Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 428
Filtrar
1.
ACS Biomater Sci Eng ; 10(6): 3747-3758, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38753577

RESUMEN

Improving the clinical translation of nanomedicine requires better knowledge about how nanoparticles interact with biological environments. As researchers are recognizing the importance of understanding the protein corona and characterizing how nanocarriers respond in biological systems, new tools and techniques are needed to analyze nanocarrier-protein interactions, especially for smaller size (<10 nm) nanoparticles like polyamidoamine (PAMAM) dendrimers. Here, we developed a streamlined, semiquantitative approach to assess dendrimer-protein interactions using a nondenaturing electrophoresis technique combined with mass spectrometry. With this protocol, we detect fluorescently tagged dendrimers and proteins simultaneously, enabling us to analyze when dendrimers migrate with proteins. We found that PAMAM dendrimers mostly interact with complement proteins, particularly C3 and C4a, which aligns with previously published data, verifying that our approach can be used to isolate and identify dendrimer-protein interactions.


Asunto(s)
Dendrímeros , Dendrímeros/química , Dendrímeros/metabolismo , Prueba de Estudio Conceptual , Electroforesis , Humanos , Proteínas/química , Proteínas/metabolismo , Nanopartículas/química , Unión Proteica
2.
J Biomater Appl ; 38(6): 733-742, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37933579

RESUMEN

The application of hydrogels for anti-cancer drug delivery has garnered considerable interest in the medical field. Current cancer treatment approaches, such as chemotherapy and radiation therapy, often induce severe side effects, causing significant distress and substantial health complications to patients. Hydrogels present an appealing solution as they can be precisely injected into specific sites within the body, facilitating the sustainable release of encapsulated drugs. This localized treatment approach holds great potential for reducing toxicity levels and improving drug delivery efficacy. In this study we developed a hydrogel delivery system containing polyamidoamine (PAMAM) dendrimer and polyethylene glycol (PEG) for solubility enhancement and sustained delivery of hydrophobic anti-cancer drugs. The three selected model drugs, e.g. silibinin, camptothecin, and methotrexate, possess limited aqueous solubility and thus face restricted application. In the presence of vinyl sulfone functionalized PAMAM dendrimer at 45 mg/mL concentration, drug solubility is increased by 37-fold, 4-fold, and 10-fold for silibinin, camptothecin, and methotrexate, respectively. By further crosslinking of the functionalized PAMAM dendrimer and thiolated PEG, we successfully developed a fast-crosslinking hydrogel capable of encapsulating a significant payload of solubilized cancer drugs for sustained release. In water, the drug encapsulated hydrogels release 30%-80% of their loads in 1-4 days. MTT assays of J82 and MCF7 cells with various doses of drug encapsulated hydrogels reveal that cytotoxicity is observed for all three drugs on both J82 and MCF7 cell lines after 48 h. Notably, camptothecin exhibits higher cytotoxicity to both cell lines than silibinin and methotrexate, achieving up to 95% cell death at experimental conditions, despite its lower solubility. Our experiments provide evidence that the PAMAM dendrimer-mediated hydrogel system significantly improves the solubility of hydrophobic drugs and facilitates their sustained release. These findings position the system as a promising platform for controlled delivery of hydrophobic drugs for intratumoral cancer treatment.


Asunto(s)
Antineoplásicos , Dendrímeros , Humanos , Dendrímeros/química , Dendrímeros/metabolismo , Solubilidad , Metotrexato , Hidrogeles/química , Silibina , Preparaciones de Acción Retardada , Camptotecina , Células MCF-7
3.
Cell Mol Neurobiol ; 43(8): 3847-3884, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37725199

RESUMEN

Alzheimer's disease (AD) is the primary cause of dementia and is characterized by the death of brain cells due to the accumulation of insoluble amyloid plaques, hyperphosphorylation of tau protein, and the formation of neurofibrillary tangles within the cells. AD is also associated with other pathologies such as neuroinflammation, dysfunction of synaptic connections and circuits, disorders in mitochondrial function and energy production, epigenetic changes, and abnormalities in the vascular system. Despite extensive research conducted over the last hundred years, little is established about what causes AD or how to effectively treat it. Given the severity of the disease and the increasing number of affected individuals, there is a critical need to discover effective medications for AD. The US Food and Drug Administration (FDA) has approved several new drug molecules for AD management since 2003, but these drugs only provide temporary relief of symptoms and do not address the underlying causes of the disease. Currently, available medications focus on correcting the neurotransmitter disruption observed in AD, including cholinesterase inhibitors and an antagonist of the N-methyl-D-aspartate (NMDA) receptor, which temporarily alleviates the signs of dementia but does not prevent or reverse the course of AD. Research towards disease-modifying AD treatments is currently underway, including gene therapy, lipid nanoparticles, and dendrimer-based therapy. These innovative approaches aim to target the underlying pathological processes of AD rather than just managing the symptoms. This review discusses the novel aspects of pathogenesis involved in the causation of AD of AD and in recent developments in the therapeutic armamentarium for the treatment of AD such as gene therapy, lipid nanoparticles, and dendrimer-based therapy, and many more.


Asunto(s)
Enfermedad de Alzheimer , Dendrímeros , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Dendrímeros/metabolismo , Dendrímeros/uso terapéutico , Inhibidores de la Colinesterasa , Ovillos Neurofibrilares/metabolismo , Encéfalo/metabolismo , Péptidos beta-Amiloides/metabolismo
4.
Adv Drug Deliv Rev ; 200: 115005, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37419213

RESUMEN

Vision impairment and loss due to posterior segment ocular disorders, including age-related macular degeneration and diabetic retinopathy, are a rapidly growing cause of disability globally. Current treatments consist primarily of intravitreal injections aimed at preventing disease progression and characterized by high cost and repeated clinic visits. Nanotechnology provides a promising platform for drug delivery to the eye, with potential to overcome anatomical and physiological barriers to provide safe, effective, and sustained treatment modalities. However, there are few nanomedicines approved for posterior segment disorders, and fewer that target specific cells or that are compatible with systemic administration. Targeting cell types that mediate these disorders via systemic administration may unlock transformative opportunities for nanomedicine and significantly improve patient access, acceptability, and outcomes. We highlight the development of hydroxyl polyamidoamine dendrimer-based therapeutics that demonstrate ligand-free cell targeting via systemic administration and are under clinical investigation for treatment of wet age-related macular degeneration.


Asunto(s)
Dendrímeros , Oftalmopatías , Degeneración Macular , Humanos , Dendrímeros/metabolismo , Ojo/metabolismo , Sistemas de Liberación de Medicamentos , Oftalmopatías/metabolismo , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo
5.
Int J Biol Macromol ; 246: 125714, 2023 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-37423440

RESUMEN

Severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) and influenza viruses have spread around the world at an unprecedented rate. Despite multiple vaccines, new variants of SARS-CoV-2 and influenza have caused a remarkable level of pathogenesis. The development of effective antiviral drugs to treat SARS-CoV-2 and influenza remains a high priority. Inhibiting viral cell surface attachment represents an early and efficient means to block virus infection. Sialyl glycoconjugates, on the surface of human cell membranes, play an important role as host cell receptors for influenza A virus and 9-O-acetyl-sialylated glycoconjugates are receptors for MERS, HKU1 and bovine coronaviruses. We designed and synthesized multivalent 6'-sialyllactose-counjugated polyamidoamine dendrimers through click chemistry at room temperature concisely. These dendrimer derivatives have good solubility and stability in aqueous solutions. SPR, a real-time analysis quantitative method for of biomolecular interactions, was used to study the binding affinities of our dendrimer derivatives by utilizing only 200 micrograms of each dendrimer. Three SARS-CoV-2 S-protein receptor binding domain (wild type and two Omicron mutants) bound to multivalent 9-O-acetyl-6'-sialyllactose-counjugated and 6'-sialyllactose-counjugated dendrimers bound to a single H3N2 influenza A virus's HA protein (A/Hong Kong/1/1968), the SPR study results suggest their potential anti-viral activities.


Asunto(s)
COVID-19 , Dendrímeros , Gripe Humana , Animales , Bovinos , Humanos , Glicoproteína de la Espiga del Coronavirus/química , SARS-CoV-2/metabolismo , Dendrímeros/farmacología , Dendrímeros/metabolismo , Gripe Humana/tratamiento farmacológico , Hemaglutininas , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Antivirales/química , Unión Proteica
6.
J Control Release ; 358: 27-42, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37054778

RESUMEN

Glutamate carboxypeptidase II (GCPII), localized on the surface of astrocytes and activated microglia, regulates extracellular glutamate concentration in the central nervous system (CNS). We have previously shown that GCPII is upregulated in activated microglia in the presence of inflammation. Inhibition of GCPII activity could reduce glutamate excitotoxicity, which may decrease inflammation and promote a 'normal' microglial phenotype. 2-(3-Mercaptopropyl) pentanedioic acid (2-MPPA) is the first GCPII inhibitor that underwent clinical trials. Unfortunately, immunological toxicities have hindered 2-MPPA clinical translation. Targeted delivery of 2-MPPA specifically to activated microglia and astrocytes that over-express GCPII has the potential to mitigate glutamate excitotoxicity and attenuate neuroinflammation. In this study, we demonstrate that 2-MPPA when conjugated to generation-4, hydroxyl-terminated polyamidoamine (PAMAM) dendrimers (D-2MPPA) localize specifically in activated microglia and astrocytes only in newborn rabbits with cerebral palsy (CP), not in controls. D-2MPPA treatment led to higher 2-MPPA levels in the injured brain regions compared to 2-MPPA treatment, and the extent of D-2MPPA uptake correlated with the injury severity. D-2MPPA was more efficacious than 2-MPPA in decreasing extracellular glutamate level in ex vivo brain slices of CP kits, and in increasing transforming growth factor beta 1 (TGF-ß1) level in primary mixed glial cell cultures. A single systemic intravenous dose of D-2MPPA on postnatal day 1 (PND1) decreased microglial activation and resulted in a change in microglial morphology to a more ramified form along with amelioration of motor deficits by PND5. These results indicate that targeted dendrimer-based delivery specifically to activated microglia and astrocytes can improve the efficacy of 2-MPPA by attenuating glutamate excitotoxicity and microglial activation.


Asunto(s)
Parálisis Cerebral , Dendrímeros , Animales , Conejos , Parálisis Cerebral/metabolismo , Dendrímeros/metabolismo , Ácido Glutámico , Encéfalo/metabolismo , Microglía/metabolismo , Inflamación/tratamiento farmacológico
7.
Exp Dermatol ; 32(7): 986-995, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37015835

RESUMEN

Glabrous skin is hair-free skin with a high density of sweat glands, which is found on the palms, and soles of mammalians, covered with a thick stratum corneum. Dry hands are often an occupational problem which deserves attention from dermatologists. Urea is found in the skin as a component of the natural moisturizing factor and of sweat. We report the discovery of dendrimer structures of crystalized urea in the stratum corneum of palmar glabrous skin using laser scanning microscopy. The chemical and structural nature of the urea crystallites was investigated in vivo by non-invasive techniques. The relation of crystallization to skin hydration was explored. We analysed the index finger, small finger and tenar palmar area of 18 study participants using non-invasive optical methods, such as laser scanning microscopy, Raman microspectroscopy and two-photon tomography. Skin hydration was measured using corneometry. Crystalline urea structures were found in the stratum corneum of about two-thirds of the participants. Participants with a higher density of crystallized urea structures exhibited a lower skin hydration. The chemical nature and the crystalline structure of the urea were confirmed by Raman microspectroscopy and by second harmonic generated signals in two-photon tomography. The presence of urea dendrimer crystals in the glabrous skin seems to reduce the water binding capacity leading to dry hands. These findings highlight a new direction in understanding the mechanisms leading to dry hands and open opportunities for the development of better moisturizers and hand disinfection products and for diagnostic of dry skin.


Asunto(s)
Dendrímeros , Urea , Animales , Humanos , Dendrímeros/metabolismo , Epidermis/metabolismo , Agua/metabolismo , Mano , Mamíferos
8.
BMC Oral Health ; 23(1): 141, 2023 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-36906526

RESUMEN

BACKGROUND: Intrafibrillar remineralization within the hybrid layers (HLs) has recently attracted extensive attention in achieving durable resin-dentin bonds. The polyhydroxy-terminated poly(amidoamine) dendrimer (PAMAM-OH) at fourth generation becomes a desirable candidate to induce intrafibrillar remineralization to protect exposed collagen fibrils within HLs based on the size exclusion effect of fibrillar collagen. However, the remineralization process in vivo is time-consuming, during which the exposed collagen fibrils are vulnerable to enzymatic degradation, resulting in unsatisfactory remineralization. Thereby, if PAMAM-OH itself possesses concomitant anti-proteolytic activity during the induction of remineralization, it would be very beneficial to obtain satisfactory remineralization. METHODS: Binding capacity tests using adsorption isotherm and confocal laser scanning microscopy (CLSM) were performed to assess if the PAMAM-OH had adsorption capacity on dentin. Anti-proteolytic testings were detected by MMPs assay kit, in-situ zymography and ICTP assay. Adhesive infiltration of resin-dentin interface and tensile bond strength before and after thermomechanical cycling were implemented to assess if the PAMAM-OH adversely affected resin-dentin bonds. RESULTS: Anti-proteolytic testings performed using MMPs assay kit, in-situ zymography and ICTP assay indicated that PAMAM-OH inhibited exogenous soluble MMP-9 as well as had inhibitory effect on the endogenous proteases. Adhesive infiltration of resin-dentin interface and tensile bond strength before and after thermomechanical cycling were implemented to indicate that the PAMAM-OH pretreatment had no adverse effects on immediate dentin bonding and prolonged the durability of resin-dentin bonds. CONCLUSIONS: PAMAM-OH possesses anti-proteolytic activity and prevents exposed collagen fibrils within HLs from degradation, which lays the foundation for the satisfactory intrafibrillar remineralization induced by PAMAM-OH within HLs to achieve durable resin-dentin bonds in the next work.


Asunto(s)
Dendrímeros , Recubrimiento Dental Adhesivo , Colágeno/metabolismo , Dendrímeros/análisis , Dendrímeros/metabolismo , Recubrimiento Dental Adhesivo/métodos , Dentina/metabolismo , Recubrimientos Dentinarios/química , Ensayo de Materiales , Metaloproteinasas de la Matriz/metabolismo , Resistencia a la Tracción
9.
Biomater Sci ; 11(4): 1530-1539, 2023 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-36607143

RESUMEN

The development of efficient nanomedicines to repress the repolarization of M1 phenotype macrophages and therefore inhibit pro-inflammatory cytokine overexpression for anti-inflammatory therapy is still a challenging task. We report here an original gene delivery nanoplatform based on pyrrolidinium-modified amphiphilic generation 1 phosphorus dendron (C12G1) nanomicelles with a rigid phosphorous dendron structure. The nanomicelles display higher gene delivery efficiency than the counterpart materials of pyrrolidinium-modified G1 phosphorus dendrimers, and meanwhile exhibit excellent cytocompatibility. The C12G1 nanomicelles can be employed to co-deliver the miRNA-146a mimic (miR-146a mimic) and miRNA-429 inhibitor (miR-429i) to inhibit the Toll-like receptor-4 signaling pathway and p38 mitogen-activated protein kinase signaling pathway, respectively, thus causing repression of M1 phenotype alveolar macrophage polarization. The developed C12G1/miR-mixture polyplexes enable efficient therapy of lipopolysaccharide-activated alveolar macrophages in vitro and an acute lung injury mouse model in vivo. The generated cationic phosphorus dendron nanomicelles may hold promising potential for anti-inflammatory gene therapy of other inflammatory diseases.


Asunto(s)
Lesión Pulmonar Aguda , Dendrímeros , MicroARNs , Ratones , Animales , MicroARNs/metabolismo , Dendrímeros/metabolismo , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Macrófagos/metabolismo , Antiinflamatorios/química
10.
Biomater Sci ; 11(5): 1589-1606, 2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36692071

RESUMEN

Polyamidoamines (PAMAMs) are a class of dendrimer with monodispersity and controlled topology, which can deliver biologically active macromolecules (e.g., genes and proteins) to specific regions with high efficiency and minimum side effects. In detail, PAMAMs can be functionalized easily by core modification or surface amendment to encapsulate a wide range of biomacromolecules. Besides, self-assembled, cross-linked and hybrid PAMAMs with customized therapeutic purposes are developed as delivery vehicles, which makes PAMAMs promising for biomacromolecule therapy. In this review, we comprehensively summarize the application of PAMAMs in biomacromolecule delivery from the synthesis of functionalized PAMAM carriers to the development of PAMAM-based drug delivery systems. The underlying strategies for PAMAM functionalization and assembly are first systematically discussed, and then the current applications of PAMAMs for biomacromolecule delivery are reviewed. Finally, a brief perspective on the further applications of PAMAMs concludes, aiming to provide insights into developing PAMAM-based biomacromolecule delivery systems.


Asunto(s)
Dendrímeros , Dendrímeros/metabolismo , Sistemas de Liberación de Medicamentos , Portadores de Fármacos
11.
Biomater Sci ; 10(21): 6258-6266, 2022 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-36111777

RESUMEN

Apoptosis of cardiomyocytes is a critical outcome of myocardial ischemia-reperfusion injury (MIRI), which leads to the permanent impairment of cardiac function. Upregulated E2F1 is implicated in inducing cardiomyocyte apoptosis, and thus intervention of the E2F1 signaling pathway via RNA interference may hold promising potential for rescuing the myocardium from MIRI. To aid efficient E2F1 siRNA (siE2F1) delivery into cardiomyocytes that are normally hard to transfect, a spherical, α-helical polypeptide (SPP) with potent membrane activity was developed via dendrimer-initiated ring-opening polymerization of N-carboxyanhydride followed by side-chain functionalization with guanidines. Due to its multivalent structure, SPP outperformed its linear counterpart (LPP) to feature potent siRNA binding affinity and membrane activity. Thus, SPP effectively delivered siE2F1 into cardiomyocytes and suppressed E2F1 expression both in vitro and in vivo after intramyocardial injection. The E2F1-miR421-Pink1 signaling pathway was disrupted, thereby leading to the reduction of MIRI-induced mitochondrial damage, apoptosis, and inflammation of cardiomyocytes and ultimately recovering the systolic function of the myocardium. This study provides an example of membrane-penetrating nucleic acid delivery materials, and it also provides a promising approach for the genetic manipulation of cardiomyocyte apoptosis for the treatment of MIRI.


Asunto(s)
Dendrímeros , Daño por Reperfusión Miocárdica , Ratas , Animales , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Dendrímeros/metabolismo , Ratas Sprague-Dawley , Apoptosis , Péptidos/genética , Péptidos/farmacología , Péptidos/metabolismo , Guanidinas/farmacología , Guanidinas/uso terapéutico , Proteínas Quinasas/metabolismo , Proteínas Quinasas/farmacología , Proteínas Quinasas/uso terapéutico , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F1/farmacología
12.
Curr Drug Metab ; 23(9): 708-722, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35713127

RESUMEN

Gliomas are the most prevailing intracranial tumors, which account for approximately 36% of the primary brain tumors of glial cells. Glioblastoma multiforme (GBM) possesses a higher degree of malignancy among different gliomas. The blood-brain barrier (BBB) protects the brain against infections and toxic substances by preventing foreign molecules or unwanted cells from entering the brain parenchyma. Nano-carriers such as liposomes, nanoparticles, dendrimers, etc. boost the brain permeability of various anticancer drugs or other drugs. The favorable properties like small size, better solubility, and the modifiable surface of dendrimers have proven their broad applicability in the better management of GBM. However, in vitro and in vivo toxicities caused by dendrimers have been a significant concern. The presence of multiple functionalities on the surface of dendrimers enables the grafting of target ligand and/or therapeutic moieties. Surface engineering improves certain properties like targeting efficiency, pharmacokinetic profile, therapeutic effect, and toxicity reduction. This review will be focused on the role of different surface-modified dendrimers in the effective management of GBM.


Asunto(s)
Antineoplásicos , Neoplasias Encefálicas , Dendrímeros , Glioblastoma , Glioma , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Dendrímeros/metabolismo , Dendrímeros/farmacología , Encéfalo/metabolismo , Antineoplásicos/farmacología , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología
13.
Int J Mol Sci ; 23(10)2022 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-35628503

RESUMEN

Tumor cells with stem cell properties are considered to play major roles in promoting the development and malignant behavior of aggressive cancers. Therapeutic strategies that efficiently eradicate such tumor stem cells are of highest clinical need. Herein, we performed the validation of the polycationic phosphorus dendrimer-based approach for small interfering RNAs delivery in in vitro stem-like cells as models. As a therapeutic target, we chose Lyn, a member of the Src family kinases as an example of a prominent enzyme class widely discussed as a potent anti-cancer intervention point. Our selection is guided by our discovery that Lyn mRNA expression level in glioma, a class of brain tumors, possesses significant negative clinical predictive value, promoting its potential as a therapeutic target for future molecular-targeted treatments. We then showed that anti-Lyn siRNA, delivered into Lyn-expressing glioma cell model reduces the cell viability, a fact that was not observed in a cell model that lacks Lyn-expression. Furthermore, we have found that the dendrimer itself influences various parameters of the cells such as the expression of surface markers PD-L1, TIM-3 and CD47, targets for immune recognition and other biological processes suggested to be regulating glioblastoma cell invasion. Our findings prove the potential of dendrimer-based platforms for therapeutic applications, which might help to eradicate the population of cancer cells with augmented chemotherapy resistance. Moreover, the results further promote our functional stem cell technology as suitable component in early stage drug development.


Asunto(s)
Neoplasias Encefálicas , Dendrímeros , Glioblastoma , Glioma , Neoplasias Encefálicas/metabolismo , Dendrímeros/metabolismo , Dendrímeros/farmacología , Glioblastoma/metabolismo , Glioma/metabolismo , Humanos , Células Madre Neoplásicas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
14.
J Neuroendocrinol ; 34(5): e13094, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35107859

RESUMEN

The concept that different systems control episodic and surge secretion of gonadotropin-releasing hormone (GnRH) was well established by the time that GnRH was identified and formed the framework for studies of the physiological roles of GnRH, and later kisspeptin. Here, we focus on recent studies identifying the neural mechanisms underlying these two modes of secretion, with an emphasis on their core components. There is now compelling data that kisspeptin neurons in the arcuate nucleus that also contain neurokinin B (NKB) and dynorphin (i.e., KNDy cells) and their projections to GnRH dendrons constitute the GnRH pulse generator in mice and rats. There is also strong evidence for a similar role for KNDy neurons in sheep and goats, and weaker data in monkeys and humans. However, whether KNDy neurons act on GnRH dendrons and/or GnRH soma and dendrites that are found in the mediobasal hypothalamus (MBH) of these species remains unclear. The core components of the GnRH/luteinising hormone surge consist of an endocrine signal that initiates the process and a neural trigger that drives GnRH secretion during the surge. In all spontaneous ovulators, the core endocrine signal is a rise in estradiol secretion from the maturing follicle(s), with the site of estrogen positive feedback being the rostral periventricular kisspeptin neurons in rodents and neurons in the MBH of sheep and primates. There is considerable species variations in the neural trigger, with three major classes. First, in reflex ovulators, this trigger is initiated by coitus and carried to the hypothalamus by neural or vascular pathways. Second, in rodents, there is a time of day signal that originates in the suprachiasmatic nucleus and activates rostral periventricular kisspeptin neurons and GnRH soma and dendrites. Finally, in sheep nitric oxide-producing neurons in the ventromedial nucleus, KNDy neurons and rostral kisspeptin neurons all appear to participate in driving GnRH release during the surge.


Asunto(s)
Dendrímeros , Hormona Liberadora de Gonadotropina , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Dendrímeros/metabolismo , Dinorfinas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Ratones , Neuroquinina B/metabolismo , Ratas , Ovinos
15.
Biol Chem ; 403(3): 345-360, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34883001

RESUMEN

The activity and interacting ability of a polyamidoamine (PAMAM) dendrimer modified with 4-N-methylpiperazine-1,8-naphthalimide units (termed D) and complexed by Cu(ii) ions, towards healthy and cancer cells were studied. Comparative electron paramagnetic resonance (EPR) studies of the Cu(ii)-D complex are presented: coordination mode, chemical structure, flexibility and stability of these complexes, in the absence and presence of myeloid cancer cells and peripheral blood mononuclear cells (PBMC). The interactions of Cu(ii) ions in the biological media at different equilibrium times were studied, highlighting different stability and interacting conditions with the cells. Furthermore, flow cytometry and confocal analysis, trace the peculiar properties of the dendrimers in PBMC and U937 cells. Indeed, a new probe (Fly) was used as a potential fluorescent tool for biological imaging of Cu(ii). The study highlights that dendrimer and, mainly, the Cu(ii) metallodendrimer are cytotoxic agents for the cells, specifically for U937 tumor cells, inducing mitochondrial dysfunction, ROS increase and lysosome involvement. The metallodendrimer shows antitumor selectivity, fewer affecting healthy PBMC, inducing a massive apoptotic cell death on U937 cells, in line with the high stability of this complex, as verified by EPR studies. The results underline the potentiality of this metallodendrimer to be used as anticancer drug.


Asunto(s)
Antineoplásicos , Dendrímeros , Neoplasias , Antineoplásicos/química , Antineoplásicos/farmacología , Dendrímeros/química , Dendrímeros/metabolismo , Dendrímeros/farmacología , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Leucocitos Mononucleares , Naftalimidas/farmacología , Poliaminas
16.
Biomaterials ; 280: 121323, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34942563

RESUMEN

A large number of cytokines or growth factors have been used in the treatment of inflammation. However, they are highly dependent on an optimal delivery system with sufficient loading efficiency and protection of growth factors from proteolytic degradation. To develop the immunotherapy capacity of peptide dendrimers themselves, inspired by the structure and immunoregulatory functions of mannose-capped lipoarabinomannan (ManLAM), we thus propose a hypothesis that mannose-decorated globular lysine dendrimers (MGLDs) with precise molecular design can elicit anti-inflammatory activity through targeting and reprogramming macrophages to M2 phenotype. To achieve this, a series of mannose-decorated globular lysine dendrimers (MGLDs) was developed. Size-controlled MGLDs obtained were spherical with positive surface charges. The mean size ranged from 50-200 nm in varying generations and modification degrees. The initial screening study revealed that MGLDs have superior biocompatibility. When cocultured with MGLDs, mouse bone marrow-derived macrophages (BMDMs) acquired an anti-inflammatory M2 phenotype characterized by significant mannose receptor (MR) clustering on the cell surface and the elongated shape, an increased production of transforming growth factor (TGF)-ß1, interleukin (IL)-4 and IL-10, a downregulated secretory of IL-1ß, IL-6, and tumor necrosis factor (TNF)-α, and increased ability to induce fibroblast proliferation. Then in vivo studies further demonstrated that topical administration of optimized MGLDs accelerates wound repair of full-thickness cutaneous defects in type 2 diabetic mice via M2 macrophage polarization. Mechanistically, MGLDs treatment showed an enhanced closure rate, collagen deposition, and angiogenesis, along with mitigated inflammation modulated by a suppressed secretory of pro-inflammation cytokines, and increased production of TGF-ß1. These findings provide the first evidence that the bioinspired design of MGLDs can direct M2 macrophage polarization, which may be beneficial in the therapy of injuries and inflammation.


Asunto(s)
Dendrímeros , Diabetes Mellitus Experimental , Animales , Dendrímeros/metabolismo , Diabetes Mellitus Experimental/patología , Lisina , Macrófagos/metabolismo , Manosa/metabolismo , Ratones , Cicatrización de Heridas
17.
Adv Sci (Weinh) ; 9(4): e2103098, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34894089

RESUMEN

The multivalent binding effect has been the subject of extensive studies to modulate adhesion behaviors of various biological and engineered systems. However, precise control over the strong avidity-based binding remains a significant challenge. Here, a set of engineering strategies are developed and tested to systematically enhance the multivalent binding of peptides in a stepwise manner. Poly(amidoamine) (PAMAM) dendrimers are employed to increase local peptide densities on a substrate, resulting in hierarchically multivalent architectures (HMAs) that display multivalent dendrimer-peptide conjugates (DPCs) with various configurations. To control binding behaviors, effects of the three major components of the HMAs are investigated: i) poly(ethylene glycol) (PEG) linkers as spacers between conjugated peptides; ii) multiple peptides on the DPCs; and iii) various surface arrangements of HMAs (i.e., a mixture of DPCs each containing different peptides vs DPCs cofunctionalized with multiple peptides). The optimized HMA configuration enables significantly enhanced target cell binding with high selectivity compared to the control surfaces directly conjugated with peptides. The engineering approaches presented herein can be applied individually or in combination, providing guidelines for the effective utilization of biomolecular multivalent interactions using DPC-based HMAs.


Asunto(s)
Neoplasias de la Mama/metabolismo , Adhesión Celular , Nanopartículas/metabolismo , Péptidos/metabolismo , Línea Celular Tumoral , Dendrímeros/metabolismo , Humanos , Fenómenos Físicos , Polietilenglicoles/metabolismo
18.
Adv Sci (Weinh) ; 8(24): e2102991, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34719882

RESUMEN

The assembly of proteins in a programmable manner provides insight into the creation of novel functional nanomaterials for practical applications. Despite many advances, however, a rational protein assembly with an easy scalability in terms of size and valency remains a challenge. Here, a simple bottom-up approach to the supramolecular protein assembly with a tunable size and valency in a programmable manner is presented. The dendrimer-like protein assembly, simply called a "protein dendrimer," is constructed through a stepwise and alternate addition of a building block protein. Starting from zeroth-generation protein dendrimer (pG0 ) of 27 kDa, the protein dendrimer is sequentially grown to pG1 , pG2 , pG3 , to pG4 with a molecular mass of 94, 216, 483, and 959 kDa, respectively. The valency of the protein dendrimers at the periphery increases by a factor of two after each generation, allowing a tunable valency and easy functionalization. The protein dendrimers functionalizes with a targeting moiety and a cytotoxic protein cargo shows a typical feature of multi-valency in the avidity and a highly enhanced cellular cytotoxicity, exemplifying their utility as a protein delivery platform. The present approach can be effectively used in the creation of protein architectures with new functions for biotechnological and medical applications.


Asunto(s)
Antineoplásicos/administración & dosificación , Dendrímeros/metabolismo , Neoplasias/tratamiento farmacológico , Línea Celular Tumoral , Células Cultivadas , Humanos , Microscopía Confocal , Nanoestructuras , Neoplasias/diagnóstico por imagen
19.
Genes (Basel) ; 12(7)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34356120

RESUMEN

The transcription factor high mobility group protein A2 (HMGA2) plays an important role in the pathogenesis of some cancers including breast cancer. Polyamidoamine dendrimer generation 4 is a kind of highly branched polymeric nanoparticle with surface charge and highest density peripheral groups that allow ligands or therapeutic agents to attach it, thereby facilitating target delivery. Here, methotrexate (MTX)- modified polyamidoamine dendrimer generation 4 (G4) (G4/MTX) was generated to deliver specific small interface RNA (siRNA) for suppressing HMGA2 expression and the consequent effects on folate receptor (FR) expressing human breast cancer cell lines (MCF-7, MDA-MB-231). We observed that HMGA2 siRNA was electrostatically adsorbed on the surface of the G4/MTX nanocarrier for constructing a G4/MTX-siRNA nano-complex which was verified by changing the final particle size and zeta potential. The release of MTX and siRNA from synthesized nanocomplexes was found in a time- and pH-dependent manner. We know that MTX targets FR. Interestingly, G4/MTX-siRNA demonstrates significant cellular internalization and gene silencing efficacy when compared to the control. Besides, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay demonstrated selective cell cytotoxicity depending on the folate receptor expressing in a dose-dependent manner. The gene silencing and protein downregulation of HMGA2 by G4/MTX-siRNA was observed and could significantly induce cell apoptosis in MCF-7 and MDA-MB-231 cancer cells compared to the control group. Based on the findings, we suggest that the newly developed G4/MTX-siRNA nano-complex may be a promising strategy to increase apoptosis induction through HMGA2 suppression as a therapeutic target in human breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Proteína HMGA2/genética , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Dendrímeros/metabolismo , Dendrímeros/farmacología , Dendrímeros/uso terapéutico , Femenino , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen , Proteína HMGA2/metabolismo , Humanos , Células MCF-7 , Metotrexato/farmacología , Nylons/farmacología , ARN Interferente Pequeño/genética
20.
Nat Commun ; 12(1): 4858, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34381048

RESUMEN

Complement is an enzymatic humoral pattern-recognition defence system of the body. Non-specific deposition of blood biomolecules on nanomedicines triggers complement activation through the alternative pathway, but complement-triggering mechanisms of nanomaterials with dimensions comparable to or smaller than many globular blood proteins are unknown. Here we study this using a library of <6 nm poly(amido amine) dendrimers bearing different end-terminal functional groups. Dendrimers are not sensed by C1q and mannan-binding lectin, and hence do not trigger complement activation through these pattern-recognition molecules. While, pyrrolidone- and carboxylic acid-terminated dendrimers fully evade complement response, and independent of factor H modulation, binding of amine-terminated dendrimers to a subset of natural IgM glycoforms triggers complement activation through lectin pathway-IgM axis. These findings contribute to mechanistic understanding of complement surveillance of dendrimeric materials, and provide opportunities for dendrimer-driven engineering of complement-safe nanomedicines and medical devices.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Dendrímeros/metabolismo , Inmunoglobulina M/metabolismo , Activación de Complemento/efectos de los fármacos , Complemento C1q/metabolismo , Dendrímeros/química , Dendrímeros/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacología , Humanos , Lectina de Unión a Manosa/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa , Poliaminas/química , Poliaminas/metabolismo , Poliaminas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...