Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Immunol ; 10(7): 721-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19503103

RESUMEN

Activation of the complement system generates potent chemoattractants and leads to the opsonization of cells for immune clearance. Short-lived protease complexes cleave complement component C3 into anaphylatoxin C3a and opsonin C3b. Here we report the crystal structure of the C3 convertase formed by C3b and the protease fragment Bb, which was stabilized by the bacterial immune-evasion protein SCIN. The data suggest that the proteolytic specificity and activity depend on the formation of dimers of C3 with C3b of the convertase. SCIN blocked the formation of a productive enzyme-substrate complex. Irreversible dissociation of the complex of C3b and Bb is crucial to complement regulation and was determined by slow binding kinetics of the Mg(2+)-adhesion site in Bb. Understanding the mechanistic basis of the central complement-activation step and microbial immune evasion strategies targeting this step will aid in the development of complement therapeutics.


Asunto(s)
Proteínas Bacterianas/química , C3 Convertasa de la Vía Alternativa del Complemento/química , Proteínas Inactivadoras de Complemento/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Sitios de Unión , Catálisis , Dominio Catalítico , Complemento C3/química , Complemento C3/metabolismo , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Convertasas de Complemento C3-C5/química , Convertasas de Complemento C3-C5/metabolismo , Complemento C3b/química , Complemento C3b/metabolismo , Proteínas Inactivadoras de Complemento/inmunología , Proteínas Inactivadoras de Complemento/metabolismo , Vía Alternativa del Complemento/inmunología , Cristalografía por Rayos X , Humanos , Cinética , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Staphylococcus aureus/química , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo , Especificidad por Sustrato , Resonancia por Plasmón de Superficie
2.
EMBO J ; 35(10): 1133-49, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27013439

RESUMEN

Regulators of complement activation (RCA) inhibit complement-induced immune responses on healthy host tissues. We present crystal structures of human RCA (MCP, DAF, and CR1) and a smallpox virus homolog (SPICE) bound to complement component C3b. Our structural data reveal that up to four consecutive homologous CCP domains (i-iv), responsible for inhibition, bind in the same orientation and extended arrangement at a shared binding platform on C3b. Large sequence variations in CCP domains explain the diverse C3b-binding patterns, with limited or no contribution of some individual domains, while all regulators show extensive contacts with C3b for the domains at the third site. A variation of ~100° rotation around the longitudinal axis is observed for domains binding at the fourth site on C3b, without affecting the overall binding mode. The data suggest a common evolutionary origin for both inhibitory mechanisms, called decay acceleration and cofactor activity, with variable C3b binding through domains at sites ii, iii, and iv, and provide a framework for understanding RCA disease-related mutations and immune evasion.


Asunto(s)
Complemento C3b/química , Complemento C3b/metabolismo , Sitios de Unión , Antígenos CD55/química , Antígenos CD55/metabolismo , Activación de Complemento , Humanos , Proteína Cofactora de Membrana/química , Proteína Cofactora de Membrana/metabolismo , Dominios Proteicos , Receptores de Complemento 3b/química , Receptores de Complemento 3b/metabolismo , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/metabolismo
3.
Dev Biol ; 428(1): 88-100, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28576690

RESUMEN

Complement components have been implicated in a wide variety of functions including neurogenesis, proliferation, cell migration, differentiation, cancer, and more recently early development and regeneration. Following our initial observations indicating that C3a/C3aR signaling induces chick retina regeneration, we analyzed its role in chick eye morphogenesis. During eye development, the optic vesicle (OV) invaginates to generate a bilayer optic cup (OC) that gives rise to the retinal pigmented epithelium (RPE) and neural retina. We show by immunofluorescence staining that C3 and the receptor for C3a (the cleaved and active form of C3), C3aR, are present in chick embryos during eye morphogenesis in the OV and OC. Interestingly, C3aR is mainly localized in the nuclear compartment at the OC stage. Loss of function studies at the OV stage using morpholinos or a blocking antibody targeting the C3aR (anti-C3aR Ab), causes eye defects such as microphthalmia and defects in the ventral portion of the eye that result in coloboma. Such defects were not observed when C3aR was disrupted at the OC stage. Histological analysis demonstrated that microphthalmic eyes were unable to generate a normal optic stalk or a closed OC. The dorsal/ventral patterning defects were accompanied by an expansion of the ventral markers Pax2, cVax and retinoic acid synthesizing enzyme raldh-3 (aldh1a3) domains, an absence of the dorsal expression of Tbx5 and raldh-1 (aldh1a1) and a re-specification of the ventral RPE to neuroepithelium. In addition, the eyes showed overall decreased expression of Gli1 and a change in distribution of nuclear ß-catenin, suggesting that Shh and Wnt pathways have been affected. Finally, we observed prominent cell death along with a decrease in proliferating cells, indicating that both processes contribute to the microphthalmic phenotype. Together our results show that C3aR is necessary for the proper morphogenesis of the OC. This is the first report implicating C3aR in eye development, revealing an unsuspected hitherto regulator for proper chick eye morphogenesis.


Asunto(s)
Tipificación del Cuerpo/fisiología , Complemento C3a/metabolismo , Regulación del Desarrollo de la Expresión Génica , Receptores de Complemento/metabolismo , Epitelio Pigmentado de la Retina/embriología , Aldehído Deshidrogenasa/metabolismo , Animales , Apoptosis/fisiología , Proliferación Celular/fisiología , Embrión de Pollo , Proteínas Hedgehog/metabolismo , Microftalmía/embriología , Morfogénesis/fisiología , Factor de Transcripción PAX2/metabolismo , Receptores de Complemento/genética , Retinal-Deshidrogenasa/metabolismo , Proteínas de Dominio T Box/metabolismo , Vía de Señalización Wnt/fisiología , Proteína con Dedos de Zinc GLI1/biosíntesis , beta Catenina/metabolismo
4.
J Biol Chem ; 288(4): 2870-81, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23233676

RESUMEN

Complement is a network of interacting circulatory and cell surface proteins that recognizes, marks, and facilitates clearance of microbial invaders. To evade complement attack, the pathogenic organism Staphylococcus aureus expresses a number of secreted proteins that interfere with activation and regulation of the complement cascade. Staphylococcal complement inhibitors (SCINs) are one important class of these immunomodulators and consist of three active members (SCIN-A/-B/-C). SCINs inhibit a critical enzymatic complex, the alternative pathway C3 convertase, by targeting a functional "hot spot" on the central opsonin of complement, C3b. Although N-terminal truncation mutants of SCINs retain complement inhibitory properties, they are significantly weaker binders of C3b. To provide a structural basis for this observation, we undertook a series of crystallographic and NMR dynamics studies on full-length SCINs. This work reveals that N-terminal SCIN domains are characterized by a conformationally dynamic helical motif. C3b binding and functional experiments further demonstrate that this sequence-divergent N-terminal region of SCINs is both functionally important and context-dependent. Finally, surface plasmon resonance data provide evidence for the formation of inhibitor·enzyme·substrate complexes ((SCIN·C3bBb)·C3). Similar to the (SCIN·C3bBb)(2) pseudodimeric complexes, ((SCIN·C3bBb)·C3) interferes with the interaction of complement receptors and C3b. This activity provides an additional mechanism by which SCIN couples convertase inhibition to direct blocking of phagocytosis. Together, these data suggest that tethering multi-host protein complexes by small modular bacterial inhibitors may be a global strategy of immune evasion used by S. aureus. The work presented here provides detailed structure-activity relationships and improves our understanding of how S. aureus circumvents human innate immunity.


Asunto(s)
C3 Convertasa de la Vía Alternativa del Complemento/química , Complemento C3b/química , Proteínas Inactivadoras de Complemento/metabolismo , Staphylococcus aureus/metabolismo , Proteínas Inactivadoras de Complemento/química , Reactivos de Enlaces Cruzados/química , Cristalografía por Rayos X/métodos , Humanos , Sistema Inmunológico , Inmunidad Innata , Espectroscopía de Resonancia Magnética/métodos , Fagocitosis , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas/métodos , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie
5.
J Clin Immunol ; 34 Suppl 1: S132-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24722853

RESUMEN

Neuroinflammation plays an important role in the secondary pathophysiological mechanisms of spinal cord injury (SCI) and can exacerbate the primary trauma and thus worsen recovery. Although some aspects of the immune response are beneficial, it is thought that leukocyte recruitment and activation in the acute phase of injury results in the production of cytotoxic substances that are harmful to the nervous tissue. Therefore, suppression of excessive inflammation in the spinal cord could serve as a therapeutic strategy to attenuate tissue damage. The immunosuppressant methylprednisolone has been used in the setting of SCI, but there are complications which have attenuated the initial enthusiasm. Hence, there is interest in other immunomodulatory approaches, such as intravenous Immunoglobulin G (IVIg). Importantly, IVIg is used clinically for the treatment of several auto-immune neuropathies, such as Guillain-Barre syndrome, chronic inflammatory demyelinating polyneuropathy (CIPD) and Kawasaki disease, with a good safety profile. Thus, it is a promising treatment candidate for SCI. Indeed, IVIg has been shown by our team to attenuate the immune response and result in improved neurobehavioral recovery following cervical SCI in rats through a mechanism that involves the attenuation of neutrophil recruitment and reduction in the levels of cytokines and cytotoxic enzymes Nguyen et al. (J Neuroinflammation 9:224, 2012). Here we review published data in the context of relevant mechanisms of action that have been proposed for IVIg in other conditions. We hope that this discussion will trigger future research to provide supporting evidence for the efficiency and detailed mechanisms of action of this promising drug in the treatment of SCI, and to facilitate its clinical translation.


Asunto(s)
Inmunoglobulinas Intravenosas/uso terapéutico , Inmunosupresores/uso terapéutico , Inmunoterapia/tendencias , Inflamación Neurogénica/terapia , Neutrófilos/inmunología , Traumatismos de la Médula Espinal/terapia , Animales , Movimiento Celular/efectos de los fármacos , Citocinas/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Inmunoglobulinas Intravenosas/inmunología , Inmunosupresores/inmunología , Inflamación Neurogénica/inmunología , Ratas , Traumatismos de la Médula Espinal/inmunología
6.
J Immunol ; 186(2): 869-77, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149611

RESUMEN

The C5a anaphylatoxin receptor (C5aR; CD88) is activated as part of the complement cascade and exerts important inflammatory, antimicrobial, and regulatory functions, at least in part, via crosstalk with TLRs. However, the periodontal pathogen Porphyromonas gingivalis can control C5aR activation by generating C5a through its own C5 convertase-like enzymatic activity. In this paper, we show that P. gingivalis uses this mechanism to proactively and selectively inhibit TLR2-induced IL-12p70, whereas the same pathogen-instigated C5aR-TLR2 crosstalk upregulates other inflammatory and bone-resorptive cytokines (IL-1ß, IL-6, and TNF-α). In vivo, the ability of P. gingivalis to manipulate TLR2 activation via the C5a-C5aR axis allowed it to escape IL-12p70-dependent immune clearance and to cause inflammatory bone loss in a murine model of experimental periodontitis. In the latter regard, C5aR-deficient or TLR2-deficient mice were both resistant to periodontal bone loss, in stark contrast with wild-type control mice, which is consistent with the interdependent interactions of C5aR and TLR2 in P. gingivalis immune evasion and induction of bone-resorptive cytokines. In conclusion, P. gingivalis targets C5aR to promote its adaptive fitness and cause periodontal disease. Given the current availability of safe and effective C5aR antagonists, pharmacological blockade of C5aR could act therapeutically in human periodontitis and reduce associated systemic risks.


Asunto(s)
Pérdida de Hueso Alveolar/inmunología , Pérdida de Hueso Alveolar/microbiología , Interleucina-12/antagonistas & inhibidores , Interleucina-12/fisiología , Periodontitis/inmunología , Periodontitis/microbiología , Porphyromonas gingivalis/inmunología , Receptor de Anafilatoxina C5a/fisiología , Pérdida de Hueso Alveolar/metabolismo , Animales , Modelos Animales de Enfermedad , Evasión Inmune/genética , Evasión Inmune/inmunología , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Interleucina-10/fisiología , Interleucina-12/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/fisiología , Periodontitis/metabolismo , Porphyromonas gingivalis/crecimiento & desarrollo , Receptor Cross-Talk/inmunología , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 2/fisiología
7.
J Immunol ; 186(7): 4269-77, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21339361

RESUMEN

Exposure of nonself surfaces such as those of biomaterials or transplanted cells and organs to host blood frequently triggers innate immune responses, thereby affecting both their functionality and tolerability. Activation of the alternative pathway of complement plays a decisive role in this unfavorable reaction. Whereas previous studies demonstrated that immobilization of physiological regulators of complement activation (RCA) can attenuate this foreign body-induced activation, simple and efficient approaches for coating artificial surfaces with intact RCA are still missing. The conjugation of small molecular entities that capture RCA with high affinity is an intriguing alternative, as this creates a surface with autoregulatory activity upon exposure to blood. We therefore screened two variable cysteine-constrained phage-displayed peptide libraries for factor H-binding peptides. We discovered three peptide classes that differed with respect to their main target binding areas. Peptides binding to the broad middle region of factor H (domains 5-18) were of particular interest, as they do not interfere with either regulatory or binding activities. One peptide in this group (5C6) was further characterized and showed high factor H-capturing activity while retaining its functional integrity. Most importantly, when 5C6 was coated to a model polystyrene surface and exposed to human lepirudin-anticoagulated plasma, the bound peptide captured factor H and substantially inhibited complement activation by the alternative pathway. Our study therefore provides a promising and novel approach to produce therapeutic materials with enhanced biocompatibility.


Asunto(s)
Vía Alternativa del Complemento/inmunología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/uso terapéutico , Materiales Biocompatibles/metabolismo , Clonación Molecular , Complemento C3b/antagonistas & inhibidores , Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Factor H de Complemento/uso terapéutico , Factor I de Complemento/antagonistas & inhibidores , Factor I de Complemento/metabolismo , Proteínas Inactivadoras de Complemento/metabolismo , Hemólisis , Humanos , Biblioteca de Péptidos , Unión Proteica/inmunología , Propiedades de Superficie
8.
Proc Natl Acad Sci U S A ; 107(41): 17621-6, 2010 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-20876141

RESUMEN

The complement system is a major target of immune evasion by Staphylococcus aureus. Although many evasion proteins have been described, little is known about their molecular mechanisms of action. Here we demonstrate that the extracellular fibrinogen-binding protein (Efb) from S. aureus acts as an allosteric inhibitor by inducing conformational changes in complement fragment C3b that propagate across several domains and influence functional regions far distant from the Efb binding site. Most notably, the inhibitor impaired the interaction of C3b with complement factor B and, consequently, formation of the active C3 convertase. As this enzyme complex is critical for both activation and amplification of the complement response, its allosteric inhibition likely represents a fundamental contribution to the overall immune evasion strategy of S. aureus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Complemento C3b/metabolismo , Proteínas Inactivadoras de Complemento/metabolismo , Evasión Inmune/fisiología , Modelos Moleculares , Conformación Proteica , Staphylococcus aureus/fisiología , Regulación Alostérica/fisiología , Proteínas Bacterianas/química , Convertasas de Complemento C3-C5/metabolismo , Complemento C3b/química , Factor B del Complemento/metabolismo , Proteínas Inactivadoras de Complemento/química , Espectrometría de Masas , Dispersión del Ángulo Pequeño , Staphylococcus aureus/metabolismo , Resonancia por Plasmón de Superficie
9.
J Immunol ; 183(4): 2565-74, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19625656

RESUMEN

Staphylococcus aureus possesses an impressive arsenal of complement evasion proteins that help the bacterium escape attack of the immune system. The staphylococcal complement inhibitor (SCIN) protein exhibits a particularly high potency and was previously shown to block complement by acting at the level of the C3 convertases. However, many details about the exact binding and inhibitory mechanism remained unclear. In this study, we demonstrate that SCIN directly binds with nanomolar affinity to a functionally important area of C3b that lies near the C terminus of its beta-chain. Direct competition of SCIN with factor B for C3b slightly decreased the formation of surface-bound convertase. However, the main inhibitory effect can be attributed to an entrapment of the assembled convertase in an inactive state. Whereas native C3 is still able to bind to the blocked convertase, no generation and deposition of C3b could be detected in the presence of SCIN. Furthermore, SCIN strongly competes with the binding of factor H to C3b and influences its regulatory activities: the SCIN-stabilized convertase was essentially insensitive to decay acceleration by factor H and the factor I- and H-mediated conversion of surface-bound C3b to iC3b was significantly reduced. By targeting a key area on C3b, SCIN is able to block several essential functions within the alternative pathway, which explains the high potency of the inhibitor. Our findings provide an important insight into complement evasion strategies by S. aureus and may act as a base for further functional studies.


Asunto(s)
Complemento C3b/metabolismo , Proteínas Inactivadoras de Complemento/fisiología , Familia de Multigenes/inmunología , Staphylococcus aureus/inmunología , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , C3 Convertasa de la Vía Alternativa del Complemento/fisiología , Complemento C3b/química , Factor B del Complemento/metabolismo , Factor H de Complemento/metabolismo , Proteínas Inactivadoras de Complemento/metabolismo , Humanos , Unión Proteica/inmunología , Staphylococcus aureus/patogenicidad , Virulencia
10.
PLoS One ; 15(10): e0241285, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33125407

RESUMEN

Adaptive immunity is critical for controlling infections, which are a leading cause of morbidity and mortality in patients with spinal cord injury (SCI). In rats and mice, compromised peripheral adaptive immune responses, as shown by splenic atrophy and lowered frequencies of peripheral lymphocytes, were shown to result from high-level thoracic SCI. However, whether cervical SCI, which is the most common level of SCI in humans, impairs adaptive immunity remains largely unknown. In the present study, we induced cervical SCI in rats at the C7/T1 level by clip compression and looked at changes in peripheral adaptive immunity at 2-, 10- and 20-weeks post-injury. Specifically, we quantified changes in the frequencies of T- and B- lymphocytes in the blood and the mandibular and deep cervical lymph nodes, which drain the cervical spinal cord. We also assessed changes in serum IgG and IgM immunoglobulin levels, as well as spleen size. We found a significant decline in circulating T- and B- cell frequencies at 10 weeks post-SCI, which returned to normal at 20 weeks after injury. We found no effect of cervical SCI on T- and B- cell frequencies in the draining lymph nodes. Moreover, cervical SCI had no effect on net spleen size, although injured rats had a higher spleen/body weight ratio than sham controls at all time points of the study. Lastly, IgG and IgM immunoglobulin declined at 2 weeks, followed by a significant increase in IgM levels at 10 weeks of injury. These data indicate that cervical SCI causes a significant imbalance in circulating lymphocytes and immunoglobulin levels at 2 and 10 weeks. As we discuss in this article, these findings are largely in line with clinical observations, and we anticipate that this study will fuel more research on the effect of adaptive immunity on SCI recovery.


Asunto(s)
Inmunidad Adaptativa/fisiología , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/patología , Animales , Linfocitos B/metabolismo , Médula Cervical/inmunología , Médula Cervical/patología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Ganglios Linfáticos/inmunología , Ratones , Ratas , Ratas Wistar , Linfocitos T/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-19407382

RESUMEN

Staphylococcus aureus secretes a number of small proteins that effectively attenuate the human innate immune response. Among these, the staphylococcal complement-inhibitor protein (SCIN) disrupts the function of the complement component 3 (C3) convertase that is initiated through either the classical or the alternative pathway and thereby prevents amplification of the complement response on the bacterial surface. Recent studies have shown that SCIN may affect the activities of the C3 convertase by binding in an equimolar fashion to C3b, which is itself an integral although non-enzymatic component of the convertase. In order to better understand the nature of the C3b-SCIN interaction, the hanging-drop vapor-diffusion technique was used to crystallize human C3b in the presence of a recombinant form of SCIN. These crystals diffracted synchrotron X-rays to approximately 6 A Bragg spacing and grew in a primitive tetragonal space group (P4(1)2(1)2 or P4(3)2(1)2; unit-cell parameters a = b = 128.03, c = 468.59 A). Cell-content analysis of these crystals was consistent with the presence of either two 1:1 complexes or a single 2:2 assembly in the asymmetric unit, both of which correspond to a solvent content of 51.9%. By making use of these crystals, solution of the C3b-SCIN structure should further our understanding of complement inhibition and immune evasion by this pathogen.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/química , Proteínas Inactivadoras del Complemento C3b/metabolismo , Complemento C3b/química , Complemento C3b/metabolismo , Staphylococcus aureus/metabolismo , Complemento C3b/genética , Proteínas Inactivadoras del Complemento C3b/genética , Cristalización , Cristalografía por Rayos X , Humanos , Staphylococcus aureus/genética
12.
Alzheimers Dement (N Y) ; 4: 37-45, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29955650

RESUMEN

INTRODUCTION: Translational inhibition of amyloid precursor protein (APP) by Posiphen has been shown to reduce APP and its fragments in cell culture, animal models, and mildly cognitively impaired patients, making it a promising drug candidate for the treatment of Alzheimer's disease. METHODS: We used a mouse model of Alzheimer's disease (APP/presenilin-1) to examine Posiphen's efficacy, pharmacodynamics, and pharmacokinetics. RESULTS: Posiphen treatment normalized impairments in spatial working memory, contextual fear learning, and synaptic function in APP/presenilin-1 mice, without affecting their visual acuity, motor skills, or motivation and without affecting wild-type mice. Posiphen had a prolonged effect in reducing APP and all related peptides for at least 9 hours after the last dose. Its concentration was higher in the brain than in plasma, and the most abundant metabolite was N8-norPosiphen. DISCUSSION: This is the first study demonstrating the therapeutic efficacy of inhibiting the translation of APP and its fragments in an Alzheimer's disease model.

13.
J Neurotrauma ; 34(6): 1209-1226, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-27775474

RESUMEN

The immune system plays a critical and complex role in the pathobiology of spinal cord injury (SCI), exerting both beneficial and detrimental effects. Increasing evidence suggests that there are injury level-dependent differences in the immune response to SCI. Patients with traumatic SCI have elevated levels of circulating autoantibodies against components of the central nervous system, but the role of these antibodies in SCI outcomes remains unknown. In rodent models of mid-thoracic SCI, antibody-mediated autoimmunity appears to be detrimental to recovery. However, whether autoantibodies against the spinal cord are generated following cervical SCI (cSCI), the most common level of injury in humans, remains undetermined. To address this knowledge gap, we investigated the antibody responses following cSCI in a rat model of injury. We found increased immunoglobulin G (IgG) and IgM antibodies in the spinal cord in the subacute phase of injury (2 weeks), but not in more chronic phases (10 and 20 weeks). At 2 weeks post-cSCI, antibodies were detected at the injury epicenter and co-localized with the astroglial scar and neurons of the ventral horn. These increased levels of antibodies corresponded with enhanced activation of immune responses in the spleen. Higher counts of antibody-secreting cells were observed in the spleen of injured rats. Further, increased levels of secreted IgG antibodies and enhanced proliferation of T-cells in splenocyte cultures from injured rats were found. These findings suggest the potential development of autoantibody responses following cSCI in the rat. The impact of the post-traumatic antibody responses on functional outcomes of cSCI is a critical topic that requires further investigation.


Asunto(s)
Autoanticuerpos/inmunología , Médula Cervical/lesiones , Traumatismos de la Médula Espinal/inmunología , Animales , Células Productoras de Anticuerpos/inmunología , Astrocitos/inmunología , Modelos Animales de Enfermedad , Femenino , Ratas , Ratas Wistar , Bazo/inmunología
14.
Nat Commun ; 4: 2312, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23942241

RESUMEN

Identifying the initiation signals for tissue regeneration in vertebrates is one of the major challenges in regenerative biology. Much of the research thus far has indicated that certain growth factors have key roles. Here we show that complement fragment C3a is sufficient to induce complete regeneration of the embryonic chick retina from stem/progenitor cells present in the eye, independent of fibroblast growth factor receptor signaling. Instead, C3a induces retina regeneration via STAT3 activation, which in turn activates the injury- and inflammation-responsive factors, IL-6, IL-8 and TNF-α. This activation sets forth regulation of Wnt2b, Six3 and Sox2, genes associated with retina stem and progenitor cells. Thus, our results establish a mechanism for retina regeneration based on injury and inflammation signals. Furthermore, our results indicate a unique function for complement anaphylatoxins that implicate these molecules in the induction and complete regeneration of the retina, opening new avenues of experimentation in the field.


Asunto(s)
Complemento C3a/metabolismo , Regeneración/fisiología , Retina/metabolismo , Factor de Transcripción STAT3/metabolismo , Ingeniería de Tejidos/métodos , Animales , Embrión de Pollo , Activación Enzimática , Proteínas del Ojo/metabolismo , Regeneración Tisular Dirigida , Proteínas de Homeodominio/metabolismo , Interleucina-6/biosíntesis , Interleucina-6/metabolismo , Interleucina-8/biosíntesis , Interleucina-8/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Cultivo de Órganos , Regeneración/inmunología , Retina/embriología , Retina/crecimiento & desarrollo , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción STAT3/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo , Proteína Wnt3/metabolismo , Proteína Homeobox SIX3
15.
Immunobiology ; 218(4): 496-505, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22795972

RESUMEN

Therapeutic modulation of the complement system has become increasingly important in line with the growing recognition of the role of complement in numerous diseases. Compstatin, a peptidic inhibitor that acts at the central level of the complement cascade, is currently in clinical evaluation but routes to improve its efficacy have not yet been fully explored. Here, we report improvements in both the inhibitory potency and pharmacokinetic parameters of compstatin that broaden its clinical applications. Selective modification of the compstatin N-terminus with non-proteinogenic amino acids resulted in the first analogue with subnanomolar binding affinity (KD=0.5nM) and other similarly potent derivatives with improved solubility in clinically relevant solvents. Detailed structure-activity relationship studies based on biophysical and computational methods revealed key structural determinants for the observed improvements. Importantly, pharmacokinetic evaluation in non-human primates revealed target-driven elimination kinetics with plasma half-life values exceeding expectations for peptidic drugs (close to 12h). This successful optimization strategy is expected to pave the way for systemic administration of compstatin in a range of clinical conditions.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Péptidos Cíclicos , Animales , Semivida , Haplorrinos , Humanos , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacocinética , Péptidos Cíclicos/farmacología , Relación Estructura-Actividad
16.
ACS Chem Biol ; 6(7): 753-60, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21520911

RESUMEN

Disulfide bonds are essential for the structural stability and biological activity of many bioactive peptides. However, these bonds are labile to reducing agents, which can limit the therapeutic utility of such peptides. Substitution of a disulfide bond with a reduction-resistant cystathionine bridge is an attractive means of improving stability while imposing minimal structural perturbation to the peptide. We have applied this approach to the therapeutic complement inhibitor compstatin, a disulfide-containing peptide currently in clinical trials for age-related macular degeneration, in an effort to maintain its potent activity while improving its biological stability. Thioether-containing compstatin analogues were produced via solid-phase peptide synthesis utilizing orthogonally protected cystathionine amino acid building blocks and solid-supported peptide cyclization. Overall, the affinity of these analogues for their biological target and potent inhibition of complement activation were largely maintained when compared to those of the parent disulfide-containing peptides. Thus, the improved stability to reduction conferred by the thioether bond makes this new class of compstatin peptides a promising alternative for therapeutic applications. Additionally, the versatility of this synthesis allows for exploration of disulfide-to-thioether substitution in a variety of other therapeutic peptides.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Sulfuros/química , Complemento C3/antagonistas & inhibidores , Complemento C3/metabolismo , Disulfuros/química , Oxidación-Reducción , Péptidos Cíclicos/síntesis química
17.
Dev Cell ; 21(6): 1026-37, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22118769

RESUMEN

Collective cell migration is a mode of movement crucial for morphogenesis and cancer metastasis. However, little is known about how migratory cells coordinate collectively. Here we show that mutual cell-cell attraction (named here coattraction) is required to maintain cohesive clusters of migrating mesenchymal cells. Coattraction can counterbalance the natural tendency of cells to disperse via mechanisms such as contact inhibition and epithelial-to-mesenchymal transition. Neural crest cells are coattracted via the complement fragment C3a and its receptor C3aR, revealing an unexpected role of complement proteins in early vertebrate development. Loss of coattraction disrupts collective and coordinated movements of these cells. We propose that coattraction and contact inhibition act in concert to allow cell collectives to self-organize and respond efficiently to external signals, such as chemoattractants and repellents.


Asunto(s)
Comunicación Celular/fisiología , Movimiento Celular/fisiología , Complemento C3a/fisiología , Animales , Adhesión Celular/fisiología , Factores Quimiotácticos/genética , Factores Quimiotácticos/fisiología , Complemento C3a/genética , Transición Epitelial-Mesenquimal/fisiología , Modelos Neurológicos , Datos de Secuencia Molecular , Células Madre Multipotentes/fisiología , Cresta Neural/citología , Cresta Neural/embriología , Células-Madre Neurales/fisiología , Receptores de Complemento/genética , Receptores de Complemento/fisiología , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiología , Xenopus laevis/embriología , Xenopus laevis/genética , Xenopus laevis/fisiología , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
18.
Science ; 330(6012): 1816-20, 2010 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-21205667

RESUMEN

Activation of the complement cascade induces inflammatory responses and marks cells for immune clearance. In the central complement-amplification step, a complex consisting of surface-bound C3b and factor B is cleaved by factor D to generate active convertases on targeted surfaces. We present crystal structures of the pro-convertase C3bB at 4 angstrom resolution and its complex with factor D at 3.5 angstrom resolution. Our data show how factor B binding to C3b forms an open "activation" state of C3bB. Factor D specifically binds the open conformation of factor B through a site distant from the catalytic center and is activated by the substrate, which displaces factor D's self-inhibitory loop. This concerted proteolytic mechanism, which is cofactor-dependent and substrate-induced, restricts complement amplification to C3b-tagged target cells.


Asunto(s)
C3 Convertasa de la Vía Alternativa del Complemento/química , Complemento C3b/química , Factor B del Complemento/química , Factor D del Complemento/química , Sitios de Unión , Dominio Catalítico , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Complemento C3b/metabolismo , Factor B del Complemento/metabolismo , Factor D del Complemento/metabolismo , Vía Alternativa del Complemento , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína
19.
J Mol Biol ; 402(1): 17-29, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20654625

RESUMEN

The human complement system plays an essential role in innate and adaptive immunity by marking and eliminating microbial intruders. Activation of complement on foreign surfaces results in proteolytic cleavage of complement component 3 (C3) into the potent opsonin C3b, which triggers a variety of immune responses and participates in a self-amplification loop mediated by a multi-protein assembly known as the C3 convertase. The human pathogen Staphylococcus aureus has evolved a sophisticated and potent complement evasion strategy, which is predicated upon an arsenal of potent inhibitory proteins. One of these, the staphylococcal complement inhibitor (SCIN), acts at the level of the C3 convertase (C3bBb) and impairs downstream complement function by trapping the convertase in a stable but inactive state. Previously, we have shown that SCIN binds C3b directly and competitively inhibits binding of human factor H and, to a lesser degree, that of factor B to C3b. Here, we report the co-crystal structures of SCIN bound to C3b and C3c at 7.5 and 3.5 A limiting resolution, respectively, and show that SCIN binds a critical functional area on C3b. Most significantly, the SCIN binding site sterically occludes the binding sites of both factor H and factor B. Our results give insight into SCIN binding to activated derivatives of C3, explain how SCIN can recognize C3b in the absence of other complement components, and provide a structural basis for the competitive C3b-binding properties of SCIN. In the future, this may suggest templates for the design of novel complement inhibitors based upon the SCIN structure.


Asunto(s)
Complemento C3b/química , Complemento C3c/química , Proteínas Inactivadoras de Complemento/química , Staphylococcus aureus/inmunología , Sitios de Unión , Complemento C3b/metabolismo , Complemento C3c/metabolismo , Proteínas Inactivadoras de Complemento/metabolismo , Cristalografía por Rayos X , Humanos , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA