These results further open the possibility of therapeutically utilizing LPMs as delivery vehicles to carry nanoparticle-encapsulated oligonucleotide modalities to potentially address inflammatory diseases, infectious diseases and even cancer.


Asunto(s)
Ácido Clodrónico , Pulmón , Macrófagos Peritoneales , Nanopartículas , Animales , Ácido Clodrónico/farmacología , Ácido Clodrónico/administración & dosificación , Nanopartículas/química , Nanopartículas/administración & dosificación , Ratones , Pulmón/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Alveolares/metabolismo , ARN Interferente Pequeño/administración & dosificación , Factor de Transcripción GATA6/metabolismo , Liposomas , Ratones Endogámicos C57BL , Carbocianinas/química , Movimiento Celular/efectos de los fármacos , Citometría de Flujo
2.
Biomed Opt Express ; 14(11): 5555-5568, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-38021147

RESUMEN

In this work, we introduce ratiometric diffuse in vivo flow cytometry (R-DiFC) for quantitative measurement of circulating fluorescent red blood cell (fRBC) sensors for systemic blood sodium levels. Unlike in our previous work in measuring circulating fRBC sensors, R-DiFC allows simultaneous measurement of two fluorophores encapsulated in the sensor, the ratio of which enables self-calibration of the fluorescence signal with different fRBC depths in biological tissue. We show that the R-DiFC signal varies significantly less than either fluorescence signal alone. This work holds promise for personalized monitoring of systemic sodium for bipolar patients in the future.

3.
J Biomed Opt ; 28(7): 077001, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37484977

RESUMEN

Significance: Diffuse in vivo flow cytometry (DiFC) is an emerging fluorescence sensing method to non-invasively detect labeled circulating cells in vivo. However, due to signal-to-noise ratio (SNR) constraints largely attributed to background tissue autofluorescence (AF), DiFC's measurement depth is limited. Aim: The dual ratio (DR)/dual slope is an optical measurement method that aims to suppress noise and enhance SNR to deep tissue regions. We aim to investigate the combination of DR and near-infrared (NIR) DiFC to improve circulating cells' maximum detectable depth and SNR. Approach: Phantom experiments were used to estimate the key parameters in a diffuse fluorescence excitation and emission model. This model and parameters were implemented in Monte Carlo to simulate DR DiFC while varying noise and AF parameters to identify the advantages and limitations of the proposed technique. Results: Two key factors must be true to give DR DiFC an advantage over traditional DiFC: first, the fraction of noise that DR methods cannot cancel cannot be above the order of 10% for acceptable SNR. Second, DR DiFC has an advantage, in terms of SNR, if the distribution of tissue AF contributors is surface-weighted. Conclusions: DR cancelable noise may be designed (e.g., through the use of source multiplexing), and indications point to the AF contributors' distribution being truly surface-weighted in vivo. Successful and worthwhile implementation of DR DiFC depends on these considerations, but results point to DR DiFC having possible advantages over traditional DiFC.


Asunto(s)
Fantasmas de Imagen , Relación Señal-Ruido
4.
ArXiv ; 2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37292468

RESUMEN

SIGNIFICANCE: Diffuse in-vivo Flow Cytometry (DiFC) is an emerging fluorescence sensing method to non-invasively detect labeled circulating cells in-vivo. However, due to Signal-to-Noise Ratio (SNR) constraints largely attributed to background tissue autofluorescence, DiFC's measurement depth is limited. multiplies Aim: The Dual-Ratio (DR) / dual-slope is a new optical measurement method that aims to suppress noise and enhance SNR to deep tissue regions. We aim to investigate the combination of DR and Near-InfraRed (NIR) DiFC to improve circulating cells' maximum detectable depth and SNR. APPROACH: Phantom experiments were used to estimate the key parameters in a diffuse fluorescence excitation and emission model. This model and parameters were implemented in Monte-Carlo to simulate DR DiFC while varying noise and autofluorescence parameters to identify the advantages and limitations of the proposed technique. RESULTS: Two key factors must be true to give DR DiFC an advantage over traditional DiFC; first, the fraction of noise that DR methods cannot cancel cannot be above the order of 10% for acceptable SNR. Second, DR DiFC has an advantage, in terms of SNR, if the distribution of tissue autofluorescence contributors is surface-weighted. CONCLUSIONS: DR cancelable noise may be designed for (e.g. through the use of source multiplexing), and indications point to the autofluorescence contributors' distribution being truly surface-weighted in-vivo. Successful and worthwhile implementation of DR DiFC depends on these considerations, but results point to DR DiFC having possible advantages over traditional DiFC.

5.
J Biomed Opt ; 27(9)2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36114606

RESUMEN

Significance: Diffuse in vivo flow cytometry (DiFC) is an emerging technique for enumerating rare fluorescently labeled circulating cells noninvasively in the bloodstream. Thus far, we have reported red and blue-green versions of DiFC. Use of near-infrared (NIR) fluorescent light would in principle allow use of DiFC in deeper tissues and would be compatible with emerging NIR fluorescence molecular contrast agents. Aim: We describe the design of a NIR-DiFC instrument and demonstrate its use in optical flow phantoms in vitro and in mice in vivo. Approach: We developed an improved optical fiber probe design for efficient collection of fluorescence from individual circulating cells and efficient rejection of instrument autofluorescence. We built a NIR-DiFC instrument. We tested this with NIR fluorescent microspheres and cell lines labeled with OTL38 fluorescence contrast agent in a flow phantom model. We also tested NIR-DiFC in nude mice injected intravenously with OTL38-labeled L1210A cells. Results: NIR-DiFC allowed detection of circulating tumor cells (CTCs) in flow phantoms with mean signal-to-noise ratios (SNRs) of 19 to 32 dB. In mice, fluorescently labeled CTCs were detectable with mean SNR of 26 dB. NIR-DiFC also exhibited orders significantly lower autofluorescence and false-alarm rates than blue-green DiFC. Conclusions: NIR-DiFC allows use of emerging NIR contrast agents. Our work could pave the way for future use of NIR-DiFC in humans.


Asunto(s)
Medios de Contraste , Células Neoplásicas Circulantes , Animales , Recuento de Células , Citometría de Flujo/métodos , Colorantes Fluorescentes , Humanos , Ratones , Ratones Desnudos , Células Neoplásicas Circulantes/patología
6.
J Biomed Opt ; 27(6)2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35726129

RESUMEN

SIGNIFICANCE: "Diffuse in vivo flow cytometry" (DiFC) is an emerging technology for fluorescence detection of rare circulating cells directly in large deep-seated blood vessels in mice. Because DiFC uses highly scattered light, in principle, it could be translated to human use. However, an open question is whether fluorescent signals from single cells would be detectable in human-scale anatomies. AIM: Suitable blood vessels in a human wrist or forearm are at a depth of ∼2 to 4 mm. The aim of this work was to study the impact of DiFC instrument geometry and wavelength on the detected DiFC signal and on the maximum depth of detection of a moving cell. APPROACH: We used Monte Carlo simulations to compute fluorescence Jacobian (sensitivity) matrices for a range of source and detector separations (SDS) and tissue optical properties over the visible and near infrared spectrum. We performed experimental measurements with three available versions of DiFC (488, 640, and 780 nm), fluorescent microspheres, and tissue mimicking optical flow phantoms. We used both computational and experimental data to estimate the maximum depth of detection at each combination of settings. RESULTS: For the DiFC detection problem, our analysis showed that for deep-seated blood vessels, the maximum sensitivity was obtained with NIR light (780 nm) and 3-mm SDS. CONCLUSIONS: These results suggest that-in combination with a suitable molecularly targeted fluorescent probes-circulating cells and nanosensors could, in principle, be detectable in circulation in humans.


Asunto(s)
Colorantes Fluorescentes , Animales , Citometría de Flujo/métodos , Humanos , Ratones , Microesferas , Método de Montecarlo , Fantasmas de Imagen
8.
Front Oncol ; 10: 601085, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240820

RESUMEN

MOTIVATION: Circulating tumor cells (CTCs) are widely studied using liquid biopsy methods that analyze fractionally-small peripheral blood (PB) samples. However, little is known about natural fluctuations in CTC numbers that may occur over short timescales in vivo, and how these may affect detection and enumeration of rare CTCs from small blood samples. METHODS: We recently developed an optical instrument called "diffuse in vivo flow cytometry" (DiFC) that uniquely allows continuous, non-invasive counting of rare, green fluorescent protein expressing CTCs in large blood vessels in mice. Here, we used DiFC to study short-term changes in CTC numbers in multiple myeloma and Lewis lung carcinoma xenograft models. We analyzed CTC detections in over 100 h of DiFC data, and considered intervals corresponding to approximately 1%, 5%, 10%, and 20% of the PB volume. In addition, we analyzed changes in CTC numbers over 24 h (diurnal) periods. RESULTS: For rare CTCs (fewer than 1 CTC per ml of blood), the use of short DiFC intervals (corresponding to small PB samples) frequently resulted in no detections. For more abundant CTCs, CTC numbers frequently varied by an order of magnitude or more over the time-scales considered. This variance in CTC detections far exceeded that expected by Poisson statistics or by instrument variability. Rather, the data were consistent with significant changes in mean numbers of CTCs on the timescales of minutes and hours. CONCLUSIONS: The observed temporal changes can be explained by known properties of CTCs, namely, the continuous shedding of CTCs from tumors and the short half-life of CTCs in blood. It follows that the number of cells in a blood sample are strongly impacted by the timing of the draw. The issue is likely to be compounded for multicellular CTC clusters or specific CTC subtypes, which are even more rare than single CTCs. However, we show that enumeration can in principle be improved by averaging multiple samples, analysis of larger volumes, or development of methods for enumeration of CTCs directly in vivo.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...