Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 18: 2891-2910, 2023.
Article in English | MEDLINE | ID: mdl-37283712

ABSTRACT

Introduction: Hepatocellular cancer stem cells (CSCs) play crucial roles in hepatocellular cancer initiation, development, relapse, and metastasis. Therefore, eradication of this cell population is a primary objective in hepatocellular cancer therapy. We prepared a nanodrug delivery system with activated carbon nanoparticles (ACNP) as carriers and metformin (MET) as drug (ACNP-MET), which was able to selectively eliminate hepatocellular CSCs and thereby increase the effects of MET on hepatocellular cancers. Methods: ACNP were prepared by ball milling and deposition in distilled water. Suspension of ACNP and MET was mixed and the best ratio of ACNP and MET was determined based on the isothermal adsorption formula. Hepatocellular CSCs were identified as CD133+ cells and cultured in serum-free medium. We investigated the effects of ACNP-MET on hepatocellular CSCs, including the inhibitory effects, the targeting efficiency, self-renewal capacity, and the sphere-forming capacity of hepatocellular CSCs. Next, we evaluated the therapeutic efficacy of ACNP-MET by using in vivo relapsed tumor models of hepatocellular CSCs. Results: The ACNP have a similar size, a regular spherical shape and a smooth surface. The optimal ratio for adsorption was MET: ACNP=1:4. ACNP-MET could target and inhibit the proliferation of CD133+ population and decrease mammosphere formation and renewal of CD133+ population in vitro and in vivo. Conclusion: These results not only suggest that nanodrug delivery system increased the effects of MET, but also shed light on the mechanisms of the therapeutic effects of MET and ACNP-MET on hepatocellular cancers. ACNP, as a good nano-carrier, could strengthen the effect of MET by carrying drugs to the micro-environment of hepatocellular CSCs.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Metformin , Nanoparticles , Humans , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/pathology , Charcoal , Cell Line, Tumor , Metformin/pharmacology , Neoplastic Stem Cells/pathology , Nanoparticles/therapeutic use , AC133 Antigen/metabolism , AC133 Antigen/pharmacology , Tumor Microenvironment
2.
Biomed Environ Sci ; 31(6): 467-472, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30025561

ABSTRACT

There is still a need for better protection against or mitigation of the effects of ionizing radiation following conventional radiotherapy or accidental exposure. The objective of our current study was to investigate the possible roles of matrix metalloproteinase inhibitor, ilomastat, in the protection of mice from total body radiation (TBI), and the underlying protective mechanisms. Ilomastat treatment increased the survival of mice after TBI. Ilomastat pretreatment promoted recovery of hematological and immunological cells in mice after 6 Gy γ-ray TBI. Our findings suggest the potential of ilomastat to protect against or mitigate the effects of radiation.


Subject(s)
Acute Radiation Syndrome/prevention & control , Gamma Rays/adverse effects , Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , Matrix Metalloproteinase Inhibitors/therapeutic use , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/therapeutic use , Acute Radiation Syndrome/blood , Acute Radiation Syndrome/immunology , Animals , Blood Cells/drug effects , Blood Cells/radiation effects , Dose-Response Relationship, Drug , Mice , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/immunology , Spleen/drug effects , Spleen/immunology , Spleen/radiation effects , Survival Analysis , Whole-Body Irradiation
3.
Curr Alzheimer Res ; 14(6): 668-679, 2017.
Article in English | MEDLINE | ID: mdl-27978793

ABSTRACT

BACKGROUND: Amyloid peptide precursor (APP) as the precursor protein of peptide betaamyloid (ß-amyloid, Aß), which is thought to play a central role in the pathogenesis of Alzheimer's disease (AD), also has an important effect on the development and progression of AD. Through knocking-in APP gene in animals, numerous transgenic AD models have been set up for the investigation of the mechanisms behind AD pathogenesis and the screening of anti-AD drugs. However, there are some limitations to these models and here is a need for such an AD model that is economic as well as has satisfactory genetic homology with human. METHODS: We generated a new AD transgenic model by knocking a mutant human APP gene (APPsw) in zebrafish with appb promoter of zebrafish to drive the expression of APPsw. RESULTS: Fluorescent image and immunochemistry stain showed and RT-PCR and western blot assay confirmed that APPsw was successfully expressed in the brain, heart, eyes and vasculature of the transgenic zebrafish. Behavioral observation demonstrated that the transgenic zebrafish had AD-like symptoms. Histopathological observation found that there were cerebral ß-amyloidosis and angiopathy (CAA), which induced neuron loss and enlarged pervascular space. CONCLUSION: These results suggest that APPsw transgenic zebrafish well simulate the pathological characters of AD and can be used as an economic AD transgenic model. Furthermore, the new model suggested that APP can express in microvasculatures and cause the Aß generation and deposition in cerebral vessel which further destroys cerebral vascular structure resulting in the development and/or the progress of AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Cerebral Amyloid Angiopathy/etiology , Gene Expression Regulation/genetics , Mutation/genetics , Promoter Regions, Genetic/physiology , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloidosis/etiology , Animals , Animals, Genetically Modified , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Cerebral Amyloid Angiopathy/genetics , Disease Models, Animal , Exploratory Behavior/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Maze Learning/physiology , Microscopy, Electron, Transmission , Microvessels/metabolism , Microvessels/pathology , Microvessels/ultrastructure , RNA, Messenger/metabolism , Zebrafish
4.
Cutan Ocul Toxicol ; 34(1): 1-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24641121

ABSTRACT

CONTEXT: The chemical weapon sulfur mustard (SM) is a blister agent, and currently, there is no effective antidote. OBJECTIVE: To evaluate the decontamination efficacy of potassium ketoxime against SM and preliminarily elucidate its decontamination mechanism. MATERIALS AND METHODS: Potassium ketoxime reacted with SM, and SM residues were tested at different time intervals by T-135 colorimetry after the reaction. Rabbit skin was topically exposed to 2 mg/cm(2) SM, treated with potassium ketoxime 1 min later, and observed after 6, 12, and 24 h. Gas chromatography-mass spectroscopy was employed to screen and identify the main products of potassium ketoxime decontamination of SM. RESULTS: Potassium ketoxime had a great effect against SM contamination. With a mass ratio of decontaminant: SM of 50:1, decontamination rates against SM were 87.5% after 30 s, 95.9% after 1 min, and 99.0% after 5 min. Fifteen minutes after exposure to SM, the untreated group showed clear erythema lesions, whereas the experimental group showed no clear erythema lesions within 6 h. After 12 and 24 h, the areas of damaged skin in the experimental group were 0.038 and 0.125 cm(2), respectively, compared with 2.21 and 2.65 cm(2) in the control group. Histopathological analysis revealed that treatment with potassium ketoxime also reduced inflammation-induced damage. CONCLUSION: The results of this study indicate that potassium ketoxime reacted rapidly and completely with SM, and thus, it was found to be a suitable and effective skin decontaminant against SM. The decontamination reaction mechanism is mainly related to nucleophilic substitution.


Subject(s)
Chemical Warfare Agents/toxicity , Decontamination/methods , Mustard Gas/toxicity , Oximes/therapeutic use , Skin Diseases/drug therapy , Animals , Chemical Warfare Agents/analysis , Female , Male , Mustard Gas/analysis , Rabbits , Skin/chemistry , Skin/drug effects , Skin/pathology , Skin Diseases/chemically induced , Skin Diseases/pathology
5.
Toxicol Appl Pharmacol ; 259(1): 133-42, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22227104

ABSTRACT

Bisphenol (BPA) is one of the highest-volume chemicals produced worldwide, and human exposure to BPA is thought to be ubiquitous. Various rodent and in vitro studies have shown that thyroid hormone (TH) function can be impaired by BPA. However, it is still unknown if low concentrations of BPA can suppress the thyroid hormone receptor (TR) transcription. The present study aims to investigate the possible suppressing effects of low concentrations of BPA on TR transcription and the involved mechanism(s) in CV-1 cells derived from cercopithecus aethiops monkey kidneys. Using gene reporter assays, BPA at concentrations as low as 10(-9)M suppresses TR or steroid receptor coactivator-1(SRC-1)-enhanced TR transcription, but not reducing TR/SRC-1 interaction in mammalian two-hybrid and glutathione S-transferase pull-down studies. It has been further shown that both nuclear receptor co-repressor (N-CoR) and silencing mediator for retinoid and thyroid hormone receptors (SMRT) are recruited to the TR-ß1 by BPA in the presence of physiologic concentrations of T3 or T4. However, the overexpression of ß3 integrin or c-Src significantly reduces BPA-induced recruitment of N-CoR/SMRT to TR or suppression of TR transcription. Furthermore, BPA inhibits the T3/T4-mediated interassociation of the ß3 integrin/c-Src/MAPK/TR-ß1 pathways by the co-immunoprecipitation. These results indicate that low concentrations of BPA suppress the TR transcription by disrupting physiologic concentrations of T3/T4-mediated ß3 integrin/c-Src/MAPK/TR-ß1 pathways, followed by recruiting N-CoR/SMRT to TR-ß1, providing a novel insight regarding the TH disruption effects of low concentration BPA.


Subject(s)
Environmental Pollutants/toxicity , Phenols/toxicity , Signal Transduction/drug effects , Thyroid Hormone Receptors beta/genetics , Transcription, Genetic/drug effects , Animals , Benzhydryl Compounds , Cell Culture Techniques , Cell Line , Chlorocebus aethiops , Dose-Response Relationship, Drug , Genes, Reporter , Genetic Vectors , Humans , Nuclear Receptor Co-Repressor 2/metabolism , Nuclear Receptor Coactivator 1/metabolism , Plasmids , Thyroid Hormone Receptors beta/metabolism , Thyroxine/pharmacology , Thyroxine/physiology , Transcription, Genetic/physiology , Triiodothyronine/pharmacology , Triiodothyronine/physiology
6.
Yao Xue Xue Bao ; 47(11): 1447-51, 2012 Nov.
Article in Chinese | MEDLINE | ID: mdl-23387075

ABSTRACT

Blood-brain barrier (BBB) is the major obstacle for drug delivery into the central nervous system (CNS). However, there is no ideal model animal for the study of BBB permeability till now. Currently zebrafish (Danio rerio) has emerged as a powerful model organism for the study of vertebrate biology. In this study, the feasibility of using zebrafish as model animal was investigated for BBB permeability by comparing the results of administration of BBB-penetrating peptide and protein to mouse and zebrafish. The results showed that the BBBs of mouse and zebrafish were similar in molecular permeability. Additionally, zebrafish has advantageous features as a model animal, such as small size, fertile and easy to breed. Therefore, it is suggested that zebrafish may be a favored model for the study of BBB permeability.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Glycoproteins/pharmacokinetics , Peptide Fragments/pharmacokinetics , Viral Proteins/pharmacokinetics , Zebrafish/metabolism , Animals , Female , Fluorescent Dyes/pharmacokinetics , Green Fluorescent Proteins/pharmacokinetics , Male , Mice , Models, Animal , Permeability , Rhodamines/pharmacokinetics , Tissue Distribution
7.
Int J Pharm ; 405(1-2): 1-8, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21093564

ABSTRACT

A group of growth factors have been shown to play important roles in amelioration of the malfunction of the neurodegenerative diseases. However, the proteins or polypeptides passing across the blood-brain barrier (BBB) to access the brain parenchyma are relatively few so that it hinders the therapies in clinic. Here a genetically reconstructed fusion peptide of human epidermal growth factor (hEGF) with an undecapeptide YGRKKRRQRRR (P11) was used to investigate the permeability between the cell membrane and the BBB via rectal administration. The efficiency to rescue the Aß 22-35-induced dementia in mice was assessed after administration of P11-hEGF per rectal. Our results showed that P11-hEGF permeates across not only the 3T3 cell membrane in vitro, but also the endothelia of vessels after intravenous injection (IV), and the mucosa of the rectum after per rectal administration. Further results showed that the circulating P11-hEGF allowed penetrating through the blood-brain barrier and then getting into the brain manifesting biological responses. In the animal experiments, treatment with P11-hEGF not only ameliorated the dementia induced by Aß 22-35 but also rescued the dementia of the aged mice, no matter how it was administrated (IV or per rectal). These results suggest that the rectal non-invasive delivery of the P11 polypeptide-conjugated growth factor is an efficient way for BBB transduction, thus raises the hope of real therapeutic progress against neurodegenerative diseases.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Dementia/drug therapy , Epidermal Growth Factor/administration & dosage , Oligopeptides/administration & dosage , Recombinant Fusion Proteins/pharmacology , Administration, Rectal , Animals , Brain/metabolism , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Dementia/metabolism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/pharmacokinetics , Epidermal Growth Factor/pharmacology , Humans , Learning Disabilities/drug therapy , Memory Disorders/drug therapy , Mice , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/pharmacokinetics
8.
Yi Chuan ; 32(1): 73-80, 2010 Jan.
Article in Chinese | MEDLINE | ID: mdl-20085889

ABSTRACT

The expression of Pib gene in rice was induced by hormone, such as jasmonic acid and ethylene. In order to determine the necessary regions of sequence or motifs for response to jasmonic acid and ethylene in Pib promoter, the full length promoter of Pib (-3,572 approximately 2 bp) and three different 5' deletion fragments of Pib promoter (-2,692 approximately 2 bp, -1,335 approximately 2 bp, -761 approximately 2 bp) were synthesized by PCR and then were substituted for 35S upstream gus in a binary plasmid to construct re-combined plasmids of Pib promoter-gus fusions. Transgenic rice plants of the four recombined plasmids were produced by Agrobacterium-mediated transformation. Quality and quantum analysis of gus activities in transgenic plants at both protein and mRNA levels were conducted. The promotion activity of the full length promoter of Pib (-3,572 approximately 2 bp, pNAR901) was the highest in the four recombinants and the gus activities in its transgenic plant organs were enhanced obviously at 6 h after treatment with jasmonic acid or ethylene. The promotion activity of the deleted Pib promoters was significantly decreased and the response to jasmonic acid or ethylene treatment was not present when the -3,572 approximately -2,692 bp sequence was knocked out from the Pib promoter. Although the disparity in the lengths of the deleted Pib promoter of pNAR902 (-2,692 approximately 2 bp), pNAR903 (-1,335 approximately 2 bp), and pNAR904 (-761 approximately 2 bp) was more than 2 or 3 times, the response to jasmonic acid or ethylene treatment was not different among their transgenic plants. All these results indicated that the common deleted sequences (-3,572 approximately -2,692 bp) in the three deleted Pib promoter constructs were the essential region to the response to jasmonic acid and ethylene treatment. The result of pib promoter sequence searching indicated that there was only one GCCGCC motif at -2,722 bp of this common deleted segment in the Pib promoter sequence. Our rice transgenic results showed that the GCCGCC may be a cis-motif for Pib gene conferring response to jasmonic acid and ethylene for Pib gene.


Subject(s)
Carrier Proteins/genetics , Cyclopentanes/metabolism , Ethylenes/metabolism , Gene Expression Regulation, Plant , Oryza/genetics , Oxylipins/metabolism , Plant Proteins/genetics , Promoter Regions, Genetic , Transformation, Genetic , Genes, Reporter , Oryza/metabolism , Phosphate-Binding Proteins
10.
Neurosci Lett ; 463(2): 119-24, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19616075

ABSTRACT

Although there is possibility of cognitive disturbance in aging people, many of them live for long life and enjoy well-functioning brain during the whole life-span. The biological basis of longevity is unknown. In this study, we investigated the influence of aging on hippocampal neural stem cells (NSCs), and the correlations between hippocampal neurogenesis and cognitive function. The result showed that the protein production and mRNA expression of nestin, and the number of BrdU(+) cells in dentate gyrus (DG) of the aged non-dementia mice were clearly higher than that in the aged dementia mice and the young adult mice. We also found that the number of NeuN(+) (neuron-specific nuclear antigen) cells in DG and CA1, choline O-acetyltransferase (ChAT, EC 2.3.1.6) production and mRNA expression in hippocampi of the aged-dementia mice were significantly reduced as compared to that of the young adult mice and the aged non-dementia mice, whereas the number of NeuN(+) cells, ChAT production and mRNA expression of the aged non-dementia mice has no difference with that of the young adult mice. Glial fibrillary acidic protein (GFAP) expression in the hippocampi of aged dementia mice significantly higher than that of the young adult mice and the aged non-dementia mice. Our results suggest that aging sometimes does not cause changing of the number of neurons and the hippocampal neurogenesis. Increment of DNA replication and neuron replacement, promotion of differentiation of neural stem cells, enhancement of neuronal proliferation, facilitation of synaptic plasticity of neurons may all benefit to the maintenance of the normal cognitive ability in the aged mice.


Subject(s)
Aging/pathology , Cognition , Hippocampus/pathology , Neurons/physiology , Stem Cells/physiology , Aging/psychology , Animals , Cell Count , Choline O-Acetyltransferase/biosynthesis , Choline O-Acetyltransferase/genetics , DNA-Binding Proteins , Dementia/pathology , Dementia/psychology , Female , Glial Fibrillary Acidic Protein , Immunohistochemistry , Intermediate Filament Proteins/biosynthesis , Intermediate Filament Proteins/genetics , Maze Learning , Mice , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurogenesis , Neurons/pathology , Nuclear Proteins/metabolism , RNA, Messenger/biosynthesis , Reaction Time , Stem Cells/pathology
11.
Exp Gerontol ; 43(9): 853-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18606218

ABSTRACT

Fuzhisan (FZS), a Chinese herbal complex prescription, has been used in the treatment of Alzheimer's disease (AD) for more than 15 years. Previous studies showed that FZS enhanced the cognitive ability in AD patients and AD model rats. FZS modulated the impaired cellular functions, and attenuated the damage caused by beta-amyloid protein, dose-dependently regulated and ameliorated the cholinergic functions of the Abeta(25-35)-induced AD-model mice. The SPECT imaging revealed that FZS improved the blood flow of the frontal and temporal lobes and the callosal gyrus in AD patients. However, little investigation of the effects of FZS on the naturally aged rats was reported. The underlying mechanism also remains to be explored. Recently we investigated the effects of the aqueous extract of FZS on the cognitive functions of the aged rats and the pharmacological basis for its therapeutic efficacy. The results showed a significant improvement made by FZS (0.3, 0.6, and 1.2 g/kg/d) for impaired cognitive functions of the aged rats. The rats manifested a shortened latency in Morris water maze test after intra-gavage administration (ig) of FZS for 30 consecutive days. The micro-positron emission tomography (microPET) using (18)F-2-fluoro-2-deoxy-D-glucose ((18)F-FDG) as the tracer demonstrated that FZS promoted the glucose metabolism in the whole brains especially the temporal and parietal regions in the aged rats. The spectrophotometry and Western blot showed that FZS obviously increased the activity and the production of choline O-acetyltransferase (ChAT, EC 2.3.1.6) and the acetylcholine (ACh) contents in the hippocampus, thus regulated and ameliorated the impaired cholinergic functions of the aged rats. The therapeutical effects of FZS on the learning and memory of the aged rats were dose-dependent. The mechanism of action of FZS in ameliorating the memory dysfunction of the aged rats is ascribed to the reinforcement of the function of the cholinergic system and the enhancement of the glucose metabolism in the brains. The results of this study, together with a survey of the findings in the clinical treatment with FZS suggest that FZS may not merely alleviate the symptoms of the dementia, but may also augment the production of the neurotransmitter ACh and the energy supply in the brain to build up fitness of the patients. FZS may be beneficial for the treatment of Alzheimer's disease or cognitive impairment in old people.


Subject(s)
Aging/drug effects , Drugs, Chinese Herbal/pharmacology , Maze Learning/drug effects , Phytotherapy/methods , Acetylcholine/metabolism , Aging/psychology , Animals , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Glucose/metabolism , Hippocampus/diagnostic imaging , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/physiology , Positron-Emission Tomography/methods , Rats , Rats, Wistar , Reaction Time/drug effects
12.
Eur J Pharmacol ; 594(1-3): 93-100, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18644361

ABSTRACT

Alzheimer's disease is a progressive brain disorder with the loss of memory and other intellectual abilities. Amyloid species and neurofibrillary tangles are the prime suspects in damaging and killing nerve cells. Abnormal accumulation of Amyloid-beta peptide (Abeta) may cause synaptic dysfunction and degeneration of neurons. Drugs that can prevent its formation and accumulation or stimulate its clearance might ultimately be of therapeutic benefit. Ciliary neurotrophic factor (CNTF), a neurotrophic cytokine, promotes the survival of various neurons in brain. However, the blood-brain barrier hinders the systemic delivery of CNTF to brain. Recently the 11-amino acid of protein transduction domain TAT has successfully assisted the delivery of many macromolecules to treat preclinical models of human disease. The present study aimed to evaluate whether P11-CNTF fusion protein (P11-CNTF) is protective against the Abeta25-35-induced dementia in mice. Immunofluorescence experiments showed that P11 effectively carried CNTF to the SH-SY5Y cells in vitro, and to the brains of mice in vivo. The learning and memory impairments of mice induced by Abeta were substantially rescued by supplement with the P11-CNTF. Furthermore, mRNAs of enzymes involved in the Abeta metabolism, e.g. neprilysin (NEP), endothelin-converting enzyme 1 (ECE-1) and insulin degrading enzyme (IDE), increased in the P11-CNTF treated dementia mice, accompanied by the proliferation of nestin- and choline acetyltransferase (ChAT)-positive cells in hippocampus. It implies that the delivery of P11-CNTF may be a novel treatment for Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides , Ciliary Neurotrophic Factor/pharmacology , Learning Disabilities/chemically induced , Learning Disabilities/drug therapy , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Phosphoproteins/pharmacology , Animals , Blotting, Western , Choline O-Acetyltransferase/biosynthesis , Ciliary Neurotrophic Factor/chemical synthesis , Ciliary Neurotrophic Factor/metabolism , Cloning, Molecular , DNA Primers , Dementia/chemically induced , Dementia/prevention & control , Flow Cytometry , Fluorescent Antibody Technique , Intermediate Filament Proteins/biosynthesis , Learning Disabilities/psychology , Male , Maze Learning/drug effects , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nerve Tissue Proteins/biosynthesis , Nestin , Neurofibrillary Tangles/pathology , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...