Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Clin Immunol ; 235: 108785, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34147650

RESUMO

The FDA approval of pegcetacoplan (Empaveli), a PEGylated compstatin-based C3 therapeutic, as a new treatment for paroxysmal nocturnal hemoglobinuria (PNH) marks a milestone in the history of complement drug discovery. Almost 15 years after the approval of the first complement-specific drug for PNH, the anti-C5 antibody eculizumab, a novel class of complement inhibitors with a distinct mechanism of action finally enters the clinic. This landmark decision broadens the spectrum of available complement therapeutics, offering patients with unmet clinical needs or insufficient responses to anti-C5 therapy an alternative treatment option with a broad activity profile. Here we present a brief historical account of this newly approved complement drug, consolidating its approval within the long research record of the compstatin family of peptidic C3 inhibitors.


Assuntos
Complemento C3/antagonistas & inibidores , Hemoglobinúria Paroxística/tratamento farmacológico , Peptídeos Cíclicos/farmacologia , Complemento C3/metabolismo , Aprovação de Drogas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Peptídeos Cíclicos/química
2.
Proc Natl Acad Sci U S A ; 116(20): 9953-9958, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31036650

RESUMO

The complement system is highly efficient in targeting pathogens, but lack of its apposite regulation results in host-cell damage, which is linked to diseases. Thus, complement activation is tightly regulated by a series of proteins, which primarily belong to the regulators of complement activation (RCA) family. Structurally, these proteins are composed of repeating complement control protein (CCP) domains where two to four successive domains contribute to the regulatory functions termed decay-accelerating activity (DAA) and cofactor activity (CFA). However, the precise constitution of the functional units and whether these units can be joined to form a larger composition with dual function have not been demonstrated. Herein, we have parsed the functional units for DAA and CFA by constructing chimeras of the decay-accelerating factor (DAF) that exhibits DAA and membrane cofactor protein (MCP) that exhibits CFA. We show that in a four-CCP framework, a functional unit for each of the regulatory activities is formed by only two successive CCPs wherein each participates in the function, albeit CCP2 has a bipartite function. Additionally, optimal activity requires C-terminal domains that enhance the avidity of the molecule for C3b/C4b. Furthermore, by composing a four-CCP DAF-MCP chimera with robust CFA (for C3b and C4b) and DAA (for classical and alternative pathway C3 convertases), named decay cofactor protein, we show that CCP functional units can be linked to design a dual-activity regulator. These data indicate that the regulatory determinants for these two biological processes are distinct and modular in nature.


Assuntos
Proteínas do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento/genética , Escherichia coli , Humanos , Proteína Cofatora de Membrana , Pichia , Domínios Proteicos , Engenharia de Proteínas , Estrutura Quaternária de Proteína
3.
BMC Palliat Care ; 19(1): 14, 2020 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-31987038

RESUMO

BACKGROUND: The therapeutic landscape in medical oncology continues to expand significantly. Newer therapies, especially immunotherapy, offer the hope of profound and durable responses with more tolerable side effect profiles. Integrating this information into the decision making process is challenging for patients and oncologists. Systemic anticancer treatment within the last thirty days of life is a key quality of care indicator and is one parameter used in the assessment of aggressiveness of care. METHODS: A retrospective review of medical records of all patients previously treated at Goulburn Valley Health oncology department who died between 1 January 2015 and 30 June 2018 was conducted. Information collected related to patient demographics, diagnosis, treatment, and hospital care within the last 30 days of life. These results were presented to the cancer services meeting and a quality improvement intervention program was instituted. A second retrospective review of medical records of all patients who died between 1 July 2018 and 31 December 2018 was conducted in order to measure the effect of this intervention. RESULTS: The initial audit period comprised 440 patients. 120 patients (27%) received treatment within the last 30 days of life. The re-audit period comprised 75 patients. 19 patients (25%) received treatment within the last 30 days of life. Treatment rates of chemotherapy reduced after the intervention in contrast to treatment rates of immunotherapy which increased. A separate analysis calculated the rate of mortality within 30 days of chemotherapy from the total number of patients who received chemotherapy was initially 8% and 2% in the re-audit period. Treatment within the last 30 days of life was associated with higher use of aggressive care such as emergency department presentation, hospitalisation, ICU admission and late hospice referral. Palliative care referral rates improved after the intervention. CONCLUSION: This audit demonstrated that a quality improvement intervention can impact quality of care indicators with reductions in the use of chemotherapy within the last 30 days of life. However, immunotherapy use increased which may be explained by increased access and a better risk benefit balance.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Adulto , Idoso , Institutos de Câncer/organização & administração , Institutos de Câncer/estatística & dados numéricos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/psicologia , Serviço Hospitalar de Oncologia/organização & administração , Serviço Hospitalar de Oncologia/estatística & dados numéricos , Cuidados Paliativos/métodos , Melhoria de Qualidade , Estudos Retrospectivos
4.
J Biol Chem ; 293(40): 15538-15555, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30139746

RESUMO

Innate immunity in animals including humans encompasses the complement system, which is considered an important host defense mechanism against Aspergillus fumigatus, one of the most ubiquitous opportunistic human fungal pathogens. Previously, it has been shown that the alkaline protease Alp1p secreted from A. fumigatus mycelia degrades the complement components C3, C4, and C5. However, it remains unclear how the fungal spores (i.e. conidia) defend themselves against the activities of the complement system immediately after inhalation into the lung. Here, we show that A. fumigatus conidia contain a metalloprotease Mep1p, which is released upon conidial contact with collagen and inactivates all three complement pathways. In particular, Mep1p efficiently inactivated the major complement components C3, C4, and C5 and their activation products (C3a, C4a, and C5a) as well as the pattern-recognition molecules MBL and ficolin-1, either by directly cleaving them or by cleaving them to a form that is further broken down by other proteases of the complement system. Moreover, incubation of Mep1p with human serum significantly inhibited the complement hemolytic activity and conidial opsonization by C3b and their subsequent phagocytosis by macrophages. Together, these results indicate that Mep1p associated with and released from A. fumigatus conidia likely facilitates early immune evasion by disarming the complement defense in the human host.


Assuntos
Aspergillus fumigatus/imunologia , Complemento C3/genética , Complemento C4/genética , Complemento C5/genética , Aspergilose Pulmonar Invasiva/imunologia , Metaloendopeptidases/imunologia , Animais , Aspergillus fumigatus/crescimento & desenvolvimento , Aspergillus fumigatus/patogenicidade , Colágeno/genética , Colágeno/imunologia , Complemento C3/metabolismo , Complemento C4/metabolismo , Complemento C5/metabolismo , Modelos Animais de Doenças , Proteínas Fúngicas/genética , Proteínas Fúngicas/imunologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Imunidade Inata , Aspergilose Pulmonar Invasiva/genética , Aspergilose Pulmonar Invasiva/microbiologia , Aspergilose Pulmonar Invasiva/patologia , Lectinas/genética , Lectinas/imunologia , Pulmão/imunologia , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Serina Proteases Associadas a Proteína de Ligação a Manose/genética , Serina Proteases Associadas a Proteína de Ligação a Manose/imunologia , Metaloendopeptidases/deficiência , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fagocitose , Esporos Fúngicos/crescimento & desenvolvimento , Esporos Fúngicos/imunologia , Esporos Fúngicos/patogenicidade , Ficolinas
5.
J Biol Chem ; 293(13): 4901-4912, 2018 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-29414772

RESUMO

Surfactant protein D (SP-D), a C-type lectin and pattern-recognition soluble factor, plays an important role in immune surveillance to detect and eliminate human pulmonary pathogens. SP-D has been shown to protect against infections with the most ubiquitous airborne fungal pathogen, Aspergillus fumigatus, but the fungal surface component(s) interacting with SP-D is unknown. Here, we show that SP-D binds to melanin pigment on the surface of A. fumigatus dormant spores (conidia). SP-D also exhibited an affinity to two cell-wall polysaccharides of A. fumigatus, galactomannan (GM) and galactosaminogalactan (GAG). The immunolabeling pattern of SP-D was punctate on the conidial surface and was uniform on germinating conidia, in accordance with the localization of melanin, GM, and GAG. We also found that the collagen-like domain of SP-D is involved in its interaction with melanin, whereas its carbohydrate-recognition domain recognized GM and GAG. Unlike un-opsonized conidia, SP-D-opsonized conidia were phagocytosed more efficiently and stimulated the secretion of proinflammatory cytokines by human monocyte-derived macrophages. Furthermore, SP-D-/- mice challenged intranasally with wildtype conidia or melanin ghosts (i.e. hollow melanin spheres) displayed significantly reduced proinflammatory cytokines in the lung compared with wildtype mice. In summary, SP-D binds to melanin present on the dormant A. fumigatus conidial surface, facilitates conidial phagocytosis, and stimulates the host immune response.


Assuntos
Aspergillus fumigatus/imunologia , Polissacarídeos Fúngicos/imunologia , Melaninas/imunologia , Fagocitose , Aspergilose Pulmonar/imunologia , Proteína D Associada a Surfactante Pulmonar/imunologia , Esporos Fúngicos/imunologia , Animais , Aspergillus fumigatus/genética , Polissacarídeos Fúngicos/genética , Melaninas/genética , Camundongos , Camundongos Knockout , Aspergilose Pulmonar/genética , Aspergilose Pulmonar/patologia , Proteína D Associada a Surfactante Pulmonar/genética , Esporos Fúngicos/genética
6.
PLoS Pathog ; 13(3): e1006248, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28301559

RESUMO

The pandemic influenza A(H1N1) 2009 virus caused significant morbidity and mortality worldwide thus necessitating the need to understand the host factors that influence its control. Previously, the complement system has been shown to provide protection during the seasonal influenza virus infection, however, the role of individual complement pathways is not yet clear. Here, we have dissected the role of intact complement as well as of its individual activation pathways during the pandemic influenza virus infection using mouse strains deficient in various complement components. We show that the virus infection in C3-/- mice results in increased viral load and 100% mortality, which can be reversed by adoptive transfer of naïve wild-type (WT) splenocytes, purified splenic B cells, or passive transfer of immune sera from WT, but not C3-/- mice. Blocking of C3a and/or C5a receptor signaling in WT mice using receptor antagonists and use of C3aR-/- and C5aR-/- mice showed significant mortality after blocking/ablation of C3aR, with little or no effect after blocking/ablation of C5aR. Intriguingly, deficiency of C4 and FB in mice resulted in only partial mortality (24%-32%) suggesting a necessary cross-talk between the classical/lectin and alternative pathways for providing effective protection. In vitro virus neutralization experiments performed to probe the cross-talk between the various pathways indicated that activation of the classical and alternative pathways in concert, owing to coating of viral surface by antibodies, is needed for its efficient neutralization. Examination of the virus-specific complement-binding antibodies in virus positive subjects showed that their levels vary among individuals. Together these results indicate that cooperation between the classical and alternative pathways not only result in efficient direct neutralization of the pandemic influenza virus, but also lead to the optimum generation of C3a, which when sensed by the immune cells along with the antigen culminates in generation of effective protective immune responses.


Assuntos
Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Infecções por Orthomyxoviridae/imunologia , Transferência Adotiva , Animais , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Influenza Humana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Testes de Neutralização
7.
Immunology ; 2018 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-29485195

RESUMO

We previously reported that Indian paediatric patients with atypical haemolytic-uraemic syndrome (aHUS) showed high frequencies of anti-complement factor H (FH) autoantibodies that are correlated with homozygous deletion of the genes for FH-related proteins 1 and 3 (FHR1 and FHR3) (FHR1/3-/- ). We now report that Indian paediatric aHUS patients without anti-FH autoantibodies also showed modestly higher frequencies of the FHR1/3-/- genotype. Further, when we characterized epitope specificities and binding avidities of anti-FH autoantibodies in aHUS patients, most anti-FH autoantibodies were directed towards the FH cell-surface anchoring polyanionic binding site-containing C-terminal short conservative regions (SCRs) 17-20 with higher binding avidities than for native FH. FH SCR17-20-binding anti-FH autoantibodies also bound the other cell-surface anchoring polyanionic binding site-containing region FH SCR5-8, at lower binding avidities. Anti-FH autoantibody avidities correlated with antibody titres. These anti-FH autoantibody characteristics did not differ between aHUS patients with or without the FHR1/3-/- genotype. Our data suggest a complex matrix of interactions between FHR1-FHR3 deletion, immunomodulation and anti-FH autoantibodies in the aetiopathogenesis of aHUS.

8.
J Virol ; 91(19)2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28724763

RESUMO

Poxviruses display species tropism-variola virus is a human-specific virus, while vaccinia virus causes repeated outbreaks in dairy cattle. Consistent with this, variola virus complement regulator SPICE (smallpox inhibitor of complement enzymes) exhibits selectivity in inhibiting the human alternative complement pathway and vaccinia virus complement regulator VCP (vaccinia virus complement control protein) displays selectivity in inhibiting the bovine alternative complement pathway. In the present study, we examined the species specificity of VCP and SPICE for the classical pathway (CP). We observed that VCP is ∼43-fold superior to SPICE in inhibiting bovine CP. Further, functional assays revealed that increased inhibitory activity of VCP for bovine CP is solely due to its enhanced cofactor activity, with no effect on decay of bovine CP C3-convertase. To probe the structural basis of this specificity, we utilized single- and multi-amino-acid substitution mutants wherein 1 or more of the 11 variant VCP residues were substituted in the SPICE template. Examination of these mutants for their ability to inhibit bovine CP revealed that E108, E120, and E144 are primarily responsible for imparting the specificity and contribute to the enhanced cofactor activity of VCP. Binding and functional assays suggested that these residues interact with bovine factor I but not with bovine C4(H2O) (a moiety conformationally similar to C4b). Mapping of these residues onto the modeled structure of bovine C4b-VCP-bovine factor I supported the mutagenesis data. Taken together, our data help explain why the vaccine strain of vaccinia virus was able to gain a foothold in domesticated animals.IMPORTANCE Vaccinia virus was used for smallpox vaccination. The vaccine-derived virus is now circulating and causing outbreaks in dairy cattle in India and Brazil. However, the reason for this tropism is unknown. It is well recognized that the virus is susceptible to neutralization by the complement classical pathway (CP). Because the virus encodes a soluble complement regulator, VCP, we examined whether this protein displays selectivity in targeting bovine CP. Our data show that it does exhibit selectivity in inhibiting the bovine CP and that this is primarily determined by its amino acids E108, E120, and E144, which interact with bovine serine protease factor I to inactivate bovine C4b-one of the two subunits of CP C3-convertase. Of note, the variola complement regulator SPICE contains positively charged residues at these positions. Thus, these variant residues in VCP help enhance its potency against the bovine CP and thereby the fitness of the virus in cattle.


Assuntos
Ativação do Complemento/imunologia , Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/imunologia , Proteínas da Matriz Viral/imunologia , Proteínas Virais/imunologia , Tropismo Viral/genética , Sequência de Aminoácidos , Animais , Bovinos , Proteína de Ligação ao Complemento C4b/imunologia , Fibrinogênio/metabolismo , Humanos , Alinhamento de Sequência , Especificidade da Espécie , Vaccinia virus/imunologia , Vaccinia virus/patogenicidade , Proteínas da Matriz Viral/genética , Proteínas Virais/genética
9.
Indian J Med Res ; 148(3): 284-290, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30425218

RESUMO

BACKGROUND & OBJECTIVES: Gemcitabine combined with non-cremophor-based paclitaxel is one of the standards of care in advanced inoperable pancreatic cancer. This study was undertaken to retrospectively evaluate real world non-trial outcomes with this combination. METHODS: Patients with histologically proven advanced inoperable pancreatic adenocarcinoma (PDAC), treated with non-cremophor-based paclitaxel-gemcitabine combination (PG) (gemcitabine-nanoxel or gemcitabine-abraxane) between January 2012 and June 2015, were retrospectively analyzed. Response assessment was done every 8-12 wk with computed tomography scan and responses were measured as per the Response Evaluation Criteria in Solid Tumours 1.1 criteria where feasible. Toxicity was recorded as per the Common Terminology Criteria for Adverse Events (CTCAE) v4 criteria. Progression-free survival (PFS) and overall survival (OS) were calculated using the Kaplan-Meier method. RESULTS: A total of 78 patients with PDAC were treated with the combination. Of these, 83.3 per cent of patients had metastatic disease. The median number of chemotherapy cycles administered was three. The objective response rate for the whole group was 30.8 per cent. Grade III/IV toxicities were seen in 35.9 per cent of patients. Median PFS was 5.6 months and median OS was 11.6 months. INTERPRETATION & CONCLUSIONS: Non-cremophor-based paclitaxel in combination with gemcitabine appeared efficacious for advanced pancreatic cancers in routine clinical practice. Within the confines of a single-centre retrospective analysis, gemcitabine-nanoxel and gemcitabine-abraxane appeared to have similar efficacy and toxicity in advanced pancreatic cancers.


Assuntos
Desoxicitidina/análogos & derivados , Paclitaxel , Neoplasias Pancreáticas , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Desoxicitidina/administração & dosagem , Desoxicitidina/efeitos adversos , Esquema de Medicação , Monitoramento de Medicamentos , Feminino , Humanos , Índia , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Estudos Retrospectivos , Resultado do Tratamento , Gencitabina , Neoplasias Pancreáticas
10.
Proc Natl Acad Sci U S A ; 112(41): 12794-9, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26420870

RESUMO

The complement system has evolved to annul pathogens, but its improper regulation is linked with diseases. Efficient regulation of the system is primarily provided by a family of proteins termed regulators of complement activation (RCA). The knowledge of precise structural determinants of RCA proteins critical for imparting the regulatory activities and the molecular events underlying the regulatory processes, nonetheless, is still limited. Here, we have dissected the structural requirements of RCA proteins that are crucial for one of their two regulatory activities, the cofactor activity (CFA), by using the Kaposi's sarcoma-associated herpesvirus RCA homolog Kaposica as a model protein. We have scanned the entire Kaposica molecule by sequential mutagenesis using swapping and site-directed mutagenesis, which identified residues critical for its interaction with C3b and factor I. Mapping of these residues onto the modeled structure of C3b-Kaposica-factor I complex supported the mutagenesis data. Furthermore, the model suggested that the C3b-interacting residues bridge the CUB (complement C1r-C1s, Uegf, Bmp1) and MG2 (macroglobulin-2) domains of C3b. Thus, it seems that stabilization of the CUB domain with respect to the core of the C3b molecule is central for its CFA. Identification of CFA-critical regions in Kaposica guided experiments in which the equivalent regions of membrane cofactor protein were swapped into decay-accelerating factor. This strategy allowed CFA to be introduced into decay-accelerating factor, suggesting that viral and human regulators use a common mechanism for CFA.


Assuntos
Complemento C3b/química , Fator I do Complemento/química , Herpesvirus Humano 8/química , Modelos Moleculares , Complexos Multiproteicos/química , Proteínas Virais/química , Complemento C3b/genética , Fator I do Complemento/genética , Herpesvirus Humano 8/genética , Humanos , Complexos Multiproteicos/genética , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas Virais/genética
11.
HPB (Oxford) ; 20(9): 841-847, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29706425

RESUMO

BACKGROUND: Studies evaluating neo-adjuvant chemotherapy (NACT) exclusively in gallbladder cancer (GBC) are few and there are no randomized trials on the subject. Locally advanced GBC and indications for NACT in GBC are not yet clearly defined. METHODS: We analysed 160 consecutive GBC patients who received NACT based on clinico-radiologic criteria suggesting high-risk disease (TMH Criteria) from January 2010 to February 2016. RESULTS: On initial assessment, 140 (87.5%) patients had T3/T4 disease and 105 (65%) patients were node positive. Response rate and clinical benefit rate was 52.5% and 70% respectively. Sixty six (41.2%) patients could undergo curative intent resection. With a median follow-up of 33 months, the median OS and EFS of the entire cohort were 13 and 8 months respectively. Patient undergoing curative surgery had a statistically superior OS (49 vs. 7 months; p = 0.0001) and EFS (25 months vs. 5 months; p = 0.0001) compared to those who did not. CONCLUSION: Locally advanced GBC remains a disease with poor prognosis. Chemotherapy with neoadjuvant intent in locally advanced/borderline resectable GBC showed good response rates. This resulted in curative surgical resection or disease stabilisation in significant proportion of patients. Patients who undergo definitive surgery after favourable response to NACT experience good survival.


Assuntos
Colecistectomia , Neoplasias da Vesícula Biliar/terapia , Terapia Neoadjuvante , Adulto , Idoso , Quimioterapia Adjuvante , Colecistectomia/efeitos adversos , Colecistectomia/mortalidade , Bases de Dados Factuais , Feminino , Neoplasias da Vesícula Biliar/diagnóstico por imagem , Neoplasias da Vesícula Biliar/mortalidade , Neoplasias da Vesícula Biliar/patologia , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante/efeitos adversos , Terapia Neoadjuvante/mortalidade , Estadiamento de Neoplasias , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
12.
Toxicol Mech Methods ; 26(5): 319-26, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27101948

RESUMO

Organophosphate (OP)-based pesticides and nerve agents are highly toxic compounds which interrupt the catalytic mechanism of acetylcholinesterase (AChE) by phosphorylating the hydroxyl moiety of serine residue. The inhibited enzyme can be reactivated by the nucleophilic action of oxime reactivators. To analyze the effect of different AChE sources on reactivation efficacy of reactivators, several in vivo studies have carried out using variety of AChE sources like pig, rat and monkey. Investigations on species differences provide a better insight for the development of new reactivators. Hence, present study was mainly targeted on comparative analysis of the reactivation of electric eel and human AChE inhibited by different OP. A series of butene-linked bis-pyridinium mono oximes which vary in functional groups present at the second pyridinium ring have been examined against sarin, VX, tabun and ethyl-paraoxon-poisoned AChE. In case of tabun-inhibited AChEs, tested oximes were better than reference oximes. For VX-poisoned human AChE, reactivator K251 (kr2;1.51 mM (-) (1 )min (-) (1)) showed good reactivation efficacy with standard oximes. Studies stipulated that butene-linked oximes consisting of different functional moieties are good reactivators and found to have better efficacy to reactivate nerve agent-inhibited human AChE in comparison to eel AChE.


Assuntos
Acetilcolinesterase/metabolismo , Inibidores da Colinesterase/farmacologia , Reativadores da Colinesterase/farmacologia , Membrana Eritrocítica/enzimologia , Organofosfatos/farmacologia , Oximas/farmacologia , Adulto , Animais , Relação Dose-Resposta a Droga , Electrophorus , Membrana Eritrocítica/efeitos dos fármacos , Humanos , Cinética , Masculino , Fatores de Tempo
13.
Org Biomol Chem ; 13(10): 2827-48, 2015 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-25597899

RESUMO

Owing to the rising threats of neurotoxic organophosphosphorus compounds, facile and efficient decontamination systems are required. Since the last few decades, the search for promising α-nucleophiles for straightforward and eco-friendly decontamination reactions using α-nucleophiles has been considerably boosted up. Among these, hydroxamic acids have been widely studied due to their potential α-nucleophilicity towards carbon and phosphorus based esters. This account summarizes our research on α-nucleophilicity of hydroxamate ions in water and micelles towards esterolytic reactions. Efforts of our group in the last few years have been collectively judged and compared with the crucial findings of researchers in the relevant field. The present article sheds light on the rich chemistry of the hydroxamate ion as a perfect candidate to degrade organophosphorus esters (i.e. nerve agents, pesticides and their simulants) in water, in micelles of conventional surfactants, and in functionalized micelles. The current report also provides an insight into the possible nature and mechanisms of these reactions. A brief account of the biological activities of hydroxamic acids that have recently spurred research in medicine against some fatal diseases has been included.


Assuntos
Química Orgânica/métodos , Ésteres/química , Ácidos Hidroxâmicos/química , Íons/química , Micelas , Organofosfatos/química , Carbono/química , Ácidos Carboxílicos/química , Catálise , Concentração de Íons de Hidrogênio , Cinética , Fosfatos , Solventes/química , Ácidos Sulfônicos/química , Tensoativos/química , Água/química
14.
J Virol ; 87(1): 282-95, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23077301

RESUMO

Herpesvirus saimiri is known to encode a homolog of human complement regulators named complement control protein homolog (CCPH). We have previously reported that this virally encoded inhibitor effectively inactivates complement by supporting factor I-mediated inactivation of complement proteins C3b and C4b (termed cofactor activity), as well as by accelerating the irreversible decay of the classical/lectin and alternative pathway C3 convertases (termed decay-accelerating activity). To fine map its functional sites, in the present study, we have generated a homology model of CCPH and performed substitution mutagenesis of its conserved residues. Functional analyses of 24 substitution mutants of CCPH indicated that (i) amino acids R118 and F144 play a critical role in imparting C3b and C4b cofactor activities, (ii) amino acids R35, K142, and K191 are required for efficient decay of the C3 convertases, (iii) positively charged amino acids of the linker regions, which are dubbed to be critical for functioning in other complement regulators, are not crucial for its function, and (iv) S100K and G110D mutations substantially enhance its decay-accelerating activities without affecting the cofactor activities. Overall, our data point out that ionic interactions form a major component of the binding interface between CCPH and its interacting partners.


Assuntos
Fator I do Complemento/genética , Fator I do Complemento/metabolismo , Herpesvirus Saimiriíneo 2/imunologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Substituição de Aminoácidos , Análise Mutacional de DNA , Herpesvirus Saimiriíneo 2/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica
15.
J Immunol ; 189(3): 1431-9, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22732591

RESUMO

Variola and vaccinia viruses, the two most important members of the family Poxviridae, are known to encode homologs of the human complement regulators named smallpox inhibitor of complement enzymes (SPICE) and vaccinia virus complement control protein (VCP), respectively, to subvert the host complement system. Intriguingly, consistent with the host tropism of these viruses, SPICE has been shown to be more human complement-specific than VCP, and in this study we show that VCP is more bovine complement-specific than SPICE. Based on mutagenesis and mechanistic studies, we suggest that the major determinant for the switch in species selectivity of SPICE and VCP is the presence of oppositely charged residues in the central complement control modules, which help enhance their interaction with factor I and C3b, the proteolytically cleaved form of C3. Thus, our results provide a molecular basis for the species selectivity in poxviral complement regulators.


Assuntos
Complemento C3b/antagonistas & inibidores , Via Alternativa do Complemento/imunologia , Vaccinia virus/imunologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Substituição de Aminoácidos/imunologia , Animais , Bovinos , Complemento C3b/genética , Complemento C3b/metabolismo , Fator I do Complemento/antagonistas & inibidores , Fator I do Complemento/metabolismo , Via Alternativa do Complemento/genética , Humanos , Evasão da Resposta Imune/genética , Dados de Sequência Molecular , Especificidade da Espécie , Eletricidade Estática , Vaccinia virus/metabolismo , Proteínas Virais/genética
16.
J Gastrointest Oncol ; 15(1): 63-85, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38482233

RESUMO

Background: The discovery of biomarkers has facilitated the treatment of cancer. At present, the relationship between activin A receptor type-1 (ACVR1) and gastric cancer is gradually discovered. The aim of this study was to explore the expression of ACVR1 in gastric cancer and its clinical significance, to study the relationship between ACVR1 and tumor microenvironment (TME) for the prognosis of gastric cancer, and to further identify new targets for immunotherapy in gastric cancer. Methods: ACVR1 was first selected as a study gene according to several cancer and gastric cancer public datasets. Its pancancer expression was explored using the UCSC Xena database. The expression level, prognosis, and clinicopathological features of ACVR1 in gastric cancer were analyzed using The Cancer Genome Atlas (TCGA) database. Immunohistochemistry (IHC)-based experiments were conducted to study the expression of ACVR1 at the protein level. The IHC data were analyzed for correlations between ACVR1 expression and various clinicopathological factors and prognosis. The correlation of this gene with the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, immune infiltration, immune checkpoints, drug therapy, tumor mutation burden (TMB), microsatellite instability (MSI), and mismatch repair (MMR) system was analyzed using R software. Results: TCGA data showed that the expression of ACVR1 was higher in gastric cancer tissues than in paracancerous tissues. Moreover, the IHC experiments indicated that ACVR1 was upregulated in gastric cancer tissues at the protein level. Both univariate Cox and multivariate Cox results showed that the increase of ACVR1 was closely associated with tumor stage, size, lymph node metastasis, and age. High ACVR1 expression was linked to a poor prognosis of gastric cancer. The results also revealed that ACVR1 was closely related to suppressive immune cells and pathways. Analyses of immune checkpoints, antitumor drug, TMB, and immune microenvironment indicated that ACVR1 had an antitumor immune effect, promoting gastric cancer development and leading to poor immunotherapy. Conclusions: High ACVR1 expression can be used as an independent prognostic factor to predict the prognostic survival of patients with gastric cancer. ACVR1 expression in gastric cancer tissues was significantly correlated with immune infiltration and may thus serve as a potential therapeutic target for gastric cancer immunotherapy.

17.
J Immunol ; 184(12): 7116-24, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20483772

RESUMO

Complement factor B (fB) is a key constituent of the alternative pathway (AP). Its central role in causing inflammation and tissue injury through activation of the AP urges the need for its therapeutic targeting. In the current study, we have screened phage-displayed random peptide libraries against fB and identified a novel cyclic hendecapeptide that inhibits activation of fB and the AP. Structure-activity studies revealed that: 1) the cysteine-constrained structure of the peptide is essential for its activity; 2) Ile5, Arg6, Leu7, and Tyr8 contribute significantly to its inhibitory activity; and 3) retro-inverso modification of the peptide results in loss of its activity. Binding studies performed using surface plasmon resonance suggested that the peptide has two binding sites on fB, which are located on the Ba and Bb fragments. Studies on the mechanism of inhibition revealed that the peptide does not block the interaction of fB with the activated form of C3, thereby suggesting that the peptide inhibits fB activation primarily by inhibiting its cleavage by factor D. The peptide showed a weak effect on preformed C3 and C5 convertases. Like inhibition of fB cleavage, the peptide also inhibited C2 cleavage by activated C1s and activation of the classical as well as lectin pathways. Based on its inhibitory activities, we named the peptide Complin.


Assuntos
Complemento C2/metabolismo , Fator B do Complemento/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Via Alternativa do Complemento/fisiologia , Sequência de Aminoácidos , Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/genética , Ensaio de Imunoadsorção Enzimática , Humanos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Estrutura Quaternária de Proteína , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
18.
J Immunol ; 185(10): 6128-37, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20956343

RESUMO

Vaccinia virus encodes a structural and functional homolog of human complement regulators named vaccinia virus complement control protein (VCP). This four-complement control protein domain containing secretory protein is known to inhibit complement activation by supporting the factor I-mediated inactivation of complement proteins, proteolytically cleaved form of C3 (C3b) and proteolytically cleaved form of C4 (C4b) (termed cofactor activity), and by accelerating the irreversible decay of the classical and to a limited extent of the alternative pathway C3 convertases (termed decay-accelerating activity [DAA]). In this study, we have mapped the VCP domains important for its cofactor activity and DAA by swapping its individual domains with those of human decay-accelerating factor (CD55) and membrane cofactor protein (MCP; CD46). Our data indicate the following: 1) swapping of VCP domain 2 or 3, but not 1, with homologous domains of decay-accelerating factor results in loss in its C3b and C4b cofactor activities; 2) swapping of VCP domain 1, but not 2, 3, or 4 with corresponding domains of MCP results in abrogation in its classical pathway DAA; and 3) swapping of VCP domain 1, 2, or 3, but not 4, with homologous MCP domains have marked effect on its alternative pathway DAA. These functional data together with binding studies with C3b and C4b suggest that in VCP, domains 2 and 3 provide binding surface for factor I interaction, whereas domain 1 mediates dissociation of C2a and Bb from the classical and alternative pathway C3 convertases, respectively.


Assuntos
Proteínas do Sistema Complemento/metabolismo , Vaccinia virus/química , Vaccinia virus/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Antígenos CD55/química , Antígenos CD55/imunologia , Antígenos CD55/metabolismo , Proteínas do Sistema Complemento/química , Proteínas do Sistema Complemento/imunologia , Eletroforese em Gel de Poliacrilamida , Humanos , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície , Vaccinia virus/imunologia , Proteínas Virais/imunologia
19.
J Immunol ; 184(4): 1956-67, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20089702

RESUMO

Kaposica, the complement regulator of Kaposi's sarcoma-associated herpesvirus, inhibits complement by supporting factor I-mediated inactivation of the proteolytically activated form of C3 (C3b) and C4 (C4b) (cofactor activity [CFA]) and by accelerating the decay of classical and alternative pathway C3-convertases (decay-accelerating activity [DAA]). Previous data suggested that electrostatic interactions play a critical role in the binding of viral complement regulators to their targets, C3b and C4b. We therefore investigated how electrostatic potential on Kaposica influences its activities. We built a homology structure of Kaposica and calculated the electrostatic potential of the molecule, using the Poisson-Boltzmann equation. Mutants were then designed to alter the overall positive potential of the molecule or of each of its domains and linkers by mutating Lys/Arg to Glu/Gln, and the functional activities of the expressed mutants were analyzed. Our data indicate that 1) positive potential at specific sites and not the overall positive potential on the molecule guides the CFAs and classical pathway DAA; 2) positive potential around the linkers between complement control protein domains (CCPs) 1-2 and 2-3 is more important for DAAs than for CFAs; 3) positive potential in CCP1 is crucial for binding to C3b and C4b, and thereby its functional activities; 4) conversion to negative or enhancement of negative potential for CCPs 2-4 has a marked effect on C3b-linked activities as opposed to C4b-linked activities; and 5) reversal of the electrostatic potential of CCP4 to negative has a differential effect on classical and alternative pathway DAAs. Together, our data provide functional relevance to conservation of positive potential in CCPs 1 and 4 and the linkers of viral complement regulators.


Assuntos
Proteínas Inativadoras do Complemento/fisiologia , Herpesvirus Humano 8/imunologia , Eletricidade Estática , Proteínas Virais/fisiologia , Proteínas Inativadoras do Complemento/genética , Via Alternativa do Complemento/genética , Via Alternativa do Complemento/imunologia , Herpesvirus Humano 8/genética , Humanos , Mutagênese Sítio-Dirigida , Proteínas Virais/genética
20.
J Gastrointest Oncol ; 13(2): 871-884, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35557598

RESUMO

Background and Objective: Gastric cancer carries a poor prognosis despite advances in treatment. Despite curative-intent surgery, the risk of recurrence is high. Perioperative treatment may improve rates of complete surgical resection and reduce the rate of recurrence. Treatment practices vary worldwide, while perioperative treatment is considered standard-of-care practice in Western countries, upfront surgery followed by adjuvant therapy is preferred in Asian countries. The current literature is complex to navigate with a plethora of studies available for review. The aim of this review is to summarise current evidence regarding the role of perioperative treatment in resectable gastric cancer and to explore future directions in research. Methods: We searched the PubMed database for peer-reviewed original articles from phase III trials, published between 2002 to 2021 with regard to the treatment of resectable gastric cancer. Current active clinical trials regarding the use of targeted therapy and immune checkpoint inhibitors in perioperative and adjuvant therapy were identified using the ClinicalTrials.gov database from the US National Library of Medicine. Key Content and Findings: Compared to surgery alone, the use of perioperative chemotherapy prior to resection of gastric cancer and the use of adjuvant chemotherapy after upfront surgery both improve survival in those with resectable gastric cancer. However, treatment practices vary worldwide. In clinical practice, patient factors such as functional status should be considered when considering treatment approach. Many current clinical trials explore the role of targeted therapy and immune checkpoint inhibitors in the perioperative setting, which appear to be promising. Conclusions: Gastric cancer continues to carry a poor prognosis. The addition of targeted agents and immune checkpoint inhibitors in the perioperative setting appear to be promising although further research is required in this area to assess efficacy. Further clinical research is required to identify new agents and approaches to treatment to improve the survival of these patients.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA