Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33906946

RESUMO

Intracarotid arterial hyperosmolar mannitol (ICAHM) blood-brain barrier disruption (BBBD) is effective and safe for delivery of therapeutics for central nervous system malignancies. ICAHM osmotically alters endothelial cells and tight junction integrity to achieve BBBD. However, occurrence of neuroinflammation following hemispheric BBBD by ICAHM remains unknown. Temporal proteomic changes in rat brains following ICAHM included increased damage-associated molecular patterns, cytokines, chemokines, trophic factors, and cell adhesion molecules, indicative of a sterile inflammatory response (SIR). Proteomic changes occurred within 5 min of ICAHM infusion and returned to baseline by 96 h. Transcriptomic analyses following ICAHM BBBD further supported an SIR. Immunohistochemistry revealed activated astrocytes, microglia, and macrophages. Moreover, proinflammatory proteins were elevated in serum, and proteomic and histological findings from the contralateral hemisphere demonstrated a less pronounced SIR, suggesting neuroinflammation beyond regions of ICAHM infusion. Collectively, these results demonstrate ICAHM induces a transient SIR that could potentially be harnessed for neuroimmunomodulation.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Imunidade Inata/genética , Inflamação/genética , Manitol/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Artérias Carótidas/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/sangue , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Neoplasias do Sistema Nervoso Central/genética , Quimiocinas/sangue , Citocinas/sangue , Células Endoteliais/efeitos dos fármacos , Humanos , Inflamação/sangue , Ratos , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/genética
2.
J Cell Mol Med ; 24(22): 13278-13288, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33067927

RESUMO

Image-guided pulsed focused ultrasound (pFUS) is a non-invasive technique that can increase tropism of intravenously (IV)-infused mesenchymal stromal cells (MSC) to sonicated tissues. MSC have shown promise for cardiac regenerative medicine strategies but can be hampered by inefficient homing to the myocardium. This study sonicated the left ventricles (LV) in rats with magnetic resonance imaging (MRI)-guided pFUS and examined both proteomic responses and subsequent MSC tropism to treated myocardium. T2-weighted MRI was used for pFUS targeting of the entire LV. pFUS increased numerous pro- and anti-inflammatory cytokines, chemokines, and trophic factors and cell adhesion molecules in the myocardial microenvironment for up to 48 hours post-sonication. Cardiac troponin I and N-terminal pro-B-type natriuretic peptide were elevated in the serum and myocardium. Immunohistochemistry revealed transient hypoxia and immune cell infiltration in pFUS-targeted regions. Myocardial tropism of IV-infused human MSC following pFUS increased twofold-threefold compared with controls. Proteomic and histological changes in myocardium following pFUS suggested a reversible inflammatory and hypoxic response leading to increased tropism of MSC. MR-guided pFUS could represent a non-invasive modality to improve MSC therapies for cardiac regenerative medicine approaches.


Assuntos
Coração/diagnóstico por imagem , Imageamento por Ressonância Magnética , Células-Tronco Mesenquimais/citologia , Miocárdio/metabolismo , Ultrassonografia/métodos , Animais , Citocinas/metabolismo , Feminino , Ventrículos do Coração/metabolismo , Humanos , Hipóxia , Imuno-Histoquímica , Inflamação , Transplante de Células-Tronco Mesenquimais , Permeabilidade , Proteômica , Ratos , Ratos Sprague-Dawley
3.
Proc Natl Acad Sci U S A ; 114(1): E75-E84, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-27994152

RESUMO

MRI-guided pulsed focused ultrasound (pFUS) combined with systemic infusion of ultrasound contrast agent microbubbles (MB) causes localized blood-brain barrier (BBB) disruption that is currently being advocated for increasing drug or gene delivery in neurological diseases. The mechanical acoustic cavitation effects of opening the BBB by low-intensity pFUS+MB, as evidenced by contrast-enhanced MRI, resulted in an immediate damage-associated molecular pattern (DAMP) response including elevations in heat-shock protein 70, IL-1, IL-18, and TNFα indicative of a sterile inflammatory response (SIR) in the parenchyma. Concurrent with DAMP presentation, significant elevations in proinflammatory, antiinflammatory, and trophic factors along with neurotrophic and neurogenesis factors were detected; these elevations lasted 24 h. Transcriptomic analysis of sonicated brain supported the proteomic findings and indicated that the SIR was facilitated through the induction of the NFκB pathway. Histological evaluation demonstrated increased albumin in the parenchyma that cleared by 24 h along with TUNEL+ neurons, activated astrocytes, microglia, and increased cell adhesion molecules in the vasculature. Infusion of fluorescent beads 3 d before pFUS+MB revealed the infiltration of CD68+ macrophages at 6 d postsonication, as is consistent with an innate immune response. pFUS+MB is being considered as part of a noninvasive adjuvant treatment for malignancy or neurodegenerative diseases. These results demonstrate that pFUS+MB induces an SIR compatible with ischemia or mild traumatic brain injury. Further investigation will be required before this approach can be widely implemented in clinical trials.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/fisiopatologia , Sistemas de Liberação de Medicamentos/métodos , Técnicas de Transferência de Genes , Sonicação/métodos , Ultrassonografia/métodos , Animais , Astrócitos/metabolismo , Moléculas de Adesão Celular/metabolismo , Feminino , Proteínas de Choque Térmico HSP70/metabolismo , Inflamação/patologia , Interleucina-1/metabolismo , Interleucina-18/metabolismo , Macrófagos/imunologia , Microglia/metabolismo , Doenças Neurodegenerativas/terapia , Tecido Parenquimatoso/patologia , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
4.
J Cell Mol Med ; 22(12): 6015-6025, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30216653

RESUMO

Mesenchymal stromal cell (MSC) therapies combined with renal pulsed focused ultrasound (pFUS) pretreatment increase MSC homing and improve cisplatin-induced acute kidney injury (AKI) better than MSC alone. However, mechanisms underlying improved outcomes remain unknown. We hypothesize pFUS up-regulates renal interferon-γ (IFNγ) and stimulates MSC to produce interleukin-10 (IL-10) after migrating to kidneys. To demonstrate initially, MSC cultured with IFNγ up-regulated IL-10. More MSC-derived IL-10 was detected in kidneys when IFNγ-stimulated MSC were infused and they improved AKI better than unstimulated MSC. Next, IFNγ-knockout mice with AKI received pFUS+MSC, but MSC-derived IL-10 expression and AKI were similar to using MSC alone. AKI in wild-type mice receiving pFUS and IL-10-deficient MSC was also unimproved compared to administering IL-10-deficient MSC alone. Indoleamine 2,3-dioxygenase (IDO), an anti-inflammatory enzyme up-regulated in MSC by IFNγ, was up-regulated during AKI, but was not further elevated in MSC from pFUS-treated kidneys, suggesting that IDO is not involved in improved AKI healing by pFUS+MSC. These data suggest IFNγ is up-regulated by pFUS and after i.v.-infused MSC home to pFUS-treated kidneys, IFNγ stimulates additional IL-10 production by MSC to improve AKI. Analogous mechanisms of ultrasound-treated tissue microenvironments stimulating therapeutic MSC may exist in other pathologies where adjuvant ultrasound techniques are successful.


Assuntos
Injúria Renal Aguda/terapia , Interferon gama/genética , Interleucina-10/genética , Transplante de Células-Tronco Mesenquimais , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Animais , Cisplatino/efeitos adversos , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Humanos , Rim/lesões , Rim/metabolismo , Rim/patologia , Células-Tronco Mesenquimais , Camundongos , Camundongos Knockout , Ondas Ultrassônicas
5.
J Transl Med ; 15(1): 252, 2017 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-29237455

RESUMO

BACKGROUND: Image-guided high intensity focused ultrasound has been used as an extracorporeal cardiac pacing tool and to enhance homing of stem cells to targeted tissues. However, molecular changes in the myocardium after sonication have not been widely investigated. Magnetic-resonance (MR)-guided pulsed focused ultrasound (pFUS) was targeted to the rat myocardium over a range of pressures and the microenvironmental and histological effects were evaluated over time. METHODS: Eight-to-ten-week-old Sprague-Dawley rats received T2-weighted MR images to target pFUS to the left ventricular and septum without cardiac or respiratory gating. Rats were sonicated through the thoracic wall at peak negative pressures (PNP) from 1 to 8 MPa at a center frequency of 1 MHz, 10 ms pulse duration and 1 Hz pulse repetition frequency for 100 pulses per focal target. Following pFUS, myocardium was harvested over 24 h and subjected to imaging, proteomic, and histological measurements. RESULTS: pFUS to the myocardium increased expression of cytokines, chemokines, and trophic factors characterized by an initial increase in tumor necrosis factor (TNF)-α followed by increases in pro- and anti-inflammatory factors that returned to baseline by 24 h. Immediately after pFUS, there was a transient (< 1 h) increase in N-terminal pro b-type natriuretic peptide (NT-proBNP) without elevation of other cardiac injury markers. A relationship between PNP and expression of TNF-α and NT-proBNP was observed with significant changes (p < 0.05 ANOVA) ≥ 4 MPa compared to untreated controls. Contrast-enhanced ex vivo T1-weighted MRI revealed vascular leakage in sonicated myocardium that was accompanied by the presence of albumin upon immunohistochemistry. Histology revealed infiltration of neutrophils and macrophages without morphological myofibril changes in sonicated tissue accompanied by pulmonary hemorrhage at PNP > 4 MPa. CONCLUSIONS: MR-guided pFUS to myocardium induced transient proteomic and histological changes. The temporal proteomic changes in the myocardium indicate a short-lived sterile inflammatory response consistent with ischemia or contusion. Further study of myocardial function and strain is needed to determine if pFUS could be developed as an experimental model of cardiac injury and chest trauma.


Assuntos
Coração/diagnóstico por imagem , Ablação por Ultrassom Focalizado de Alta Intensidade , Imageamento por Ressonância Magnética , Miocárdio/metabolismo , Miocárdio/patologia , Animais , Biomarcadores/metabolismo , Feminino , Granulócitos/metabolismo , Macrófagos/metabolismo , Proteômica , Ratos Sprague-Dawley
6.
Stem Cells ; 33(4): 1173-86, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25534849

RESUMO

Maximal homing of infused stem cells to diseased tissue is critical for regenerative medicine. Pulsed focused ultrasound (pFUS) is a clinically relevant platform to direct stem cell migration. Through mechanotransduction, pFUS establishes local gradients of cytokines, chemokines, trophic factors (CCTF) and cell adhesion molecules (CAM) in treated skeletal muscle that subsequently infused mesenchymal stromal cells (MSC) can capitalize to migrate into the parenchyma. Characterizing molecular responses to mechanical pFUS effects revealed tumor necrosis factor-alpha (TNFα) drives cyclooxygenase-2 (COX2) signaling to locally increase CCTF/CAM that are necessary for MSC homing. pFUS failed to increase chemoattractants and induce MSC homing to treated muscle in mice pretreated with ibuprofen (nonspecific COX inhibitor) or etanercept (TNFα inhibitor). pFUS-induced MSC homing was also suppressed in COX2-knockout mice, demonstrating ibuprofen blocked the mechanically induced CCTF/CAM by acting on COX2. Anti-inflammatory drugs, including ibuprofen, are administered to muscular dystrophy (MD) patients, and ibuprofen also suppressed pFUS-induced homing to muscle in a mouse model of MD. Drug interactions with cell therapies remain unexplored and are not controlled for during clinical cell therapy trials. This study highlights potentially negative drug-host interactions that suppress stem cell homing and could undermine cell-based approaches for regenerative medicine.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Mecanotransdução Celular/fisiologia , Células-Tronco Mesenquimais/fisiologia , Distrofias Musculares , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Ondas Ultrassônicas , Animais , Células Cultivadas , Feminino , Humanos , Mecanotransdução Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos da radiação , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C3H , Camundongos Knockout , Distrofias Musculares/patologia , Fator de Necrose Tumoral alfa/biossíntese , Adulto Jovem
7.
Stem Cells ; 33(4): 1241-53, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25640064

RESUMO

Animal studies have shown that mesenchymal stromal cell (MSC) infusions improve acute kidney injury (AKI) outcomes when administered early after ischemic/reperfusion injury or within 24 hours after cisplatin administration. These findings have spurred several human clinical trials to prevent AKI. However, no specific therapy effectively treats clinically obvious AKI or rescues renal function once advanced injury is established. We investigated if noninvasive image-guided pulsed focused ultrasound (pFUS) could alter the kidney microenvironment to enhance homing of subsequently infused MSC. To examine the efficacy of pFUS-enhanced cell homing in disease, we targeted pFUS to kidneys to enhance MSC homing after cisplatin-induced AKI. We found that pFUS enhanced MSC homing at 1 day post-cisplatin, prior to renal functional deficits, and that enhanced homing improved outcomes of renal function, tubular cell death, and regeneration at 5 days post-cisplatin compared to MSC alone. We then investigated whether pFUS+MSC therapy could rescue established AKI. MSC alone at 3 days post-cisplatin, after renal functional deficits were obvious, significantly improved 7-day survival of animals. Survival was further improved by pFUS and MSC. pFUS prior to MSC injections increased IL-10 production by MSC that homed to kidneys and generated an anti-inflammatory immune cell profile in treated kidneys. This study shows pFUS is a neoadjuvant approach to improve MSC homing to diseased organs. pFUS with MSC better prevents AKI than MSC alone and allows rescue therapy in established AKI, which currently has no meaningful therapeutic options.


Assuntos
Injúria Renal Aguda/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Ondas Ultrassônicas , Injúria Renal Aguda/patologia , Animais , Feminino , Humanos , Células-Tronco Mesenquimais/efeitos da radiação , Camundongos , Camundongos Endogâmicos C3H , Resultado do Tratamento
9.
Stem Cells ; 31(11): 2551-60, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23922277

RESUMO

Stem cells are promising therapeutics for cardiovascular diseases, and i.v. injection is the most desirable route of administration clinically. Subsequent homing of exogenous stem cells to pathological loci is frequently required for therapeutic efficacy and is mediated by chemoattractants (cell adhesion molecules, cytokines, and growth factors). Homing processes are inefficient and depend on short-lived pathological inflammation that limits the window of opportunity for cell injections. Noninvasive pulsed focused ultrasound (pFUS), which emphasizes mechanical ultrasound-tissue interactions, can be precisely targeted in the body and is a promising approach to target and maximize stem cell delivery by stimulating chemoattractant expression in pFUS-treated tissue prior to cell infusions. We demonstrate that pFUS is nondestructive to murine skeletal muscle tissue (no necrosis, hemorrhage, or muscle stem cell activation) and initiates a largely M2-type macrophage response. We also demonstrate that local upregulation of chemoattractants in pFUS-treated skeletal muscle leads to enhance homing, permeability, and retention of human mesenchymal stem cells (MSC) and human endothelial precursor cells (EPC). Furthermore, the magnitude of MSC or EPC homing was increased when pFUS treatments and cell infusions were repeated daily. This study demonstrates that pFUS defines transient "molecular zip codes" of elevated chemoattractants in targeted muscle tissue, which effectively provides spatiotemporal control and tunability of the homing process for multiple stem cell types. pFUS is a clinically translatable modality that may ultimately improve homing efficiency and flexibility of cell therapies for cardiovascular diseases.


Assuntos
Citocinas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Músculo Esquelético/metabolismo , Células-Tronco/metabolismo , Ultrassom/métodos , Animais , Técnicas de Cultura de Células , Feminino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos C3H , Modelos Animais , Músculo Esquelético/citologia , Músculo Esquelético/diagnóstico por imagem , Análise Espaço-Temporal , Células-Tronco/citologia , Células-Tronco/diagnóstico por imagem , Ultrassonografia
10.
Stem Cells ; 30(6): 1216-27, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22593018

RESUMO

Bone marrow stromal cells (BMSCs) have shown significant promise in the treatment of disease, but their therapeutic efficacy is often limited by inefficient homing of systemically administered cells, which results in low number of cells accumulating at sites of pathology. BMSC home to areas of inflammation where local expression of integrins and chemokine gradients is present. We demonstrated that nondestructive pulsed focused ultrasound (pFUS) exposures that emphasize the mechanical effects of ultrasound-tissue interactions induced local and transient elevations of chemoattractants (i.e., cytokines, integrins, and growth factors) in the murine kidney. pFUS-induced upregulation of cytokines occurred through approximately 1 day post-treatment and returned to contralateral kidney levels by day 3. This window of significant increases in cytokine expression was accompanied by local increases of other trophic factors and integrins that have been shown to promote BMSC homing. When BMSCs were intravenously administered following pFUS treatment to a single kidney, enhanced homing, permeability, and retention of BMSC was observed in the treated kidney versus the contralateral kidney. Histological analysis revealed up to eight times more BMSC in the peritubular regions of the treated kidneys on days 1 and 3 post-treatment. Furthermore, cytokine levels in pFUS-treated kidneys following BMSC administration were found to be similar to controls, suggesting modulation of cytokine levels by BMSC. pFUS could potentially improve cell-based therapies as a noninvasive modality to target homing by establishing local chemoattractant gradients and increasing expression of integrins to enhance tropism of cells toward treated tissues.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/diagnóstico por imagem , Transplante de Medula Óssea/métodos , Rim/citologia , Rim/diagnóstico por imagem , Células Estromais/transplante , Ultrassom/métodos , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/diagnóstico por imagem , Técnicas de Cultura de Células , Citocinas/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células Estromais/citologia , Ultrassonografia
12.
Trends Neurosci ; 45(6): 459-470, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35461727

RESUMO

The blood-brain barrier (BBB) continues to represent one of the most significant challenges for successful drug-based treatments of neurological disease. Mechanical modulation of the BBB using focused ultrasound (FUS) and microbubbles (MBs) has shown considerable promise in enhancing the delivery of therapeutics to the brain, but questions remain regarding possible long-term effects of such forced disruption. This review examines the evidence for inflammation associated with ultrasound-induced BBB disruption and potential strategies for managing such inflammatory effects to improve both the efficacy and safety of therapeutic ultrasound in neurological applications.


Assuntos
Barreira Hematoencefálica , Doenças Neuroinflamatórias , Transporte Biológico , Encéfalo , Humanos , Imageamento por Ressonância Magnética , Microbolhas
13.
Drug Metab Dispos ; 39(10): 1961-6, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21737567

RESUMO

Electron paramagnetic resonance (EPR) imaging using nitroxides as molecular probes is potentially a powerful tool for the detection and physiological characterization of micrometastatic lesions. Encapsulating nitroxides in anti-HER2 immunoliposomes at high concentrations to take advantage of the "self-quenching" phenomenon of nitroxides allows generation of robust EPR signals in HER2-overexpressing breast tumor cells with minimal background from indifferent tissues or circulating liposomes. We investigated the in vivo pharmacological properties of nitroxides encapsulated in sterically stabilized liposomes designed for long circulation times. We show that circulation times of nitroxides can be extended from hours to days; this increases the proportion of liposomes in circulation to enhance tumor targeting. Furthermore, nitroxides encapsulated in sterically stabilized anti-HER2 immunoliposomes can be delivered to HER2-overexpressing tumors at micromolar concentrations, which should be imageable by EPR. Lastly, after in vivo administration, liposomally encapsulated nitroxide signal also appears in the liver, spleen, and kidneys. Although these organs are spatially distinct and would not hinder tumor imaging in our model, understanding nitroxide signal retention in these organs is essential for further improvements in EPR imaging contrast between tumors and other tissues. These results lay the foundation to use liposomally delivered nitroxides and EPR imaging to visualize tumor cells in vivo.


Assuntos
Neoplasias da Mama/metabolismo , Óxidos N-Cíclicos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Lipossomos/farmacocinética , Sondas Moleculares/farmacocinética , Animais , Neoplasias da Mama/diagnóstico , Diagnóstico por Imagem , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Feminino , Lipossomos/administração & dosagem , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pirrolidinas/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Distribuição Tecidual , Transplante Heterólogo , Células Tumorais Cultivadas
14.
Theranostics ; 11(2): 602-613, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33391495

RESUMO

Mechanical forces from non-ablative pulsed focused ultrasound (pFUS) generate pro-inflammatory tumor microenvironments (TME), marked by increased cytokines, chemokines, and trophic factors, as well as immune cell infiltration and reduced tumor growth. pFUS also causes DNA damage within tumors, which is a potent activator of immunity and could contribute to changes in the TME. This study investigated mechanisms behind the mechanotransductive effects of pFUS causing DNA damage in several tumor cell types. Methods: 4T1 (murine breast tumor), B16 (murine melanoma), C6 (rat glioma), or MDA-MB-231 (human breast tumor) cells were sonicated in vitro (1.1MHz; 6MPa PNP; 10ms pulses; 10% duty cycle; 300 pulses). DNA damage was detected by TUNEL, apoptosis was measured by immunocytochemistry for cleaved caspase-3. Calcium, superoxide, and H2O2 were detected by fluorescent indicators and modulated by BAPTA-AM, mtTEMPOL, or Trolox, respectively. Results: pFUS increased TUNEL reactivity (range = 1.6-2.7-fold) in all cell types except C6 and did not induce apoptosis in any cell line. All lines displayed cytosolic Ca2+ transients during sonication. pFUS increased superoxide (range = 1.6-2.0-fold) and H2O2 (range = 2.3-2.8-fold) in all cell types except C6. BAPTA-AM blocked increased TUNEL reactivity, superoxide and H2O2 formation, while Trolox also blocked increased TUNEL reactivity increased after pFUS. mtTEMPOL allowed H2O2 formation and did not block increased TUNEL reactivity after pFUS. Unsonicated C6 cells had higher baseline concentrations of cytosolic Ca2+, superoxide, and H2O2, which were not associated with greater baseline TUNEL reactivity than the other cell lines. Conclusions: Mechanotransduction of pFUS directly induces DNA damage in tumor cells by cytosolic Ca2+ transients causing formation of superoxide and subsequently, H2O2. These results further suggest potential clinical utility for pFUS. However, the lack of pFUS-induced DNA damage in C6 cells demonstrates a range of potential tumor responses that may arise from physiological differences such as Ca2+ or redox homeostasis.


Assuntos
Neoplasias da Mama/patologia , Cálcio/metabolismo , Citosol/metabolismo , Dano ao DNA , Mecanotransdução Celular , Espécies Reativas de Oxigênio/metabolismo , Ondas Ultrassônicas , Apoptose , Neoplasias da Mama/metabolismo , Neoplasias da Mama/radioterapia , Proliferação de Células , Feminino , Humanos , Células Tumorais Cultivadas
15.
Cancers (Basel) ; 13(7)2021 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-33801627

RESUMO

Focused ultrasound (FUS) has shown promise as a non-invasive treatment modality for solid malignancies. FUS targeting to tumors has been shown to initiate pro-inflammatory immune responses within the tumor microenvironment. Pulsed FUS (pFUS) can alter the expression of cytokines, chemokines, trophic factors, cell adhesion molecules, and immune cell phenotypes within tissues. Here, we investigated the molecular and immune cell effects of pFUS on murine B16 melanoma and 4T1 breast cancer flank tumors. Temporal changes following sonication were evaluated by proteomics, RNA-seq, flow-cytometry, and histological analyses. Proteomic profiling revealed molecular changes occurring over 24 h post-pFUS that were consistent with a shift toward inflamed tumor microenvironment. Over 5 days post-pFUS, tumor growth rates were significantly decreased while flow cytometric analysis revealed differences in the temporal migration of immune cells. Transcriptomic analyses following sonication identified differences in gene expression patterns between the two tumor types. Histological analyses further demonstrated reduction of proliferation marker, Ki-67 in 4T1, but not in B16 tumors, and activated cleaved-caspase 3 for apoptosis remained elevated up to 3 days post-pFUS in both tumor types. This study revealed diverse biological mechanisms following pFUS treatment and supports its use as a possible adjuvant to ablative tumor treatment to elicit enhanced anti-tumor responses and slow tumor growth.

16.
Nat Neurosci ; 24(2): 245-258, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33462481

RESUMO

Cerebrovascular injuries can cause severe edema and inflammation that adversely affect human health. Here, we observed that recanalization after successful endovascular thrombectomy for acute large vessel occlusion was associated with cerebral edema and poor clinical outcomes in patients who experienced hemorrhagic transformation. To understand this process, we developed a cerebrovascular injury model using transcranial ultrasound that enabled spatiotemporal evaluation of resident and peripheral myeloid cells. We discovered that injurious and reparative responses diverged based on time and cellular origin. Resident microglia initially stabilized damaged vessels in a purinergic receptor-dependent manner, which was followed by an influx of myelomonocytic cells that caused severe edema. Prolonged blockade of myeloid cell recruitment with anti-adhesion molecule therapy prevented severe edema but also promoted neuronal destruction and fibrosis by interfering with vascular repair subsequently orchestrated by proinflammatory monocytes and proangiogenic repair-associated microglia (RAM). These data demonstrate how temporally distinct myeloid cell responses can contain, exacerbate and ultimately repair a cerebrovascular injury.


Assuntos
Encéfalo/imunologia , Inflamação/imunologia , AVC Isquêmico/imunologia , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Inflamação/diagnóstico por imagem , Inflamação/patologia , AVC Isquêmico/diagnóstico por imagem , AVC Isquêmico/patologia , Imageamento por Ressonância Magnética , Camundongos , Microglia , Células Mieloides
17.
Biochim Biophys Acta ; 1788(10): 2301-8, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19712666

RESUMO

Electron paramagnetic resonance imaging (EPRI) allows detection and localization of paramagnetic spin probes in vivo and in real time. We have shown that nitroxide spin probes entrapped in the intracellular milieu can be imaged by EPRI. Therefore, with the development of a tumor-targetable vehicle that can efficiently deliver nitroxides into cells, it should be possible to use nitroxide spin probes to label and image cells in a tumor. In this study, we assess the potential of liposomes as a delivery vehicle for imaging probes. We demonstrate that liposomes can stably encapsulate nitroxides at very high concentrations (>100 mM), at which nitroxides exhibit concentration-dependent quenching of their EPR signal-a process analogous to the quenching of fluorescent molecules. The encapsulating liposomes thus appear spectroscopically "dark". When the liposomes are endocytosed and degraded by cells, the encapsulated nitroxides are liberated and diluted into the much larger intracellular volume. The consequent relief of quenching generates a robust intracellular nitroxide signal that can be imaged. We show that through endocytosis of nitroxide-loaded liposomes, CV1 cells can achieve intracellular nitroxide concentrations of approximately 1 mM. By using tissue phantom models, we verify that this concentration is more than sufficient for in vivo EPR imaging.


Assuntos
Espectroscopia de Ressonância de Spin Eletrônica , Endocitose/fisiologia , Processamento de Imagem Assistida por Computador , Lipossomos/química , Óxido Nítrico/metabolismo , Marcadores de Spin , Animais , Células Cultivadas , Chlorocebus aethiops , Diagnóstico por Imagem , Camundongos , Camundongos Endogâmicos C3H
18.
Breast Cancer Res Treat ; 124(1): 121-31, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20066490

RESUMO

Electron paramagnetic resonance (EPR) imaging is an emerging modality that can detect and localize paramagnetic molecular probes (so-called spin probes) in vivo. We previously demonstrated that nitroxide spin probes can be encapsulated in liposomes at concentrations exceeding 100 mM, at which nitroxides exhibit a concentration-dependent quenching of their EPR signal that is analogous to the self-quenching of fluorescent molecules. Therefore, intact liposomes encapsulating high concentrations of nitroxides exhibit greatly attenuated EPR spectral signals, and endocytosis of such liposomes represents a cell-activated contrast-generating mechanism. After endocytosis, the encapsulated nitroxide is liberated and becomes greatly diluted in the intracellular milieu. This dequenches the nitroxides to generate a robust intracellular EPR signal. It is therefore possible to deliver a high concentration of nitroxides to cells while minimizing background signal from unendocytosed liposomes. We report here that intracellular EPR signal can be selectively generated in a specific cell type by exploiting its expression of Human Epidermal Growth Factor Receptor 2 (HER2). When targeted by anti-HER2 immunoliposomes encapsulating quenched nitroxides, Hc7 cells, which are novel HER2-overexpressing cells derived from the MCF7 breast tumor cell line, endocytose the liposomes copiously, in contrast to the parent MCF7 cells or control CV1 cells, which do not express HER2. HER2-dependent liposomal delivery enables Hc7 cells to accumulate 750 µM nitroxide intracellularly. Through the use of phantom models, we verify that this concentration of nitroxides is more than sufficient for EPR imaging, thus laying the foundation for using EPR imaging to visualize HER2-overexpressing Hc7 tumors in animals.


Assuntos
Anticorpos Monoclonais/metabolismo , Neoplasias da Mama/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Fragmentos Fab das Imunoglobulinas/metabolismo , Sondas Moleculares , Óxidos de Nitrogênio/metabolismo , Receptor ErbB-2/metabolismo , Marcadores de Spin , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Espectroscopia de Ressonância de Spin Eletrônica/instrumentação , Endocitose , Feminino , Humanos , Lipossomos , Microscopia de Fluorescência , Imagens de Fantasmas , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Transfecção , Trastuzumab , Regulação para Cima
19.
J Org Chem ; 75(19): 6463-7, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20828113

RESUMO

Electron paramagnetic resonance imaging (EPRI) using nitroxides is an emergent imaging method for studying in vivo physiology, including O(2) distribution in various tissues. Such imaging capabilities would allow O(2) mapping in tumors and in different brain regions following hypoxia or drug abuse. We have recently demonstrated that the anion of 3-carboxy-2,2,5,5-tetramethyl-1-pyrrolidinyloxyl (2) can be entrapped in brain tissue to quantitate O(2) concentration in vivo. To increase the sensitivity of O(2) measurement by EPR imaging, we synthesized 3-carboxy-2,2,5,5-tetra((2)H(3))methyl-1-(3,4,4-(2)H(3),1-(15)N)pyrrolidinyloxyl (7). EPR spectroscopic measurements demonstrate that this fully isotopically substituted nitroxide markedly improves signal-to-noise ratio and, therefore, the sensitivity of EPR imaging. The new isotopically substituted nitroxide shows increased sensitivity to changes in O(2) concentration, which will enable more accurate O(2) measurement in tissues using EPRI.


Assuntos
Deutério/química , Sondas Moleculares/química , Pirrolidinas/química , Espectroscopia de Ressonância de Spin Eletrônica , Estrutura Molecular , Isótopos de Nitrogênio , Oxigênio/análise , Pirrolidinas/síntese química , Estereoisomerismo
20.
J Org Chem ; 75(14): 4737-41, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20540511

RESUMO

Nitroxides with narrow linewidths are essential for low-frequency EPR spectroscopy and in vivo EPR imaging. In developing a framework for designing narrow-line nitroxides, we sought to understand the unexpectedly narrow line width of 4-oxo-2,2,6,6-tetramethyl-1-piperidinyloxyl (5). Computational modeling revealed that the carbonyl double bond in the 4-position allows conformational diversity that results in the observed narrowing of the EPR spectral line. In view of this finding, we synthesized two new nitroxides bearing an exocyclic double bond: 4-methoxycarbonylmethylidene-2,2,6,6-tetramethyl-1-piperidinyloxyl (7) and 4-acetoxymethoxycarbonylmethylidene-2,2,6,6-tetramethyl-1-piperidinyloxyl (9). These nitroxides, like nitroxide 5, exhibited narrow linewidths-consistent with the results of modeling. Nitroxide 8 (4-carboxymethylidene-2,2,6,6-tetramethyl-1-piperidinyloxyl), as a prototype, allows for a variety of structural diversity, such as nitroxide 9,that can, for instance, target tissue compartments for in vivo EPR imaging.


Assuntos
Íons/química , Óxidos de Nitrogênio/química , Piperidinas/química , Cristalografia por Raios X , Espectroscopia de Ressonância de Spin Eletrônica , Estrutura Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA