Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 438
1.
Int. j. morphol ; 42(1): 127-136, feb. 2024. ilus
Article En | LILACS | ID: biblio-1528822

SUMMARY: The objective of this study was to investigate the therapeutic wound healing potential and molecular mechanisms of shikonin as small molecules in vitro. A mouse burn model was used to explore the potential therapeutic effect of shikonin; we traced proliferating cells in vivo to locate the active area of skin cell proliferation. Through the results of conventional pathological staining, we found that shikonin has a good effect on the treatment of burned skin and promoted the normal distribution of skin keratin at the damaged site. At the same time, shikonin also promoted the proliferation of skin cells at the damaged site; importantly, we found a significant increase in the number of fibroblasts at the damaged site treated with shikonin. Most importantly, shikonin promotes fibroblasts to repair skin wounds by regulating the PI3K/AKT signaling pathway. This study shows that shikonin can effectively promote the proliferation of skin cell, and local injection of fibroblasts in burned skin can play a certain therapeutic role.


El objetivo de este trabajo fue investigar el potencial terapéutico de cicatrización de heridas y los mecanismos moleculares de la shikonina como moléculas pequeñas in vitro. Se utilizó un modelo de quemaduras en ratones para explorar el posible efecto terapéutico de la shikonina; Rastreamos las células en proliferación in vivo para localizar el área activa de proliferación de células de la piel. A través de los resultados de la tinción para patología convencional, encontramos que la shikonina tiene un buen efecto en el tratamiento de la piel quemada y promueve la distribución normal de la queratina de la piel en el sitio dañado. Al mismo tiempo, la shikonina también promovió la proliferación de células de la piel en el sitio dañado. Es importante destacar que encontramos un aumento significativo en la cantidad de fibroblastos en el sitio dañado tratado con shikonina. Lo más importante es que la shikonina promueve la función reparadora de fibroblastos en las heridas de la piel regulando la vía de señalización PI3K/ AKT. Este estudio muestra que la shikonina puede promover eficazmente la proliferación de células de la piel y que la inyección local de fibroblastos en la piel quemada puede desempeñar un cierto papel terapéutico.


Animals , Mice , Wound Healing/drug effects , Burns/drug therapy , Naphthoquinones/administration & dosage , Skin , In Vitro Techniques , Naphthoquinones/pharmacology , Phosphatidylinositol 3-Kinases , Cell Proliferation/drug effects , Disease Models, Animal , Proto-Oncogene Proteins c-akt , Fibroblasts , Mice, Inbred C57BL
2.
ACS Biomater Sci Eng ; 9(9): 5405-5417, 2023 09 11.
Article En | MEDLINE | ID: mdl-37638660

Microwave (MW) thermal therapy has been widely used for the treatment of cancer in clinics, but it still shows limited efficacy and a high recurrence rate owing to non-selective heat delivery and thermo-resistance. Regulating glycolysis shows great promise to improve MW thermal therapy since glycolysis plays an important role in thermo-resistance, progression, metabolism, and recurrence. Herein, we developed a delivery nanosystem of shikonin (SK)-loaded and hyaluronic acid (HA)-modified hollow Fe-MOF (HFM), HFM@SK@HA, as an efficient glycolysis-meditated agent to improve the efficacy of MW thermal therapy. The HFM@SK@HA nanosystem shows a high SK loading capacity of 31.7 wt %. The loaded SK can be effectively released from the HFM@SK@HA under the stimulation of an acidic tumor microenvironment and MW irradiation, overcoming the intrinsically low solubility and severe toxicity of SK. We also find that the HFM@SK@HA can not only greatly improve the heating effect of MW in the tumor site but also mediate MW-enhancing dynamic therapy efficiency by catalyzing the endogenous H2O2 to generate reactive oxygen species (ROS). As such, the MW irradiation treatment in the presence of HFM@SK@HA in vitro enables a highly improved anti-tumor efficacy due to the combined effect of released SK and generated ROS on inhibiting glycolysis in cancer cells. Our in vivo experiments show that the tumor inhibition rate is up to 94.75% ± 3.63% with no obvious recurrence during the 2 weeks after treatment. This work provides a new strategy for improving the efficacy of MW thermal therapy.


Iron , Metal Nanoparticles , Metal-Organic Frameworks , Naphthoquinones , Neoplasms , Metal-Organic Frameworks/chemistry , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Neoplasms/therapy , Iron/chemistry , Naphthoquinones/administration & dosage , Naphthoquinones/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Microwaves/therapeutic use , Warburg Effect, Oncologic/drug effects , Hep G2 Cells , Cell Line, Tumor , L Cells , Female , Animals , Mice , Humans
3.
Transplant Proc ; 55(7): 1731-1738, 2023 Sep.
Article En | MEDLINE | ID: mdl-37391330

BACKGROUND: In diabetes mellitus, diabetic nephropathy (DN) is a typical complication and pivotal cause of chronic kidney disease. The DN disease burden is among the highest in the world and is associated with high morbidity, mortality, and disease burden. Safe and effective medications are urgently needed for the treatment of DN. Interest has been increasing in Shikonin, extracted from the naphthoquinone plant, particularly in determining its renal protective effect. METHODS: In this study, we explored Shikonin's effects and potential mechanisms on a streptozotocin (STZ)-induced DN experimental model. An STZ-induced rat diabetic model was established, and the rats were treated with different doses of Shikonin (10/50 mg/kg) for 4 weeks. Blood, urine, and renal tissue samples were collected after the last administration. Renal tissues were examined to detect each group's physiologic, biochemical, histopathologic, and molecular changes. RESULTS: The results showed that Shikonin administration could significantly alleviate the STZ-induced elevation of blood urea nitrogen, serum creatinine, urinary protein content, and renal pathologic injury. Furthermore, Shikonin significantly decreased oxidative stress, inflammation, and Toll-like receptor 4/myeloid differentiation primary response 88/nuclear factor-κB expression levels in DN kidney tissues. Shikonin showed a dose-dependent effect, with the best outcome at 50 mg/kg. CONCLUSION: Shikonin could effectively alleviate DN-related nephropathy damage and reveal the underlying pharmacologic mechanism. Based on the results, a Shikonin combination can be used in clinical treatment.


Anti-Inflammatory Agents, Non-Steroidal , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Naphthoquinones , Animals , Rats , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/etiology , Naphthoquinones/administration & dosage , Naphthoquinones/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Diabetes Mellitus, Experimental/complications , Streptozocin , Blood Urea Nitrogen , Albuminuria , Oxidative Stress/drug effects , Male , Rats, Sprague-Dawley
4.
Carbohydr Polym ; 282: 119087, 2022 Apr 15.
Article En | MEDLINE | ID: mdl-35123755

The efficient triggering of prodrug release has become a challengeable task for stimuli-responsive nanomedicine utilized in cancer therapy due to the subtle differences between normal and tumor tissues and heterogeneity. In this work, a dual ROS-responsive nanocarriers with the ability to self-regulate the ROS level was constructed, which could gradually respond to the endogenous ROS to achieve effective, hierarchical and specific drug release in cancer cells. In brief, DOX was conjugated with MSNs via thioketal bonds and loaded with ß-Lapachone. TPP modified chitosan was then coated to fabricate nanocarriers for mitochondria-specific delivery. The resultant nanocarriers respond to the endogenous ROS and release Lap specifically in cancer cells. Subsequently, the released Lap self-regulated the ROS level, resulting in the specific DOX release and mitochondrial damage in situ, enhancing synergistic oxidation-chemotherapy. The tumor inhibition Ratio was achieved to 78.49%. The multi-functional platform provides a novel remote drug delivery system in vivo.


Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Naphthoquinones/administration & dosage , Neoplasms/drug therapy , Oxidative Stress , Prodrugs/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Chitosan/administration & dosage , Chitosan/chemistry , Chitosan/pharmacokinetics , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Liberation , Female , Humans , Mice, Inbred BALB C , Mitochondria/physiology , Nanoparticles/chemistry , Naphthoquinones/chemistry , Naphthoquinones/pharmacokinetics , Neoplasms/metabolism , Neoplasms/pathology , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Oxidation-Reduction , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Reactive Oxygen Species/metabolism , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacokinetics , Tumor Burden/drug effects
5.
J Cancer Res Clin Oncol ; 148(3): 633-645, 2022 Mar.
Article En | MEDLINE | ID: mdl-34860309

PURPOSE: Liposarcoma (LPS) represent the largest group of malignant soft tissue tumours comprising a heterogeneous group of subtypes in which the degrees of chemoresistance and radiosensitivity strongly vary. Consequently, it is of utmost interest to establish novel therapeutic regimens based on molecular targets. METHODS: Immunohistochemical staining of survivin was performed in tissue microarrays comprising 49 primary LPS specimens. LPS cell lines were treated with survivin antagonist YM155 and doxorubicin or etoposide alone as well as in combination. Changes in cell viability were investigated and the synergistic effect of a combined therapy analysed. RESULTS: Immunohistochemistry revealed an abundant expression of survivin in LPS that significantly concurred with less-differentiated tumour subtypes and grading. In vitro, we demonstrated the impact of the survivin inhibitor YM155 on dedifferentiated LPS (DDLPS) and, even more imposing, pleomorphic LPS (PLS) tumour cell viability with a strong induction of apoptosis. A combined treatment of doxorubicin or etoposide with YM155 augmented the cytotoxic effects on DDLPS and PLS cells. CONCLUSION: These findings support the significant role of survivin in the oncogenesis and progression of LPS subtypes providing a rationale to target survivin in eligible in-vivo models and to pioneer clinical applications of survivin-specific substances unfolding their therapeutic potential in LPS patients prospectively.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Liposarcoma/classification , Liposarcoma/drug therapy , Survivin/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Doxorubicin/administration & dosage , Etoposide/administration & dosage , Female , Follow-Up Studies , Humans , Imidazoles/administration & dosage , Liposarcoma/metabolism , Liposarcoma/pathology , Male , Middle Aged , Naphthoquinones/administration & dosage , Prognosis , Retrospective Studies , Survival Rate , Tumor Cells, Cultured
6.
Mar Drugs ; 19(11)2021 Nov 01.
Article En | MEDLINE | ID: mdl-34822493

Atopic dermatitis (AD) is a chronic inflammatory skin disease in which skin barrier dysfunction leads to dryness, pruritus, and erythematous lesions. AD is triggered by immune imbalance and oxidative stress. Echinochrome A (Ech A), a natural pigment isolated from sea urchins, exerts antioxidant and beneficial effects in various inflammatory disease models. In the present study, we tested whether Ech A treatment alleviated AD-like skin lesions. We examined the anti-inflammatory effect of Ech A on 2,4-dinitrochlorobenzene (DNCB)-induced AD-like lesions in an NC/Nga mouse model. AD-like skin symptoms were induced by treatment with 1% DNCB for 1 week and 0.4% DNCB for 5 weeks in NC/Nga mice. The results showed that Ech A alleviated AD clinical symptoms, such as edema, erythema, and dryness. Treatment with Ech A induced the recovery of epidermis skin lesions as observed histologically. Tewameter® and Corneometer® measurements indicated that Ech A treatment reduced transepidermal water loss and improved stratum corneum hydration, respectively. Ech A treatment also inhibited inflammatory-response-induced mast cell infiltration in AD-like skin lesions and suppressed the expression of proinflammatory cytokines, such as interferon-γ, interleukin-4, and interleukin-13. Collectively, these results suggest that Ech A may be beneficial for treating AD owing to its anti-inflammatory effects.


Anti-Inflammatory Agents/pharmacology , Naphthoquinones/pharmacology , Sea Urchins , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Aquatic Organisms , Dermatitis, Atopic/drug therapy , Disease Models, Animal , Interleukin-3/metabolism , Male , Mice , Mice, Inbred Strains , Naphthoquinones/administration & dosage , Naphthoquinones/chemistry , Skin/drug effects , Water Loss, Insensible/drug effects
7.
Mar Drugs ; 19(10)2021 Sep 28.
Article En | MEDLINE | ID: mdl-34677449

Echinochrome A (Ech A, 7-ethyl-2,3,5,6,8-pentahydroxy-1,4-naphthoquinone) has been known to exhibit anti-oxidative and anti-inflammatory effects. However, no study has been carried out on the efficacy of Ech A against skin photoaging; this process is largely mediated by oxidative stress. Six-week-old male SKH-1 hairless mice (n = 36) were divided into five groups. Except for a group that were not treated (n = 4), all mice underwent ultraviolet-B (UVB) exposure for 8 weeks while applying phosphate-buffered saline or Ech A through intraperitoneal injection. UVB impaired skin barrier function, showing increased transepidermal water loss and decreased stratum corneum hydration. UVB induced dermal collagen degeneration and mast cell infiltration. Ech A injection was found to significantly lower transepidermal water loss while attenuating tissue inflammatory changes and collagen degeneration compared to the control. Furthermore, Ech A was found to decrease the relative expression of matrix metalloproteinase, tryptase, and chymase. Taken together, these results suggest that Ech A protects against UVB-induced photoaging in both functional and histologic aspects, causing a lowering of collagen degradation and inflammatory cell infiltration.


Collagen/metabolism , Naphthoquinones/pharmacology , Protective Agents/pharmacology , Skin Aging/drug effects , Animals , Aquatic Organisms , Disease Models, Animal , Male , Mast Cells/drug effects , Mice , Mice, Hairless , Naphthoquinones/administration & dosage , Protective Agents/administration & dosage , Ultraviolet Rays , Water Loss, Insensible/drug effects
8.
PLoS Negl Trop Dis ; 15(6): e0009533, 2021 06.
Article En | MEDLINE | ID: mdl-34185780

Visceral leishmaniasis is a vector-borne protozoan infection that is fatal if untreated. There is no vaccination against the disease, and the current chemotherapeutic agents are ineffective due to increased resistance and severe side effects. Buparvaquone is a potential drug against the leishmaniases, but it is highly hydrophobic resulting in poor bioavailability and low therapeutic efficacy. Herein, we loaded the drug into silicon nanoparticles produced from barley husk, which is an agricultural residue and widely available. The buparvaquone-loaded nanoparticles were several times more selective to kill the intracellular parasites being non-toxic to macrophages compared to the pure buparvaquone and other conventionally used anti-leishmanial agents. Furthermore, the in vivo results revealed that the intraperitoneally injected buparvaquone-loaded nanoparticles suppressed the parasite burden close to 100%. By contrast, pure buparvaquone suppressed the burden only by 50% with corresponding doses. As the conclusion, the biogenic silicon nanoparticles are promising carriers to significantly improve the therapeutic efficacy and selectivity of buparvaquone against resistant visceral leishmaniasis opening a new avenue for low-cost treatment against this neglected tropical disease threatening especially the poor people in developing nations.


Antiprotozoal Agents/therapeutic use , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Nanoparticles/administration & dosage , Naphthoquinones/therapeutic use , Animals , Antiprotozoal Agents/administration & dosage , Drug Carriers , Female , Hordeum , Injections, Intraperitoneal , Macrophages/drug effects , Mice, Inbred BALB C , Naphthoquinones/administration & dosage , Naphthoquinones/adverse effects , Silicon/chemistry
9.
Nat Commun ; 12(1): 3299, 2021 06 03.
Article En | MEDLINE | ID: mdl-34083537

Bioenergetic perturbations driving neoplastic growth increase the production of reactive oxygen species (ROS), requiring a compensatory increase in ROS scavengers to limit oxidative stress. Intervention strategies that simultaneously induce energetic and oxidative stress therefore have therapeutic potential. Phenformin is a mitochondrial complex I inhibitor that induces bioenergetic stress. We now demonstrate that inflammatory mediators, including IFNγ and polyIC, potentiate the cytotoxicity of phenformin by inducing a parallel increase in oxidative stress through STAT1-dependent mechanisms. Indeed, STAT1 signaling downregulates NQO1, a key ROS scavenger, in many breast cancer models. Moreover, genetic ablation or pharmacological inhibition of NQO1 using ß-lapachone (an NQO1 bioactivatable drug) increases oxidative stress to selectively sensitize breast cancer models, including patient derived xenografts of HER2+ and triple negative disease, to the tumoricidal effects of phenformin. We provide evidence that therapies targeting ROS scavengers increase the anti-neoplastic efficacy of mitochondrial complex I inhibitors in breast cancer.


Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Phenformin/pharmacology , STAT1 Transcription Factor/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Synergism , Electron Transport Complex I/antagonists & inhibitors , Energy Metabolism/drug effects , Female , Glutathione/antagonists & inhibitors , Glutathione/biosynthesis , Humans , Interferon-gamma/administration & dosage , Interferon-gamma/deficiency , Interferon-gamma/metabolism , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/administration & dosage , Oxidative Stress/drug effects , Phenformin/administration & dosage , Poly I-C/administration & dosage , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/agonists , Xenograft Model Antitumor Assays
10.
Clin Pharmacol Drug Dev ; 10(8): 824-839, 2021 08.
Article En | MEDLINE | ID: mdl-34107166

Napabucasin is an orally administered reactive oxygen species generator that is bioactivated by the intracellular antioxidant nicotinamide adenine dinucleotide phosphate:quinone oxidoreductase 1. Napabucasin induces cell death in cancer cells, including cancer stem cells. This phase 1 study (NCT03411122) evaluated napabucasin drug-drug interaction potential for 7 cytochrome P450 (CYP) enzymes and the breast cancer resistance protein transporter/organic anion transporter 3. Healthy volunteers who tolerated napabucasin during period 1 received probe drugs during period 2, and in period 3 received napabucasin (240 mg twice daily; days 1-11) plus a phenotyping cocktail containing omeprazole (CYP2C19), caffeine (CYP1A2), flurbiprofen (CYP2C9), bupropion (CYP2B6), dextromethorphan (CYP2D6), midazolam (CYP3A) (all oral; day 6), intravenous midazolam (day 7), repaglinide (CYP2C8; day 8), and rosuvastatin (breast cancer resistance protein/organic anion transporter 3; day 9). Drug-drug interaction potential was evaluated in 17 of 30 enrolled volunteers. Napabucasin coadministration increased the area under the plasma concentration-time curve from time 0 extrapolated to infinity (geometric mean ratio [90% confidence interval]) of caffeine (124% [109.0%-141.4%]), intravenous midazolam (118% [94.4%-147.3%]), repaglinide (127% [104.7%-153.3%]), and rosuvastatin (213% [42.5%-1068.3%]) and decreased the area under the plasma concentration-time curve from time 0 extrapolated to infinity of dextromethorphan (71% [47.1%-108.3%]), bupropion (79% [64.6%-97.0%]), and hydroxybupropion (45% [15.7%-129.6%]). No serious adverse events/deaths were reported. Generally, napabucasin is not expected to induce/inhibit drug clearance to a clinically meaningful degree.


ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Benzofurans/administration & dosage , Cytochrome P-450 Enzyme System/metabolism , Naphthoquinones/administration & dosage , Neoplasm Proteins/metabolism , Administration, Oral , Adult , Benzofurans/pharmacokinetics , Bupropion/administration & dosage , Bupropion/pharmacokinetics , Caffeine/administration & dosage , Caffeine/pharmacokinetics , Dextromethorphan/administration & dosage , Dextromethorphan/pharmacokinetics , Drug Interactions , Female , Flurbiprofen/administration & dosage , Flurbiprofen/pharmacokinetics , Gene Expression Regulation/drug effects , Half-Life , Healthy Volunteers , Humans , Male , Midazolam/administration & dosage , Midazolam/pharmacokinetics , Naphthoquinones/pharmacokinetics , Omeprazole/administration & dosage , Omeprazole/pharmacokinetics , Rosuvastatin Calcium/administration & dosage , Rosuvastatin Calcium/pharmacokinetics , Young Adult
11.
J Ethnopharmacol ; 277: 114184, 2021 Sep 15.
Article En | MEDLINE | ID: mdl-33961996

ETHNOPHARMACOLOGICAL RELEVANCE: Rubia yunnanensis Diels is a traditional Chinese medicine that has diverse pharmacological activities, including antituberculosis, antirheumatism and anticancers. Rubioncolin C (RC), a natural naphthohydroquinone dimer isolated from the roots and rhizomes of R. yunnanensis Diels, has shown potent antitumor activity. However, the antitumor activity and its potential mechanism of RC in triple-negative breast cancer (TNBC) cell lines remained unclear. AIM OF THE STUDY: This study was aim to investigate the anti-proliferation and anti-metastasis activity as well as the potential mechanism of RC on triple-negative breast cancer cells in vitro and in vivo. MATERIALS AND METHODS: The sulforhodamine B assay, colony formation assay and cell cycle analysis were used to determine the anti-proliferative activity of RC on TNBC. The anti-metastatic activity in vitro of RC was detected through the scratch wound assay, cell migration and invasion assays and gelatin zymography. The flow cytometry, JC-1, GFP-LC3B plasmid transfection, MDC, Lysotracker red and Carboxy-H2DCFDA, DHE, and MitoSOX™ Red staining were performed to investigate the effect of RC on apoptosis, autophagy and ROS level. The apoptosis inhibitor, autophagy inhibitors and ROS inhibitors were used to further verify the antitumor mechanism of RC. The protein levels related with cell cycle, apoptosis, and autophagy were examined with western blotting. In addition, the anti-tumor activity of RC in vivo was assessed in an experimental metastatic model. RESULTS: In the present study, RC suppressed the proliferation of TNBC cells in a time- and dose-dependent manner via regulating cell cycle. Further experiments showed that RC inhibited the migration and invasion of TNBC cells by downregulating MMPs and inhibiting EMT. Moreover, we demonstrated that RC induced obviously apoptotic and autophagic cell death, activated MAPK signaling pathway and inhibited mTOR/Akt/p70S6K and NF-κB signaling pathways. Furthermore, the excessive ROS was produced after treatment with RC. The antioxygen NAC and GSH could rescue the cell viability and reestablish the ability of cell metastasis, and inhibit the RC-induced apoptosis and autophagy. In a mice lung metastasis model of breast cancer, RC inhibited lung metastasis, and induced autophagy and apoptosis. CONCLUSION: These findings clarified the antitumor mechanism of RC on TNBC cell lines and suggested that RC is a key active ingredient for the cancer treatment of R. yunnanensis, which would help RC develop as a new potential chemotherapeutic agent for TNBC treatment.


Antineoplastic Agents, Phytogenic/pharmacology , Naphthoquinones/pharmacology , Rubia/chemistry , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/isolation & purification , Autophagy/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred BALB C , Naphthoquinones/administration & dosage , Naphthoquinones/isolation & purification , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Reactive Oxygen Species/metabolism , Time Factors
12.
Cancer Sci ; 112(9): 3507-3519, 2021 Sep.
Article En | MEDLINE | ID: mdl-34050696

Necroptosis is an alternative form of programmed cell death that generally occurs under apoptosis-deficient conditions. Our previous work showed that connexin32 (Cx32) promotes the malignant progress of hepatocellular carcinoma (HCC) by enhancing the ability of resisting apoptosis in vivo and in vitro. Whether triggering necroptosis is a promising strategy to eliminate the apoptosis-resistant HCC cells with high Cx32 expression remains unknown. In this study, we found that Cx32 expression was positively correlated with the expression of necroptosis protein biomarkers in human HCC specimens, cell lines, and a xenograft model. Treatment with shikonin, a well-used necroptosis inducer, markedly caused necroptosis in HCC cells. Interestingly, overexpressed Cx32 exacerbated shikonin-induced necroptosis, but downregulation of Cx32 alleviated necroptosis in vitro and in vivo. Mechanistically, Cx32 was found to bind to Src and promote Src-mediated caspase 8 phosphorylation and inactivation, which ultimately reduced the activated caspase 8-mediated proteolysis of receptor-interacting serine-threonine protein kinase 1/3, the key molecule for necroptosis activation. In conclusion, we showed that Cx32 contributed to the activation of necroptosis in HCC cells through binding to Src and then mediating the inactivation of caspase 8. The present study suggested that necroptosis inducers could be more favorable than apoptosis inducers to eliminate HCC cells with high expression of Cx32.


Carcinoma, Hepatocellular/metabolism , Caspase 8/metabolism , Connexins/metabolism , Liver Neoplasms/metabolism , Necroptosis/genetics , Nuclear Receptor Coactivator 1/metabolism , Signal Transduction/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Connexins/genetics , Gene Knockdown Techniques , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Naphthoquinones/administration & dosage , Necroptosis/drug effects , Nuclear Receptor Coactivator 1/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Signal Transduction/drug effects , Transfection , Tumor Burden/drug effects , Tumor Burden/genetics , Gap Junction beta-1 Protein
13.
Int J Nanomedicine ; 16: 2615-2631, 2021.
Article En | MEDLINE | ID: mdl-33854311

BACKGROUND: Plumbagin, a naphthoquinone extracted from the officinal leadwort presenting promising anti-cancer properties, has its therapeutic potential limited by its inability to reach tumors in a specific way at a therapeutic concentration following systemic injection. The purpose of this study is to assess whether a novel tumor-targeted, lipid-polymer hybrid nanoparticle formulation of plumbagin would suppress the growth of B16-F10 melanoma in vitro and in vivo. METHODS: Novel lipid-polymer hybrid nanoparticles entrapping plumbagin and conjugated with transferrin, whose receptors are present in abundance on many cancer cells, have been developed. Their cellular uptake, anti-proliferative and apoptosis efficacy were assessed on various cancer cell lines in vitro. Their therapeutic efficacy was evaluated in vivo after tail vein injection to mice bearing B16-F10 melanoma tumors. RESULTS: The transferrin-bearing lipid-polymer hybrid nanoparticles loaded with plumbagin resulted in the disappearance of 40% of B16-F10 tumors and regression of 10% of the tumors following intravenous administration. They were well tolerated by the mice. CONCLUSION: These therapeutic effects, therefore, make transferrin-bearing lipid-polymer hybrid nanoparticles entrapping plumbagin a highly promising anti-cancer nanomedicine.


Lipids/chemistry , Melanoma, Experimental/drug therapy , Nanoparticles/chemistry , Naphthoquinones/administration & dosage , Naphthoquinones/therapeutic use , Polymers/chemistry , Skin Neoplasms/drug therapy , Transferrin/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coumarins/pharmacology , Coumarins/therapeutic use , Drug Liberation , Endocytosis/drug effects , Female , Humans , Injections, Intravenous , Melanoma, Experimental/pathology , Mice, Inbred BALB C , Naphthoquinones/pharmacology , Skin Neoplasms/pathology , Thiazoles/pharmacology , Thiazoles/therapeutic use
14.
Brief Bioinform ; 22(3)2021 05 20.
Article En | MEDLINE | ID: mdl-32789496

TP53 mutation is one of the most common genetic changes in hepatocellular carcinoma (HCC). It is of great clinical significance to tailor specialized prognostication approach and to explore more therapeutic options for TP53-mutant HCCs. In this study, a total of 1135 HCC patients were retrospectively analyzed. We developed a random forest-based prediction model to estimate TP53 mutational status, tackling the problem of limited sample size in TP53-mutant HCCs. A multi-step process was performed to develop robust poor prognosis-associated signature (PPS). Compared with previous established population-based signatures, PPS manifested superior ability to predict survival in TP53-mutant patients. After in silico screening of 2249 drug targets and 1770 compounds, we found that three targets (CANT1, CBFB and PKM) and two agents (irinotecan and YM-155) might have potential therapeutic implications in high-PPS patients. The results of drug targets prediction and compounds prediction complemented each other, presenting a comprehensive view of potential treatment strategy. Overall, our study has not only provided new insights into personalized prognostication approaches, but also thrown light on integrating tailored risk stratification with precision therapy.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Hepatocellular , Liver Neoplasms , Mutation , Precision Medicine , Tumor Suppressor Protein p53 , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Computer Simulation , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/administration & dosage , Irinotecan/administration & dosage , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Male , Naphthoquinones/administration & dosage , Survival Rate , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Microbiol Res ; 244: 126666, 2021 Mar.
Article En | MEDLINE | ID: mdl-33338970

ß-lactamase inhibitors are potent synergistic drugs to deteriorate the multidrug-resistant bacteria. Here, we report the ß-lactamase inhibitory ability of kalafungin isolated from a marine sponge derived Streptomyces sp. SBRK1. The IC50 value of the kalafungin was calculated as 225.37 ± 1.95 µM against ß-lactamase. The enzyme kinetic analysis showed the Km value of 3.448 ± 0.7 µM and Vmax value of 215.356 ± 8 µM/min and the inhibition mechanism was identified as uncompetitive type. Along with the antibacterial activity, the cell surface analysis of kalafungin treated Staphylococcus aureus cells revealed destruction of cell membrane in response to ß-lactamase inhibition. Molecular docking studies have confirmed the binding property of kalafungin against ß-lactamase with two hydrogen bonds. In vivo efficacy studies in the zebrafish model by green fluorescent protein expressing S. aureus infection, survival, safety and behavioral profile were reported. The toxicity and anti-infection revealed that the compound was evidently active and safe to all organs. In conclusion, this is the first report on kalafungin with ß- lactamase inhibition and suggests that kalafungin may useful for synergic antibacterial therapy with ß-lactam drugs to overcome ß-lactamase-based resistance of any bacterial pathogens.


Anti-Bacterial Agents/administration & dosage , Bacterial Proteins/metabolism , Staphylococcal Infections/drug therapy , Staphylococcus aureus/physiology , Streptomyces/chemistry , beta-Lactamase Inhibitors/administration & dosage , Animals , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Disease Models, Animal , Humans , Kinetics , Molecular Docking Simulation , Naphthoquinones/administration & dosage , Naphthoquinones/chemistry , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Streptomyces/genetics , Streptomyces/isolation & purification , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/chemistry , beta-Lactamases/metabolism
16.
Pharmacol Res Perspect ; 9(1): e00691, 2021 02.
Article En | MEDLINE | ID: mdl-33378565

Coronaviruses represent global health threat. In this century, they have already caused two epidemics and one serious pandemic. Although, at present, there are no approved drugs and therapies for the treatment and prevention of human coronaviruses, several agents, FDA-approved, and preclinical, have shown in vitro and/or in vivo antiviral activity. An in-depth analysis of the current situation leads to the identification of several potential drugs that could have an impact on the fight against coronaviruses infections. In this review, we discuss the virology of human coronaviruses highlighting the main biological targets and summarize the current state-of-the-art of possible therapeutic options to inhibit coronaviruses infections. We mostly focus on FDA-approved and preclinical drugs targeting viral conserved elements.


Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , Coronavirus Infections/metabolism , Coronavirus/metabolism , Dipeptidyl Peptidase 4/metabolism , Severe Acute Respiratory Syndrome/metabolism , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antiviral Agents/administration & dosage , Antiviral Agents/metabolism , Azoles/administration & dosage , Azoles/metabolism , Coronavirus/drug effects , Coronavirus Infections/drug therapy , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/metabolism , Humans , Isoindoles , Naphthoquinones/administration & dosage , Naphthoquinones/metabolism , Organoselenium Compounds/administration & dosage , Organoselenium Compounds/metabolism , Severe Acute Respiratory Syndrome/drug therapy , COVID-19 Drug Treatment
17.
Cancer Res ; 81(4): 873-884, 2021 02 15.
Article En | MEDLINE | ID: mdl-33355239

The role and significance of liver-derived cytokines in cancer-associated cachexia syndrome remain elusive. Here we report that combinatorial counterbalances of the leptin and Igf1 signaling pathways in hepatocellular carcinoma (HCC) models significantly relieves cachexia. Double transgenic zebrafish models of HCC that stably displayed focal lesions, anorexia, and wasting of adipose and muscle tissues were first generated. Knockout of lepr or mc4r from these zebrafish partially restored appetite and exerted moderate or no effect on tissue wasting. However, genetic replenishment of Igf1 in a lepr-mutant background effectively relieved the cachexia-like phenotype without affecting tumor growth. Similarly, administration of napabucasin, a Stat3/Socs3 inhibitor, on the zebrafish HCC model, mammalian cell lines with exogenous IGF1, and two mouse xenograft models restored insulin sensitivity and rescued the wasting of nontumor tissues. Together, these results describe the synergistic impact of leptin and Igf1 normalization in treating certain HCC-associated cachexia as a practical strategy. SIGNIFICANCE: Disruption of leptin signaling with normalized Igf1 expression significantly rescues anorexia, muscle wasting, and adipose wasting in Ras- and Myc-driven zebrafish models of HCC.


Benzofurans/administration & dosage , Cachexia/prevention & control , Carcinoma, Hepatocellular/drug therapy , Insulin-Like Growth Factor I/administration & dosage , Liver Neoplasms/drug therapy , Liver/drug effects , Naphthoquinones/administration & dosage , 3T3-L1 Cells , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Animals, Genetically Modified , Benzofurans/pharmacology , Cachexia/genetics , Cachexia/metabolism , Cachexia/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cells, Cultured , Cytokines/metabolism , Cytokines/physiology , Disease Models, Animal , Drug Synergism , HEK293 Cells , Hep G2 Cells , Humans , Insulin Resistance/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/therapeutic use , Leptin/genetics , Leptin/metabolism , Liver/metabolism , Liver/physiology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Naphthoquinones/pharmacology , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Wasting Syndrome/genetics , Wasting Syndrome/metabolism , Wasting Syndrome/pathology , Wasting Syndrome/prevention & control , Xenograft Model Antitumor Assays , Zebrafish
18.
Microb Drug Resist ; 27(7): 924-932, 2021 Jul.
Article En | MEDLINE | ID: mdl-33275860

Background: The treatment of multidrug-resistant tuberculosis (MDR-TB) is a challenge to be overcome. The increase of resistant isolates associated with serious side effects during therapy leads to the search for substances that have anti-TB activity, which make treatment less toxic, and also act in the macrophage acidic environment promoted by the infection. Objective: The aim of this study was to investigate lapachol and ß-lapachone activities in combination with other drugs against Mycobacterium tuberculosis at neutral and acidic pH and its cytotoxicity. Design: Inhibitory and bactericidal activities against M. tuberculosis and clinical isolates were determined. Drug combination and cytotoxicity assay were carried out using standard TB drugs and/or N-acetylcysteine (NAC). Results: Both naphthoquinones presented activity against MDR clinical isolates. The combinations with the first-line TB drugs demonstrated an additive effect and ß-lapachone+NAC were synergic against H37Rv. Lapachol activity at acidic pH and its association with NAC improved the selectivity index. Lapachol and ß-lapachone produced cell morphological changes in bacilli at pH 6.0 and 6.8, respectively. Conclusion: Lapachol revealed promising anti-TB activity, especially associated with NAC.


Antitubercular Agents/pharmacology , Naphthoquinones/pharmacology , Tuberculosis, Multidrug-Resistant/drug therapy , Antitubercular Agents/administration & dosage , Cell Survival , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Humans , Hydrogen-Ion Concentration , Macrophages/drug effects , Microbial Sensitivity Tests , Naphthoquinones/administration & dosage
19.
Food Funct ; 11(11): 10219-10230, 2020 Nov 18.
Article En | MEDLINE | ID: mdl-33169745

Osteoarthritis is a chronic degenerative disease characterized by cartilage destruction. It is the fourth most disabling disease worldwide and is currently incurable. Inflammation and extracellular matrix (ECM) degradation are considered to be substantial reasons for accelerating the progression of OA. ß-Hydroxyisoamylshikonin (ß-HIVS) is a natural naphthoquinone compound with anti-inflammatory and antioxidant activity. However, the effect of ß-HIVS on OA is still unclear. In this study, we found that ß-HIVS can down-regulate the expression of NO, PEG2, IL-6, TNF-α, COX-2, and iNOS, suggesting its anti-inflammatory effects in chondrocytes; we also found that ß-HIVS may down-regulate the expression of ADAMTS5 and MMP13 and up-regulate the expression of aggrecan and collagen II to inhibit the degradation of ECM. Mechanistically, ß-HIVS inhibited the NFκB pathway by activating the Nrf2/HO-1 axis, thereby exerting its anti-inflammatory and inhibitory effects on ECM degradation. In vivo experiments also proved the therapeutic effects of ß-HIVS on OA in mice, and Nrf2 is the target of ß-HIVS. These findings indicate that ß-HIVS may become a new drug for the treatment of OA.


Anti-Inflammatory Agents/administration & dosage , Chondrocytes/drug effects , Interleukin-1beta/immunology , NF-E2-Related Factor 2/immunology , Naphthoquinones/administration & dosage , Osteoarthritis/drug therapy , Animals , Chondrocytes/immunology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , Humans , Interleukin-1beta/genetics , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/immunology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , NF-kappa B/immunology , Osteoarthritis/genetics , Osteoarthritis/immunology
20.
Int J Pharm ; 588: 119734, 2020 Oct 15.
Article En | MEDLINE | ID: mdl-32777535

Leishmaniasis is a neglected disease presenting cutaneous, mucosal and visceral forms and affecting an estimated 12 million mostly low-income people. Treatment of cutaneous leishmaniasis (CL) is recommended to expedite healing, reduce risk of scarring, prevent parasite dissemination to other mucocutaneous (common with New World species) or visceral forms and reduce the chance of relapse, but remains an unmet need. Available treatments are painful, prolonged (>20 days) and require hospitalisation, which increases the cost of therapy. Here we present the development of optimised topical self-nanoemulsifying drug delivery systems (SNEDDS) loaded with buparvaquone (BPQ, a hydroxynapthoquinone from the open Malaria Box) for the treatment of CL from New World species. The administration of topical BPQ-SNEDDS gels for 7 days resulted in a reduction of parasite load of 99.989 ± 0.019% similar to the decrease achieved with intralesionally administered Glucantime® (99.873 ± 0.204%) in a L. amazonensis BALB/c model. In vivo efficacy was supported by ex vivo permeability and in vivo tape stripping studies. BPQ-SNEDDS and their hydrogels demonstrated linear flux across non-infected CD-1 mouse skin ex vivo of 182.4 ± 63.0 µg cm-2 h-1 and 57.6 ± 10.8 µg cm-2 h-1 respectively localising BPQ within the skin in clinically effective concentrations (227.0 ± 45.9 µg and 103.8 ± 33.8 µg) respectively. These levels are therapeutic as BPQ-SNEDDS and their gels showed nanomolar in vitro efficacy against L. amazonensis and L. braziliensis amastigotes with excellent selectivity index toward parasites versus murine macrophages. In vivo tape stripping experiments indicated localisation of BPQ within the stratum corneum and dermis. Histology studies confirmed the reduction of parasitism and indicated healing in animals treated with BPQ-SNEDDS hydrogels. These results highlight the potential clinical capability of nano-enabled BPQ hydrogels towards a non-invasive treatment for CL.


Antiprotozoal Agents , Leishmaniasis, Cutaneous , Naphthoquinones , Animals , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/therapeutic use , Hydrogels/therapeutic use , Leishmaniasis, Cutaneous/drug therapy , Mice , Mice, Inbred BALB C , Naphthoquinones/administration & dosage
...