Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Res ; 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849501

ABSTRACT

Physiologically, the atria contract first, followed by the ventricles, which is the prerequisite for normal blood circulation. The above phenomenon of atrioventricular sequential contraction results from the characteristically slow conduction of electrical excitation of the atrioventricular node (AVN) between the atria and the ventricles. However, it is not clear what controls the conduction of electrical excitation within AVNs. Here, we find that AVN pacemaker cells (AVNPCs) possess an intact intrinsic GABAergic system, which plays a key role in electrical conduction from the atria to the ventricles. First, along with the discovery of abundant GABA-containing vesicles under the surface membranes of AVNPCs, key elements of the GABAergic system, including GABA metabolic enzymes, GABA receptors, and GABA transporters, were identified in AVNPCs. Second, GABA synchronously elicited GABA-gated currents in AVNPCs, which significantly weakened the excitability of AVNPCs. Third, the key molecular elements of the GABAergic system markedly modulated the conductivity of electrical excitation in the AVN. Fourth, GABAA receptor deficiency in AVNPCs accelerated atrioventricular conduction, which impaired the AVN's protective potential against rapid ventricular frequency responses, increased susceptibility to lethal ventricular arrhythmias, and decreased the cardiac contractile function. Finally, interventions targeting the GABAergic system effectively prevented the occurrence and development of atrioventricular block. In summary, the endogenous GABAergic system in AVNPCs determines the slow conduction of electrical excitation within AVNs, thereby ensuring sequential atrioventricular contraction. The endogenous GABAergic system shows promise as a novel intervention target for cardiac arrhythmias.

2.
Circ Res ; 134(3): 290-306, 2024 02 02.
Article in English | MEDLINE | ID: mdl-38197258

ABSTRACT

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is the most prevalent monogenic heart disorder. However, the pathogenesis of HCM, especially its nongenetic mechanisms, remains largely unclear. Transcription factors are known to be involved in various biological processes including cell growth. We hypothesized that SP1 (specificity protein 1), the first purified TF in mammals, plays a role in the cardiomyocyte growth and cardiac hypertrophy of HCM. METHODS: Cardiac-specific conditional knockout of Sp1 mice were constructed to investigate the role of SP1 in the heart. The echocardiography, histochemical experiment, and transmission electron microscope were performed to analyze the cardiac phenotypes of cardiac-specific conditional knockout of Sp1 mice. RNA sequencing, chromatin immunoprecipitation sequencing, and adeno-associated virus experiments in vivo were performed to explore the downstream molecules of SP1. To examine the therapeutic effect of SP1 on HCM, an SP1 overexpression vector was constructed and injected into the mutant allele of Myh6 R404Q/+ (Myh6 c. 1211C>T) HCM mice. The human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from a patient with HCM were used to detect the potential therapeutic effects of SP1 in human HCM. RESULTS: The cardiac-specific conditional knockout of Sp1 mice developed a typical HCM phenotype, displaying overt myocardial hypertrophy, interstitial fibrosis, and disordered myofilament. In addition, Sp1 knockdown dramatically increased the cell area of hiPSC-CMs and caused intracellular myofibrillar disorganization, which was similar to the hypertrophic cardiomyocytes of HCM. Mechanistically, Tuft1 was identified as the key target gene of SP1. The hypertrophic phenotypes induced by Sp1 knockdown in both hiPSC-CMs and mice could be rescued by TUFT1 (tuftelin 1) overexpression. Furthermore, SP1 overexpression suppressed the development of HCM in the mutant allele of Myh6 R404Q/+ mice and also reversed the hypertrophic phenotype of HCM hiPSC-CMs. CONCLUSIONS: Our study demonstrates that SP1 deficiency leads to HCM. SP1 overexpression exhibits significant therapeutic effects on both HCM mice and HCM hiPSC-CMs, suggesting that SP1 could be a potential intervention target for HCM.


Subject(s)
Cardiomyopathy, Hypertrophic , Induced Pluripotent Stem Cells , Humans , Mice , Animals , Induced Pluripotent Stem Cells/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Myofibrils/metabolism , Myocytes, Cardiac/metabolism , Cardiomegaly/metabolism , Transcription Factors/metabolism , Mammals
3.
Nat Commun ; 14(1): 1796, 2023 03 31.
Article in English | MEDLINE | ID: mdl-37002228

ABSTRACT

The growth and maturation of the ventricular chamber require spatiotemporally precise synergy between diverse cell types. Alternative splicing deeply affects the processes. However, the functional properties of alternative splicing in cardiac development are largely unknown. Our study reveals that an alternative splicing factor polypyrimidine tract-binding protein 1 (PTBP1) plays a key role in ventricular chamber morphogenesis. During heart development, PTBP1 colocalizes with endothelial cells but is almost undetectable in cardiomyocytes. The endothelial-specific knockout of Ptbp1, in either endocardial cells or pan-endothelial cells, leads to a typical phenotype of left ventricular noncompaction (LVNC). Mechanistically, the deletion of Ptbp1 reduces the migration of endothelial cells, disrupting cardiomyocyte proliferation and ultimately leading to the LVNC. Further study shows that Ptbp1 deficiency changes the alternative splicing of ß-arrestin-1 (Arrb1), which affects endothelial cell migration. In conclusion, as an alternative splicing factor, PTBP1 is essential during ventricular chamber development, and its deficiency can lead to congenital heart disease.


Subject(s)
Endothelial Cells , Polypyrimidine Tract-Binding Protein , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Endothelial Cells/metabolism , Alternative Splicing/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism
4.
J Cell Mol Med ; 26(10): 2981-2994, 2022 05.
Article in English | MEDLINE | ID: mdl-35429093

ABSTRACT

The neonatal heart can efficiently regenerate within a short period after birth, whereas the adult mammalian heart has extremely limited capacity to regenerate. The molecular mechanisms underlying neonatal heart regeneration remain elusive. Here, we revealed that as a coreceptor of Wnt signalling, low-density lipoprotein receptor-related protein 5 (LRP5) is required for neonatal heart regeneration by regulating cardiomyocyte proliferation. The expression of LRP5 in the mouse heart gradually decreased after birth, consistent with the time window during which cardiomyocytes withdrew from the cell cycle. LRP5 downregulation reduced the proliferation of neonatal cardiomyocytes, while LRP5 overexpression promoted cardiomyocyte proliferation. The cardiac-specific deletion of Lrp5 disrupted myocardial regeneration after injury, exhibiting extensive fibrotic scars and cardiac dysfunction. Mechanistically, the decreased heart regeneration ability induced by LRP5 deficiency was mainly due to reduced cardiomyocyte proliferation. Further study identified AKT/P21 signalling as the key pathway accounting for the regulation of cardiomyocyte proliferation mediated by LRP5. LRP5 downregulation accelerated the degradation of AKT, leading to increased expression of the cyclin-dependent kinase inhibitor P21. Our study revealed that LRP5 is necessary for cardiomyocyte proliferation and neonatal heart regeneration, providing a potential strategy to repair myocardial injury.


Subject(s)
Heart , Low Density Lipoprotein Receptor-Related Protein-5 , Myocytes, Cardiac , Regeneration , Animals , Cell Proliferation , Heart/physiology , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Mice , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Wnt Signaling Pathway
5.
Adv Sci (Weinh) ; 8(22): e2102666, 2021 11.
Article in English | MEDLINE | ID: mdl-34523247

ABSTRACT

Necrosis induces strong inflammation with undesirable implications in clinics compared with apoptosis. Fortunately, the switch between necrosis and apoptosis could be realized by tailoring the appropriate structural properties of gold nano rods (GNRs) that could precisely modulate cell death pathways. Herein, the intracellular interaction between GNRs and organelles is monitored and it is found that lysosomes dominates necrosis/apoptosis evoking. Then the surface molecule density of GNRs, which is first defined as ρsurf. molecule (Nsurf. molecules /(a × π × Diameter × Length)), mediates lysosome activities as the membrane permeabilization (LMP), the Cathepsin B and D release, the cross-talk between lysosome and different organelles, which selectively evokes apoptosis or necrosis and the production of TNF-α from macrophages. GNRs with small ρsurf. molecule mainly induce apoptosis, while with large ρsurf. molecule they greatly contribute to necrosis. Interestingly, necrosis can be suppressed by GNRs with higher ρsurf. molecule due to the overexpression of key protease caspase 8, which cleaves the RIP1-RIP3 complex and activates caspase 3 followed by necrosis to apoptosis transition. This investigation indicates that the ρsurf. molecule greatly affects the utility of nanomaterials and different structural properties of nanomaterials have different implications in clinics.


Subject(s)
Apoptosis , Gold/chemistry , Nanotubes/chemistry , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Mice, Nude , Necrosis
6.
J Integr Med ; 19(5): 408-417, 2021 09.
Article in English | MEDLINE | ID: mdl-34130942

ABSTRACT

OBJECTIVE: Bufalin is an effective drug for the treatment of liver cancer. But its high toxicity, poor water-solubility, fast metabolism and short elimination half-life limit its use in tumor treatment. How to make the drug accumulate in the tumor and reduce side effects while maintaining its efficacy are urgent problems to be solved. The goal of this study is to solve these problems. METHODS: A copolymer with tunable poly-N-isopropylacrylamide and polylactic acid was designed and synthesized. The corresponding dual targeting immunomicelles (DTIs) loaded with bufalin (DTIs-BF) were synthesized by copolymer self-assembly in an aqueous solution. The size and structure of DTIs-BF were determined by ZetaSizer Nano-ZS and transmission electron microscopy. Then, its temperature sensitivity, serum stability, critical micelle concentration (CMC), entrapment efficiency (EE), drug release and non-cytotoxicity of blank block copolymer micelles (BCMs) were evaluated. Next, the effects of DTIs-BF on cellular uptake, cytotoxicity, and tumor cell inhibition were evaluated. Finally, the accumulation of DTIs-fluorescein isothiocyanate (FITC) and the in vivo anti-tumor effect were observed using an interactive video information system. RESULTS: DTIs-BF had a small size, spherical shape, good temperature sensitivity, high serum stability, low CMC, high EE, and slow drug release. The blank BCMs had very low cytotoxicity. Compared with free bufalin, the in vitro cellular internalization and cytotoxicity of DTIs-BF against SMMC-7721 cells were significantly enhanced, and the effects were obviously better at 40 °C than 37 °C. In addition, the therapeutic effect on SMMC-7721 cells was further enhanced by the programmed cell death specifically caused by bufalin. When DTIs-FITC were injected intravenously in BALB/c nude mice bearing liver cancer, the accumulation of FITC was significantly increased in tumors. CONCLUSION: DTIs-BF is a potentially effective nano-formulation and has broad prospects in the clinical treatment of liver cancer.


Subject(s)
Antineoplastic Agents , Liver Neoplasms , Animals , Antineoplastic Agents/pharmacology , Bufanolides , Cell Line, Tumor , Liver Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Mice, Nude
7.
Protein Cell ; 12(7): 545-556, 2021 07.
Article in English | MEDLINE | ID: mdl-33548033

ABSTRACT

Activation of the heart normally begins in the sinoatrial node (SAN). Electrical impulses spontaneously released by SAN pacemaker cells (SANPCs) trigger the contraction of the heart. However, the cellular nature of SANPCs remains controversial. Here, we report that SANPCs exhibit glutamatergic neuron-like properties. By comparing the single-cell transcriptome of SANPCs with that of cells from primary visual cortex in mouse, we found that SANPCs co-clustered with cortical neurons. Tissue and cellular imaging confirmed that SANPCs contained key elements of glutamatergic neurotransmitter system, expressing genes encoding glutamate synthesis pathway (Gls), ionotropic and metabotropic glutamate receptors (Grina, Gria3, Grm1 and Grm5), and glutamate transporters (Slc17a7). SANPCs highly expressed cell markers of glutamatergic neurons (Snap25 and Slc17a7), whereas Gad1, a marker of GABAergic neurons, was negative. Functional studies revealed that inhibition of glutamate receptors or transporters reduced spontaneous pacing frequency of isolated SAN tissues and spontaneous Ca2+ transients frequency in single SANPC. Collectively, our work suggests that SANPCs share dominant biological properties with glutamatergic neurons, and the glutamatergic neurotransmitter system may act as an intrinsic regulation module of heart rhythm, which provides a potential intervention target for pacemaker cell-associated arrhythmias.


Subject(s)
Biological Clocks/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Primary Visual Cortex/metabolism , Sinoatrial Node/metabolism , Transcriptome , Action Potentials/physiology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Calcium/metabolism , Carrier Proteins/classification , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/classification , Nerve Tissue Proteins/metabolism , Neurons/cytology , Primary Visual Cortex/cytology , Receptors, Ionotropic Glutamate/classification , Receptors, Ionotropic Glutamate/genetics , Receptors, Ionotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/classification , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Single-Cell Analysis , Sinoatrial Node/cytology , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
8.
Nat Commun ; 12(1): 287, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436583

ABSTRACT

Bioelectrical impulses intrinsically generated within the sinoatrial node (SAN) trigger the contraction of the heart in mammals. Though discovered over a century ago, the molecular and cellular features of the SAN that underpin its critical function in the heart are uncharted territory. Here, we identify four distinct transcriptional clusters by single-cell RNA sequencing in the mouse SAN. Functional analysis of differentially expressed genes identifies a core cell cluster enriched in the electrogenic genes. The similar cellular features are also observed in the SAN from both rabbit and cynomolgus monkey. Notably, Vsnl1, a core cell cluster marker in mouse, is abundantly expressed in SAN, but is barely detectable in atrium or ventricle, suggesting that Vsnl1 is a potential SAN marker. Importantly, deficiency of Vsnl1 not only reduces the beating rate of human induced pluripotent stem cell - derived cardiomyocytes (hiPSC-CMs) but also the heart rate of mice. Furthermore, weighted gene co-expression network analysis (WGCNA) unveiled the core gene regulation network governing the function of the SAN in mice. Overall, these findings reveal the whole transcriptome profiling of the SAN at single-cell resolution, representing an advance toward understanding of both the biology and the pathology of SAN.


Subject(s)
Mammals/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Sinoatrial Node/cytology , Animals , Biological Clocks , Cell Aggregation , Cluster Analysis , Gene Expression Regulation , Gene Regulatory Networks , Heart Rate , Induced Pluripotent Stem Cells/cytology , Macaca fascicularis , Mice , Myocytes, Cardiac/metabolism , Neurocalcin/deficiency , Neurocalcin/metabolism , Rabbits , Species Specificity , Stochastic Processes
9.
Cell Res ; 31(4): 450-462, 2021 04.
Article in English | MEDLINE | ID: mdl-32973339

ABSTRACT

The adult mammalian heart is thought to be a terminally differentiated organ given the postmitotic nature of cardiomyocytes. Consequently, the potential for cardiac repair through cardiomyocyte proliferation is extremely limited. Low-density lipoprotein receptor-related protein 6 (LRP6) is a Wnt co-receptor that is required for embryonic heart development. In this study we investigated the role of LRP6 in heart repair through regulation of cardiomyocyte proliferation. Lrp6 deficiency increased cardiomyocyte cell cycle activity in neonatal, juvenile and adult mice. Cardiomyocyte-specific deletion of Lrp6 in the mouse heart induced a robust regenerative response after myocardial infarction (MI), led to reduced MI area and improvement in left ventricular systolic function. In vivo genetic lineage tracing revealed that the newly formed cardiomyocytes in Lrp6-deficient mouse hearts after MI were mainly derived from resident cardiomyocytes. Furthermore, we found that the pro-proliferative effect of Lrp6 deficiency was mediated by the ING5/P21 signaling pathway. Gene therapy using the adeno-associated virus (AAV)9 miRNAi-Lrp6 construct promoted the repair of heart injury in mice. Lrp6 deficiency also induced the proliferation of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Our study identifies LRP6 as a critical regulator of cardiomyocyte proliferation, which may lead to the development of a novel molecular strategy to promote myocardial regeneration and repair.


Subject(s)
Heart/physiology , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Down-Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Regeneration
10.
Circ Arrhythm Electrophysiol ; 12(4): e007097, 2019 04.
Article in English | MEDLINE | ID: mdl-30939909

ABSTRACT

BACKGROUND: Kv1.5 (Potassium voltage-gated channel subfamily A member 5) has been regarded as a promising target of interventions for atrial fibrillation (AF). SNX17 (sorting nexin 17), a member of the SNXs (sorting nexin family), regulates the intracellular trafficking of membrane proteins through its FERM (four-point-one, ezrin, radixin, moesin) domain. However, whether SNX17 regulates the trafficking process of Kv1.5 remains unknown. METHODS: A SNX17 knockout rat line was generated to test the role of SNX17 in atrial electrophysiology. The protein expression of SNX17 and membrane ion channels was detected by Western blotting. Electrophysiology changes in the atrial tissue and myocytes were analyzed by optical mapping and patch clamp, respectively. Acetylcholine and electrical stimulation were used to induce AF, and ECG recording was adopted to assess the influence of SNX17 deficiency on AF susceptibility. The spatial relationship between Kv1.5 and SNX17 was evaluated by immunostaining and confocal scanning, and the functional region of SNX17 regulating Kv1.5 trafficking was identified using plasmids with truncated SNX17 domains. RESULTS: Embryonic death occurred in homozygous SNX17 knockout rats. SNX17 heterozygous rats survived, and the level of the SNX17 protein in the atrium was decreased by ≈50%. SNX17 deficiency increased the membrane expression of Kv1.5 and atria-specific ultrarapid delayed rectifier outward potassium current ( IKur) density, resulting in a shortened action potential duration, and eventually contributing to AF susceptibility. Mechanistically, SNX17 facilitated the endocytic sorting of Kv1.5 from the plasma membrane to early endosomes via the FERM domain. CONCLUSIONS: SNX17 mediates susceptibility to AF by regulating endocytic sorting of the Kv1.5 channel through the FERM domain. SNX17 could be a potential target for the development of new drugs for AF.


Subject(s)
Atrial Fibrillation/physiopathology , Potassium Channels, Voltage-Gated/physiology , Sorting Nexins/physiology , Animals , Blotting, Western , Electrocardiography , Electrophysiologic Techniques, Cardiac , HEK293 Cells , Humans , Microscopy, Confocal , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
11.
Int J Nanomedicine ; 13: 2585-2599, 2018.
Article in English | MEDLINE | ID: mdl-29750031

ABSTRACT

INTRODUCTION: Pancreatic cancer (PC) has a poor prognosis with high mortality, due to the lack of effective early diagnostic and prognostic tools. MATERIALS AND METHODS: In order to target and diagnose PC, we developed a dual-modal imaging probe using Glypican-1 (GPC-1) antibody conjugated with Gd-Au nanoclusters (NCs; Gd-Au-NC-GPC-1). GPC-1 is a type of cell surface heparan sulfate proteoglycan, which is often highly expressed in PC. The probe was successfully prepared with a hydrodynamic diameter ranging from 13.5 to 24.4 nm. RESULTS: Spectral characteristics showed absorption at 280 nm and prominent emission at 650 nm. Confocal microscopic imaging showed effective detection of GPC-1 highly expressed PC cells by Gd-Au-NC-GPC-1, which was consistent with flow cytometry results. In vitro relaxivity characterization demonstrated that the r1 value of the probe was 17.722 s-1 mM-1 Gd, which was almost 4 times higher compared with that of Gd-diethylenetriaminepentacetate (DTPA; r1 value =4.6 s-1 mM-1 Gd). Gd-Au-NC-GPC-1 exhibited similar magnetic resonance (MR) signals when compared to Gd-DTPA even at lower Gd concentrations. Much higher MR signals were registered in PC cells (COLO-357) compared with normal cells (293T). Furthermore, Gd-Au-NC-GPC-1 could effectively detect PC cells in vivo by dual-modal fluorescence imaging/magnetic resonance imaging (FI/MRI) at 30 minutes postinjection. In addition, Gd-Au-NC-GPC-1 did not show significant biotoxicity to normal cells at tested concentrations both in vitro and in vivo. CONCLUSION: Gd-Au-NC-GPC-1 has demonstrated to be a promising dual-modal FI/MRI contrast agent for targeted diagnosis of PC.


Subject(s)
Contrast Media/chemistry , Glypicans/immunology , Nanostructures/chemistry , Optical Imaging/methods , Pancreatic Neoplasms/diagnostic imaging , Animals , Antibodies/chemistry , Antibodies/immunology , Cell Line, Tumor , Flow Cytometry , Gadolinium/chemistry , Gadolinium DTPA , Gold/chemistry , Humans , Magnetic Resonance Imaging/methods , Male , Mice, Nude , Molecular Probe Techniques , Xenograft Model Antitumor Assays
12.
Curr Drug Metab ; 19(1): 75-82, 2018.
Article in English | MEDLINE | ID: mdl-29268683

ABSTRACT

BACKGROUND: Nanomedicine, which is defined as application of nanoparticles in medicine, has offered new hopes for overcoming the drawbacks appeared in traditional chemotherapy. The size of nanomedicine normally in the range from 1 to 200 nm endows its potential applications in cancer therapy. But in clinics, there is still a gap between the in vitro physicochemical properties and the cellular level performance. METHOD: The physicochemical properties include size, shape, surface chemistry, surface topology, and surface properties strongly affect nanomedicine inter-/intra-cellular efficiency. Herein, this article reviews effects of physicochemical properties of nanomedicine on the cellular endocytosis and intracellular route. And strategies of nanomedicine optimization are also discussed from different perspectives. RESULTS: On the one hand, not as that of the traditional small molecular agents, the cellular endocytosis pathway and efficiency of nanomedicine is related to its size, structure and surface properties. On the other hand, the intracellular conditions also affect the intracellular route of nanomedicine. CONCLUSION: Nanomedicine of different scale size is internalized through different pathways. While different sensitivities to intracellular conditions determined by physicochemical properties of nanomedicine will lead to different cellular consumption. So, both the properties of nanomedicine and the intracellular conditions play important roles in cellular metabolism. Consequently, nanocarriers finely engineered as the above principles can provide practical solution to the problems appeared in cellular level for promoting traditional cancer therapy.


Subject(s)
Nanoparticles/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , Endocytosis/physiology , Humans , Nanomedicine/methods , Particle Size
13.
Int J Nanomedicine ; 12: 4019-4035, 2017.
Article in English | MEDLINE | ID: mdl-28603416

ABSTRACT

The Chinese traditional medicine Shikonin is an ideal drug due to its multiple targets to tumor cells. But in clinics, improving its aqueous solubility and tumor accumulation is still a challenge. Herein, a copolymer with tunable poly(N-isopropylacrymaide) and polylactic acid block lengths is designed, synthesized, and characterized in nuclear magnetic resonance. The corresponding thermosensitive nanomicelle (TN) with well-defined core-shell structure is then assembled in an aqueous solution. For promoting the therapeutic index, the physical-chemistry properties of TNs including narrow size, low critical micellar concentration, high serum stability, tunable volume phase transition temperature (VPTT), high drug-loading capacity, and temperature-controlled drug release are systematically investigated and regulated through the fine self-assembly. The shikonin is then entrapped in a degradable inner core resulting in a shikonin-loaded thermosensitive nanomicelle (STN) with a VPTT of ~40°C. Compared with small-molecular shikonin, the in vitro cellular internalization and cytotoxicity of STN against breast cancer cells (Michigan Cancer Foundation-7) are obviously enhanced. In addition, the therapeutic effect is further enhanced by the programmed cell death (PCD) specifically evoked by shikonin. Interestingly, both the proliferation inhibition and PCD are synergistically promoted as T > VPTT, namely the temperature-regulated passive targeting. Consequently, as intravenous injection is administered to the BALB/c nude mice bearing breast cancer, the intratumor accumulation of STNs is significantly increased as T > VPTT, which is regulated by the in-house developed heating device. The in vivo antitumor assays against breast cancer further confirm the synergistically enhanced therapeutic efficiency. The findings of this study indicate that STN is a potential effective nanoformulation in clinical cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/therapy , Hyperthermia, Induced/methods , Naphthoquinones/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Delayed-Action Preparations , Drugs, Chinese Herbal/chemistry , Female , Humans , MCF-7 Cells , Mice, Inbred BALB C , Micelles , Nanostructures/chemistry , Naphthoquinones/chemistry , Naphthoquinones/pharmacokinetics , Polyesters/chemistry , Polymers/chemistry , Solubility , Temperature , Tissue Distribution , Xenograft Model Antitumor Assays
14.
Nanomedicine (Lond) ; 12(13): 1575-1589, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28478730

ABSTRACT

AIM: This article aims to explain the necrosis mechanisms of cancer cells specifically induced by gold nanorods (GNRs). METHODS: The intracellular route and location of GNRs, the interaction between GNRs and lysosome, lysosome damage, cathepsin B release, necrosis complex formation, receptor-interacting protein 1 and TNF-α expression were systematically investigated. RESULTS: The GNRs with serum corona were internalized quickly by cancer cells and finally taken up by lysosomes. The GNRs damaged the lysosomal membrane, resulting in the leakage of cathepsin B, which promoted the activation of receptor-interacting protein 1 and necrosomes formation. Necrotic cells and their debris or ill cellular contents were engulfed by macrophages resulting in high-level release of TNF-α, which further confirmed necrosis. CONCLUSION: GNRs can specifically trigger lysosome-dependent necrosis in cancer cells.


Subject(s)
Gold/pharmacology , Lysosomes/metabolism , Nanotubes/chemistry , Animals , Apoptosis , Cathepsin B/metabolism , Cell Line, Tumor , Cricetulus , Gold/chemistry , HEK293 Cells , Humans , Lysosomes/ultrastructure , Mice , Mice, Inbred C57BL , Necrosis , Nuclear Pore Complex Proteins/metabolism , Particle Size , RNA-Binding Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
J Control Release ; 254: 107-118, 2017 05 28.
Article in English | MEDLINE | ID: mdl-28342982

ABSTRACT

This manuscript describes a synergistic therapy for mastocarcinoma by pH and temperature dual-sensitive nanogel, and effects of microstructure, composition and properties of nanogel on the cellular response mechanism. The extracellular internalization of nanogels was obviously enhanced, due to the passive targeting function at T>VPTT. Interestingly, the increased cytotoxicity was further synergistically enhanced by an unexpected apoptosis as evoked by the 5-fluorouracil loaded nanogel (FLNG). The systemically evaluation of the effectors generated from different sub-cellular organelles including endosome, lysosome, autophagosome confirmed that it was a lysomal dependent apoptosis. Such specific apoptosis was mainly attributed to its activatable protonated PEI at low pH, which caused lysosomal membrane destruction and lysosomal enzyme cathepsin B (Cat B) leakage. This Cat B was then translocated to the mitochondria resulting in mitochondrial membrane permeability increase and mitochondrial membrane potential (MMP) decrease, followed by cytochrome c (Cyt C) release. Cyt C was the main molecule that evoked apoptosis as reflected by overexpression of caspase 9. Additionally, such lysosome dependent, apoptosis was further enhanced by the passive cellular targeting at T>VPTT. Thus, the tumor growth inhibition was synergistically enhanced by the extracellular temperature dependent passive targeting and intracellular pH activatable lysosomal dependent apoptosis.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Fluorouracil/administration & dosage , Lysosomes/metabolism , Nanostructures/chemistry , Animals , Caspase 9/metabolism , Cathepsin B/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Cytochromes c/metabolism , Drug Carriers , Female , Gels , Humans , Hydrogen-Ion Concentration , Imines/chemistry , Membrane Potential, Mitochondrial , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy , Particle Size , Polyethylenes/chemistry
16.
Nanoscale Res Lett ; 12(1): 73, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28124299

ABSTRACT

Nanomedicine offers new hope to overcome the low solubility and high side toxicity to normal tissue appeared in traditional chemotherapy. The biocompatibility and intracellular drug accumulation is still a big challenge for the nano-based formulations. Herein, a medical-used biocompatible arabinoxylan (AX) is used to develop to delivery chemodrug doxorubicin (DOX). The solubility of DOX is obviously enhanced via the hydrogen bond formed with AX which results in an amphiphilic AX-DOX. A micelle with pH-cleavable bond is thus self-assembled from such AX-DOX with DOX core and AX shell. The inner DOX can be easily released out at low intracellular pH, which obviously enhanced its in vitro cytotoxicity against breast cancer cells (MCF-7). Interestingly, an unexpected apoptosis is evoked except for the proliferation inhibition. Moreover, the therapeutic effects are further synergistically promoted by the enhanced permeability and retention (EPR) and intracellular pH-triggered drug release. Consequently, the in vivo intratumor accumulation of DOX, the tumor inhibition was significantly promoted after intravenous administration to the Balb/c nude mice bearing MCF-7 tumors. These in vitro/vivo results indicated that the AX-DOX micellular formulation holds high potential in cancer therapy.

17.
Nanoscale ; 9(6): 2219-2231, 2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28120979

ABSTRACT

As the most common endocrine malignancy with a high incidence, thyroid cancer lacks a dual-modal imaging method for precise diagnosis. An accurate and multimodal imaging system is pivotal to solve this problem. Herein, dual-modality fluorescence/Computed Tomography (CT) iodinated gold nanoclusters for malignant thyroid cancer visualization have been recently fabricated. In this study, innovative iodinated gold nanoclusters (AuNCs@BSA-I) were synthesized via Bovine serum albumin (BSA) and chloramine-T. AuNCs@BSA-I not only possess an ultra-small size and brilliant biocompatibility but also exhibit excellent fluorescence/CT imaging properties. Particularly with regard to CT imaging properties, AuNCs@BSA-I rival the clinical CT contrast medium. And the fluorescence emission spectrum of AuNCs@BSA-I falls in the near infrared region (NIR). For further translational application in medicine, we established an orthotopic human thyroid cancer patient tissue derived xenograft (PDX) mouse model, highly close to the actual human thyroid cancer. The results unveil that AuNCs@BSA-I exert sensitive and accurate diagnosis characteristics. To be more specific, the AuNCs@BSA-I fluorescent/CT signals in the thyroid tumor represent characteristics of 'slow in fast out', compared to those in the normal thyroid. Moreover, AuNCs@BSA-I could distinguish minimal thyroid cancer, as small as 2 mm3. Therefore, AuNCs@BSA-I appear to be a promising nanoprobe which could be applied to preclinical medicine.


Subject(s)
Gold , Metal Nanoparticles , Thyroid Neoplasms/diagnostic imaging , Animals , Cell Line, Tumor , Chloramines , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/diagnosis , Serum Albumin, Bovine , Spectrometry, Fluorescence , Tomography, X-Ray Computed , Tosyl Compounds
18.
Nanomedicine (Lond) ; 11(15): 1993-2006, 2016 08.
Article in English | MEDLINE | ID: mdl-27256301

ABSTRACT

AIM: The main aim of this article is to explain the apoptosis mechanisms of cancer cells specifically triggered by gold nanorods (GNRs). MATERIALS & METHODS: GNRs were synthesized and optimized, the lysosome damage, cathepsin D, mitochondrial membrane potential, caspase-9, cleaved caspase-9, caspase-3 and intracellular GNRs location related to apoptosis was systematically evaluated. RESULTS: GNRs specifically induce cancer cell apoptosis while posing a negligible impact on normal cells. After incubation with GNRs, the lysosomal permeability in cancer cells as indicated by cathepsin D was markedly higher than that in normal cells and resulted in an obvious decrease in mitochondrial membrane potential. Western blot analysis further confirmed that apoptosis occurred through caspase-9 and caspase-3 activation following mitochondrial damage. Transmission electron microscope images showed that GNRs did not appear in most of the damaged mitochondria but mainly accumulated in lysosomes. CONCLUSION: These findings indicated that GNR-induced apoptosis specifically in cancer cells by affecting lysosomes and mitochondria.


Subject(s)
Apoptosis/drug effects , Gold/chemistry , Gold/therapeutic use , Lysosomes/metabolism , Mitochondria/metabolism , Nanotubes/chemistry , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival , Cricetulus , Humans , Membrane Potential, Mitochondrial , Mice , Particle Size
19.
Sci Rep ; 5: 15712, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26508306

ABSTRACT

Simultaneously blocking multiple mediators offers new hope for the treatment of complex diseases. However, the curative potential of current combination therapy by chronological administration of separate monoclonal antibodies (mAbs) or multi-specific mAbs is still moderate due to inconvenient manipulation, low cooperative effectors, poor pharmacokinetics and insufficient tumor accumulation. Here, we describe a facile strategy that arms distinct mAbs with cooperative effectors onto a long chain to form a multicomponent comb-like nano mAb. Unlike dissociative parental mAbs, the multifunctional mAb nanoarray (PL-RB) constructed from type I/II anti-CD20 mAbs shows good pharmacokinetics. This PL-RB simultaneously targets distinct epitopes on a single antigen (Ag) and neighboring Ags on different lymphocytes. This unique intra- and intercellular Ag cross-linking endows the multifunctional mAb nanoarray with potent apoptosis activity. The exceptional apoptosis, complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC) that are synchronously evoked by the nano PL-RB are further synergistically promoted via enhanced permeability and retention (EPR), which resulted in high intratumor accumulation and excellent anti-lymphoma efficiency.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Cytotoxicity, Immunologic/immunology , Lymphocytes/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD20/immunology , Apoptosis/drug effects , Apoptosis/immunology , Cytotoxicity, Immunologic/drug effects , Drug Synergism , Epitopes/immunology , Epitopes/pharmacology , Female , Humans , Lymphocytes/drug effects , Lymphoma/drug therapy , Lymphoma/immunology , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Nanotechnology/methods
20.
Oncotarget ; 6(27): 24192-204, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26284588

ABSTRACT

Although the anti-CD20 antibody Rituximab has revolutionized the treatment of Non-Hodgkin Lymphoma (NHL), resistance to treatment still existed. Thus, strategies for suppressing Rituximab-resistant NHLs are urgently needed. Here, an anti-CD20 nanocluster (ACNC) is successfully constructed from its type I and type II mAb (Rituximab and 11B8). These distinct anti-CD20 mAbs are mass grafted to a short chain polymer (polyethylenimine). Compared with parental Rituximab and 11B8, the ACNC had a reduced "off-rate". Importantly, ACNC efficiently inhibited Rituximab-resistant lymphomas in both disseminated and localized human NHL xenograft models. Further results revealed that ACNC is significantly potent in inducing caspase-dependent apoptosis and lysosome-mediated programmed cell death (PCD). This may help explain why ACNC is effective in suppressing rituximab-resistant lymphoma while Rituximab and 11B8 are not. Additionally, ACNC experienced low clearance from peripheral blood and high intratumor accumulation. This improved pharmacokinetics is attributed to the antibody-antigen reaction (active targeting) and enhanced permeability and retention (ERP) effect (passive targeting). This study suggested that ACNC might be a promising therapeutic agent for treatment of rituximab-resistant lymphomas.


Subject(s)
Antigens, CD20/chemistry , Antineoplastic Agents/chemistry , Lymphoma, B-Cell/drug therapy , Rituximab/chemistry , Animals , Antibodies, Monoclonal/chemistry , Apoptosis , Caspases/metabolism , Cell Adhesion , Cell Line, Tumor , Enzyme Activation , Female , Humans , Lymphoma, B-Cell/immunology , Lymphoma, Non-Hodgkin/immunology , Lysosomes/metabolism , Membrane Potential, Mitochondrial , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Microscopy, Confocal , Nanoparticles/chemistry , Neoplasm Transplantation , Polyethyleneimine/chemistry , Polymers/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...