Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Theranostics ; 10(7): 3138-3150, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194859

RESUMO

Multiple sclerosis (MS) is a demyelinating inflammatory disease of the central nervous system (CNS), which is a chronic progressive disease and is caused by uncontrolled activation of myelin antigen specific T cells. It has high unmet medical needs due to the difficulty of efficient drug delivery into the CNS to control tissue inflammation. In this study, we demonstrate that a fusion protein of NOD-like receptor family member X1 (NLRX1) and blood brain barrier (BBB)-permeable peptide, dNP2 ameliorates experimental autoimmune encephalomyelitis (EAE). Methods: We purified recombinant LRR or NBD regions of NLRX1 protein conjugated with dNP2. To examine intracellular delivery efficiency of the recombinant protein, we incubated the proteins with Jurkat T cells or murine splenic T cells and their delivery efficiency was analyzed by flow cytometry. To investigate the therapeutic efficacy in an EAE model, we injected the recombinant protein into mice with 3 different treatment schemes e.g., prevention, semi-therapeutic, and therapeutic. To analyze their functional roles in T cells, we treated MACS-sorted naïve CD4 T cells with the proteins during their activation and differentiation into Th1, Th17, and Treg cells. Results: dNP2-LRR protein treatment showed significantly higher delivery efficiency than TAT-LRR or LRR alone in Jurkat T cells and mouse splenic T cells. In all three treatment schemes of EAE experiments, dNP2-LRR administration showed ameliorated tissue inflammation and disease severity with reduced number of infiltrating T cells producing inflammatory cytokines such as IFNγ. In addition, dNP2-LRR inhibited T cell activation, cytokine production, and Th1 differentiation. Conclusion: These results suggest that dNP2-LRR is a novel agent, which regulates effector T cell functions and could be a promising molecule for the treatment of CNS autoimmune diseases such as multiple sclerosis.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Portadores de Fármacos/administração & dosagem , Encefalomielite Autoimune Experimental/tratamento farmacológico , Proteínas Mitocondriais/química , Linfócitos T/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Barreira Hematoencefálica , Peptídeos Penetradores de Células/farmacocinética , Portadores de Fármacos/farmacocinética , Avaliação Pré-Clínica de Medicamentos , Encefalomielite Autoimune Experimental/imunologia , Feminino , Humanos , Células Jurkat , Ativação Linfocitária/efeitos dos fármacos , Linfocinas/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacocinética , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Domínios Proteicos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Organismos Livres de Patógenos Específicos , Medula Espinal/metabolismo , Medula Espinal/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
2.
Pharmacol Res Perspect ; 7(6): e00547, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31832205

RESUMO

Activation of MrgX2, an orphan G protein-coupled receptor expressed on mast cells, leads to degranulation and histamine release. Human MrgX2 binds promiscuously to structurally diverse peptides and small molecules that tend to have basic properties (basic secretagogues), resulting in acute histamine-like adverse drug reactions of injected therapeutic agents. We set out to identify MrgX2 orthologues from other mammalian species used in nonclinical stages of drug development. Previously, the only known orthologue of human MrgX2 was from mouse, encoded by Mrgprb2. MrgX2 genes of rat, dog (beagle), minipig, pig, and Rhesus and cynomolgus monkey were identified by bioinformatic approaches and verified by their ability to mediate calcium mobilization in transfected cells in response to the classical MrgX2 agonist, compound 48/80. The peptide GSK3212448 is an inhibitor of the PRC2 epigenetic regulator that caused profound anaphylactoid reactions upon intravenous infusion to rat. We showed GSK3212448 to be a potent MrgX2 agonist particularly at rat MrgX2. We screened sets of drug-like molecules and peptides to confirm the highly promiscuous nature of MrgX2. Approximately 20% of drug-like molecules activated MrgX2 (pEC50 ranging from 4.5 to 6), with the principle determinant being basicity. All peptides tested of net charge +3 or greater exhibited agonist activity, including the cell penetrating peptides polyarginine (acetyl-Arg9-amide) and TAT (49-60), a fragment of HIV-1 TAT protein. Finally, we showed that the glycopeptide antibiotic vancomycin, which is associated with clinical pseudo-allergic reactions known as red man syndrome, is an agonist of MrgX2.


Assuntos
Anafilaxia/induzido quimicamente , Mastócitos/efeitos dos fármacos , Proteínas do Tecido Nervoso/agonistas , Fragmentos de Peptídeos/efeitos adversos , Receptores Acoplados a Proteínas G/agonistas , Receptores de Neuropeptídeos/agonistas , Vancomicina/efeitos adversos , Anafilaxia/imunologia , Animais , Degranulação Celular/efeitos dos fármacos , Degranulação Celular/imunologia , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/administração & dosagem , Peptídeos Penetradores de Células/efeitos adversos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/efeitos adversos , Células HEK293 , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Humanos , Mastócitos/imunologia , Mastócitos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/imunologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/imunologia , Receptores de Neuropeptídeos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Síndrome , Vancomicina/administração & dosagem , p-Metoxi-N-metilfenetilamina/farmacologia
3.
Mol Pharm ; 16(3): 1140-1155, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30668131

RESUMO

Chronic inflammation is closely related to the development, deterioration, and metastasis of tumors. Recently, many studies have shown that down-regulating the expression of inflammation by blocking nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways could significantly inhibit tumor growth and metastasis. The combined application of curcumin (CUR) and celecoxib (CXB) has been proven to exert a synergistic antitumor effect via inhibiting the activation of NF-κB and STAT3. TAT-NBD (TN) peptide, a fusion peptide of NF-κB essential modulator (NEMO)-binding domain peptide (NBD) and cell-penetrating peptide (TAT), can selectively block NF-κB activating pathway resulting in tumor growth inhibition. In the present study, a novel TN-modified liposome coloading both CXB and CUR (TN-CCLP) at a synergistic ratio was first constructed with the property of synchronous release, then hyaluronic acid (HA) as CD44 targeting moiety was coated on the surface of the cationic liposome via electrostatic interaction to prepare the anionic HA/TN-CCLP. In vitro results of cytotoxicity, macrophage migration inhibition, and anti-inflammation efficacy revealed that TN-CCLP and HA/TN-CCLP were significantly superior to TN-LP and CCLP, while TN-CCLP exhibited better effects than HA/TN-CCLP due to higher cellular uptake ability. Different from in vitro data, after systematically treating 4T1 breast tumor-bearing mice, HA/TN-CCLP exerted the most striking effects on anti-inflammation, inhibition of macrophage recruitment, and antitumor because of the longest circulation time and maximum tumor accumulation. In particular, HA/TN-CCLP could availably block the lung metastasis of breast cancer. Taken together, the novel CD44 targeted TN-CCLP exhibited the potential for inhibiting tumor development and metastasis through improving inflammatory infiltration of tumor tissue.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Peptídeos Penetradores de Células/uso terapêutico , Quimioterapia Combinada/métodos , Ácido Hialurônico/química , Neoplasias Inflamatórias Mamárias/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Apoptose/efeitos dos fármacos , Celecoxib/uso terapêutico , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Curcumina/uso terapêutico , Liberação Controlada de Fármacos , Sinergismo Farmacológico , Feminino , Xenoenxertos , Humanos , Receptores de Hialuronatos/metabolismo , Neoplasias Inflamatórias Mamárias/patologia , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Resultado do Tratamento , Carga Tumoral
4.
Res Vet Sci ; 114: 502-510, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28987957

RESUMO

The absence of an effective therapy against human solid tumors has fostered the development of promising antineoplastic therapeutic candidates, as the CIGB-552 peptide. This synthetic peptide has shown to be effective in reducing tumor size and increasing the lifespan in tumor-bearing mice. Therefore, this work was aimed to explore the safety profile and preliminary assessment of antitumor activity of the CIGB-552 peptide therapeutic candidate in a small population of dogs (n=9) having malignant spontaneously-arising solid tumors. The peptide was administered by subcutaneous (s.c.) route, at three dosage levels (0.075, 0.15 and 0.3mg/kg). The results showed no dose-limiting toxicities in any dogs. The antitumor activity observed in dogs receiving CIGB-552 was associated with the reduction in the tumor volume. Given the antitumor effects of CIGB-552 as mediated by COMMD1 protein, which function is highly conserved among eukaryotic organisms, and the similarities of canine and human types of cancer with respect to tumor biology, it is likely that CIGB-552 could demonstrate comparable anti-cancer activity in human patients. Synthetic peptide, COMMD1, Tumor, Dog, CIGB-552.


Assuntos
Antineoplásicos/farmacologia , Peptídeos Penetradores de Células/farmacologia , Doenças do Cão/tratamento farmacológico , Neoplasias/veterinária , Animais , Antineoplásicos/administração & dosagem , Peptídeos Penetradores de Células/administração & dosagem , Cães , Avaliação Pré-Clínica de Medicamentos/veterinária , Feminino , Masculino , Neoplasias/tratamento farmacológico
5.
J Drug Target ; 25(6): 523-531, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28140690

RESUMO

Notch pathway was found to be activated in most glioblastomas (GBMs), underlining the importance of Notch in formation and recurrence of GBM. In this study, a Notch inhibitory peptide, dominant negative MAML (dnMAML), was conjugated to elastin-like polypeptide (ELP) for tumor targeted delivery. ELP is a thermally responsive polypeptide that can be actively and passively targeted to the tumor site by localized application of hyperthermia. This complex was further modified with the addition of a cell penetrating peptide, SynB1, for improved cellular uptake and blood-brain barrier penetration. The SynB1-ELP1-dnMAML was examined for its cellular uptake, cytotoxicity, apoptosis, cell cycle inhibition and the inhibition of target genes' expression. SynB1-ELP1-dnMAML inhibited the growth of D54 and U251 cells by inducing apoptosis and cell cycle arrest, especially in the presence of hyperthermia. Hyperthermia increased overall uptake of the polypeptide by the cells and enhanced the resulting pharmacological effects of dnMAML, showing the inhibition of targets of Notch pathway such as Hes-1 and Hey-L. These results confirm that dnMAML is an effective Notch inhibitor and combination with ELP may allow thermal targeting of the SynB1-ELP1-dnMAML complex in cancer cells while avoiding the dangers of systemic Notch inhibition.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Proteínas de Ligação a DNA/administração & dosagem , Glioblastoma/tratamento farmacológico , Receptores Notch/antagonistas & inibidores , Fatores de Transcrição/administração & dosagem , Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/farmacologia , Proteínas de Ligação a DNA/farmacocinética , Proteínas de Ligação a DNA/farmacologia , Sistemas de Liberação de Medicamentos , Elastina/administração & dosagem , Glioblastoma/patologia , Humanos , Hipertermia Induzida/métodos , Peptídeos/administração & dosagem , Fatores de Transcrição/farmacocinética , Fatores de Transcrição/farmacologia
6.
J Control Release ; 210: 134-46, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26003042

RESUMO

Delivery and penetration of chemotherapeutic drugs into neoplasm through the tumor vasculature are essential mechanisms to enhance the efficiency of chemotherapy. "Vascular targeting" strategy focuses on promoting the infiltration of chemotherapeutic drugs into neoplastic tissues. In this study, we achieved a targeted therapy by coupling tumor necrosis factor α (TNFα) with TCP-1, a novel vascular-targeting peptide, in an orthotopic colorectal cancer model in mice. High dose of TCP-1-conjugated TNFα (TCP-1/TNFα: 5µg/mouse) displayed potent antitumor activity by inducing apoptosis and reducing microvessel number in tumors than unconjugated TNFα, with no evidence of increased toxicity. In the combined therapy, the antitumor action of 5-fluorouracil (5-FU) was potentiated when the mice were pretreated with a low dose of TNFα (1ng/mouse) and to a greater extent by the same concentration of TCP-1/TNFα. In this regard, TCP-1/TNFα combined with 5-FU synergistically inhibited the tumor growth, induced apoptosis and reduced cell proliferation. More importantly, TCP-1/TNFα normalized the tumor vasculature and facilitated the infiltration of immune cells to neoplasm as well as attenuated the immunosuppressing effects of TNFα in bone marrow and spleen. At the same time, TCP-1/TNFα significantly improved 5-FU absorption into the tumor mass. Taken together, these findings underscore the therapeutic potential of TCP-1 as a drug carrier in cancer therapy. TCP-1 is a novel vascular-targeting peptide and appears to be a promising agent for drug delivery. TCP-1 fused with TNFα holds great promise for colorectal cancer therapy.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Peptídeos Penetradores de Células/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Fluoruracila/administração & dosagem , Neovascularização Patológica/tratamento farmacológico , Fator de Necrose Tumoral alfa/administração & dosagem , Animais , Antimetabólitos Antineoplásicos/química , Antimetabólitos Antineoplásicos/uso terapêutico , Medula Óssea/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/uso terapêutico , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Fluoruracila/química , Fluoruracila/uso terapêutico , Proteínas de Fluorescência Verde/administração & dosagem , Proteínas de Fluorescência Verde/química , Humanos , Masculino , Camundongos Endogâmicos BALB C , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Baço/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/uso terapêutico
8.
J Control Release ; 176: 123-132, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24374002

RESUMO

Red blood cells (RBCs) based drug carrier appears to be the most appealing for protein drugs due to their unmatched biocompatability, biodegradability, and long lifespan in the circulation. Numerous methods for encapsulating protein drugs into RBCs were developed, however, most of them induce partial disruption of the cell membrane, resulting in irreversible alterations in both physical and chemical properties of RBCs. Herein, we introduce a novel method for encapsulating proteins into intact RBCs, which was meditated by a cell penetrating peptide (CPP) developed in our lab-low molecular weight protamine (LMWP). l-asparaginase, one of the primary drugs used in treatment of acute lymphoblastic leukemia (ALL), was chosen as a model protein to illustrate the encapsulation into erythrocytes mediated by CPPs. In addition current treatment of ALL using different l-asparaginase delivery and encapsulation methods as well as their associated problems were also reviewed.


Assuntos
Antineoplásicos/administração & dosagem , Peptídeos Penetradores de Células/administração & dosagem , Portadores de Fármacos/administração & dosagem , Eritrócitos , Animais , Asparaginase/administração & dosagem , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico
9.
PLoS One ; 8(1): e55104, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23372821

RESUMO

Treatment of glioblastoma is complicated by the tumors' high resistance to chemotherapy, poor penetration of drugs across the blood brain barrier, and damaging effects of chemotherapy and radiation to normal neural tissue. To overcome these limitations, a thermally responsive polypeptide was developed for targeted delivery of therapeutic peptides to brain tumors using focused hyperthermia. The peptide carrier is based on elastin-like polypeptide (ELP), which is a thermally responsive biopolymer that forms aggregates above a characteristic transition temperature. ELP was modified with cell penetrating peptides (CPPs) to enhance delivery to brain tumors and mediate uptake across the tumor cells' plasma membranes and with a peptide inhibitor of c-Myc (H1). In rats with intracerebral gliomas, brain tumor targeting of ELP following systemic administration was enhanced up to 5-fold by the use of CPPs. When the lead CPP-ELP-fused c-Myc inhibitor was combined with focused hyperthermia of the tumors, an additional 3 fold increase in tumor polypeptide levels was observed, and 80% reduction in tumor volume, delayed onset of tumor-associated neurological deficits, and at least doubled median survival time including complete regression in 80% of animals was achieved. This work demonstrates that a c-Myc inhibitory peptide can be effectively delivered to brain tumors.


Assuntos
Antineoplásicos/farmacologia , Peptídeos Penetradores de Células/farmacologia , Glioma/terapia , Hipertermia Induzida , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Peptídeos Penetradores de Células/administração & dosagem , Peptídeos Penetradores de Células/química , Modelos Animais de Doenças , Progressão da Doença , Portadores de Fármacos , Glioma/mortalidade , Glioma/patologia , Temperatura Alta , Humanos , Raios Infravermelhos/uso terapêutico , Masculino , Transplante de Neoplasias , Ratos , Distribuição Tecidual
10.
Mol Cancer Ther ; 11(7): 1547-56, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22532601

RESUMO

Elastin-like polypeptides (ELP) aggregate in response to mild hyperthermia, but remain soluble under normal physiologic conditions. ELP macromolecules can accumulate in solid tumors because of the enhanced permeability and retention effect. Tumor retention of ELPs can be further enhanced through hyperthermia-induced aggregation of ELPs by local heating of the tumor. We evaluated the therapeutic potential of ELPs in delivering doxorubicin in the E0771 syngeneic mouse breast cancer model. The ELP-Dox conjugate consisted of a cell-penetrating peptide at the N-terminus and the 6-maleimidocaproyl hydrazone derivative of doxorubicin at the C-terminus of ELP. The acid-sensitive hydrazone linker ensured release of doxorubicin in the lysosomes/endosomes after cellular uptake of the drug conjugate. ELP-Dox dosed at 5 mg doxorubicin equivalent/kg, extended the plasma half-life of doxorubicin to 5.5 hours. In addition, tumor uptake of ELP-Dox increased 2-fold when hyperthermia was applied, and was also enhanced compared to free doxorubicin. Although high levels of doxorubicin were found in the heart of animals treated with free doxorubicin, no detectable levels of doxorubicin were found in ELP-Dox-treated animals, indicating a correlation between tumor targeting and reduction of potential cardiac toxicity by ELP-Dox. At an optimal dose of 12 mg doxorubicin equivalent/kg, ELP-Dox in combination with hyperthermia induced a complete tumor growth inhibition, which was distinctly superior to free drug that only moderately inhibited tumor growth. In summary, our findings show that thermal targeting of ELP increases the potency of doxorubicin underlying the potential of exploiting ELPs to enhance the therapeutic efficacy of conventional anticancer drugs.


Assuntos
Peptídeos Penetradores de Células/farmacologia , Doxorrubicina/química , Doxorrubicina/farmacologia , Elastina/química , Hipertermia Induzida , Animais , Transporte Biológico , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/administração & dosagem , Peptídeos Penetradores de Células/química , Terapia Combinada , Modelos Animais de Doenças , Doxorrubicina/administração & dosagem , Feminino , Humanos , Dose Máxima Tolerável , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/terapia
11.
PLoS One ; 7(1): e30527, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22291977

RESUMO

BACKGROUND: In the last ten years, bioterrorism has become a serious threat and challenge to public health worldwide. Pulmonary anthrax caused by airborne Bacillus anthracis spores is a life-threatening disease often refractory to antimicrobial therapy. Inhaled spores germinate into vegetative forms that elaborate an anti-phagocytic capsule along with potent exotoxins which disrupt the signaling pathways governing the innate and adaptive immune responses and cause endothelial cell dysfunction leading to vascular injury in the lung, hypoxia, hemorrhage, and death. METHODS/PRINCIPAL FINDINGS: Using a murine model of pulmonary anthrax disease, we showed that a nuclear transport modifier restored markers of the innate immune response in spore-infected animals. An 8-day protocol of single-dose ciprofloxacin had no significant effect on mortality (4% survival) of A/J mice lethally infected with B. anthracis Sterne. Strikingly, mice were much more likely to survive infection (52% survival) when treated with ciprofloxacin and a cell-penetrating peptide modifier of host nuclear transport, termed cSN50. In B. anthracis-infected animals treated with antibiotic alone, we detected a muted innate immune response manifested by cytokines, tumor necrosis factor alpha (TNFα), interleukin (IL)-6, and chemokine monocyte chemoattractant protein-1 (MCP-1), while the hypoxia biomarker, erythropoietin (EPO), was greatly elevated. In contrast, cSN50-treated mice receiving ciprofloxacin demonstrated a restored innate immune responsiveness and reduced EPO level. Consistent with this improvement of innate immunity response and suppression of hypoxia biomarker, surviving mice in the combination treatment group displayed minimal histopathologic signs of vascular injury and a marked reduction of anthrax bacilli in the lungs. CONCLUSIONS: We demonstrate, for the first time, that regulating nuclear transport with a cell-penetrating modifier provides a cytoprotective effect, which enables the host's immune system to reduce its susceptibility to lethal B. anthracis infection. Thus, by combining a nuclear transport modifier with antimicrobial therapy we offer a novel adjunctive measure to control florid pulmonary anthrax disease.


Assuntos
Antraz/tratamento farmacológico , Antraz/mortalidade , Anti-Infecciosos/administração & dosagem , Pneumopatias/tratamento farmacológico , Pneumopatias/mortalidade , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Antraz/complicações , Antraz/patologia , Anti-Infecciosos/farmacologia , Peptídeos Penetradores de Células/administração & dosagem , Peptídeos Penetradores de Células/farmacologia , Ciprofloxacina/administração & dosagem , Ciprofloxacina/farmacologia , Citocinas/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Combinação de Medicamentos , Feminino , Pneumopatias/etiologia , Pneumopatias/patologia , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/farmacologia , Camundongos , Peptídeos Cíclicos/administração & dosagem , Peptídeos Cíclicos/farmacologia , Análise de Sobrevida , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA