Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 38(6): 110322, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35139380

RESUMEN

RAS guanosine triphosphatases (GTPases) are mutated in nearly 20% of human tumors, making them an attractive therapeutic target. Following our discovery that nucleotide-free RAS (apo RAS) regulates cell signaling, we selectively target this state as an approach to inhibit RAS function. Here, we describe the R15 monobody that exclusively binds the apo state of all three RAS isoforms in vitro, regardless of the mutation status, and captures RAS in the apo state in cells. R15 inhibits the signaling and transforming activity of a subset of RAS mutants with elevated intrinsic nucleotide exchange rates (i.e., fast exchange mutants). Intracellular expression of R15 reduces the tumor-forming capacity of cancer cell lines driven by select RAS mutants and KRAS(G12D)-mutant patient-derived xenografts (PDXs). Thus, our approach establishes an opportunity to selectively inhibit a subset of RAS mutants by targeting the apo state with drug-like molecules.


Asunto(s)
Genes ras/genética , Mutación/genética , Nucleótidos/metabolismo , Neoplasias Pancreáticas/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , Ratones , Neoplasias Pancreáticas/patología , Transducción de Señal/genética
2.
FASEB J ; 35(8): e21762, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34246197

RESUMEN

Phase II clinical trials have reported that acute treatment of surgical skin wounds with the therapeutic peptide alpha Connexin Carboxy-Terminus 1 (αCT1) improves cutaneous scar appearance by 47% 9-month postsurgery. While Cx43 and ZO-1 have been identified as molecular targets of αCT1, the mode-of-action of the peptide in scar mitigation at cellular and tissue levels remains to be further characterized. Scar histoarchitecture in αCT1 and vehicle-control treated skin wounds within the same patient were compared using biopsies from a Phase I clinical trial at 29-day postwounding. The sole effect on scar structure of a range of epidermal and dermal variables examined was that αCT1-treated scars had less alignment of collagen fibers relative to control wounds-a characteristic that resembles unwounded skin. The with-in subject effect of αCT1 on scar collagen order observed in Phase I testing in humans was recapitulated in Sprague-Dawley rats and the IAF hairless guinea pig. Transient increase in histologic collagen density in response to αCT1 was also observed in both animal models. Mouse NIH 3T3 fibroblasts and primary human dermal fibroblasts treated with αCT1 in vitro showed more rapid closure in scratch wound assays, with individual cells showing decreased directionality in movement. An agent-based computational model parameterized with fibroblast motility data predicted collagen alignments in simulated scars consistent with that observed experimentally in human and the animal models. In conclusion, αCT1 prompts decreased directionality of fibroblast movement and the generation of a 3D collagen matrix postwounding that is similar to unwounded skin-changes that correlate with long-term improvement in scar appearance.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Cicatriz/metabolismo , Conexina 43/metabolismo , Dermis/metabolismo , Fibroblastos/metabolismo , Péptidos/farmacología , Animales , Cicatriz/patología , Matriz Extracelular/metabolismo , Femenino , Cobayas , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
3.
Adv Cancer Res ; 148: 69-146, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32723567

RESUMEN

RAS was identified as a human oncogene in the early 1980s and subsequently found to be mutated in nearly 30% of all human cancers. More importantly, RAS plays a central role in driving tumor development and maintenance. Despite decades of effort, there remain no FDA approved drugs that directly inhibit RAS. The prevalence of RAS mutations in cancer and the lack of effective anti-RAS therapies stem from RAS' core role in growth factor signaling, unique structural features, and biochemistry. However, recent advances have brought promising new drugs to clinical trials and shone a ray of hope in the field. Here, we will exposit the details of RAS biology that illustrate its key role in cell signaling and shed light on the difficulties in therapeutically targeting RAS. Furthermore, past and current efforts to develop RAS inhibitors will be discussed in depth.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteínas ras/metabolismo , Animales , Antineoplásicos/farmacología , Humanos , Terapia Molecular Dirigida , Mutación , Neoplasias/patología , Oncogenes , Transducción de Señal/fisiología , Proteínas ras/antagonistas & inhibidores , Proteínas ras/genética
4.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118570, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31678118

RESUMEN

RAS is the most frequently mutated oncogene in cancer and a critical driver of oncogenesis. Therapeutic targeting of RAS has been a goal of cancer research for more than 30 years due to its essential role in tumor formation and maintenance. Yet the quest to inhibit this challenging foe has been elusive. Although once considered "undruggable", the struggle to directly inhibit RAS has seen recent success with the development of pharmacological agents that specifically target the KRAS(G12C) mutant protein, which include the first direct RAS inhibitor to gain entry to clinical trials. However, the limited applicability of these inhibitors to G12C-mutant tumors demands further efforts to identify more broadly efficacious RAS inhibitors. Understanding allosteric influences on RAS may open new avenues to inhibit RAS. Here, we provide a brief overview of RAS biology and biochemistry, discuss the allosteric regulation of RAS, and summarize the various approaches to develop RAS inhibitors.


Asunto(s)
Proteínas ras/metabolismo , Regulación Alostérica , Empalme Alternativo , Anticuerpos Monoclonales/inmunología , Humanos , Mutación , Neoplasias/metabolismo , Neoplasias/patología , Procesamiento Proteico-Postraduccional , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Proteínas ras/antagonistas & inhibidores , Proteínas ras/inmunología
5.
Int J Mol Sci ; 20(22)2019 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-31752345

RESUMEN

BACKGROUND: Autophagy is a catabolic cellular recycling pathway that is essential for maintaining intracellular homeostasis. Autophagosome formation is achieved via the coordination of the Beclin-1 protein complex. Rubicon is a Beclin-1 associated protein that suppresses autophagy by impairing the activity of the class III PI3K, Vps34. However, very little is known about the molecular mechanisms that regulate Rubicon function. METHODS: In this study, co-immunoprecipitation and kinase assays were used to investigate the ability of Hormonally Upregulated Neu-associated Kinase (HUNK) to bind to and phosphorylate Rubicon. LC3B was monitored by immunofluorescence and immunoblotting to determine whether phosphorylation of Rubicon by HUNK controls the autophagy suppressive function of Rubicon. RESULTS: Findings from this study identify Rubicon as a novel substrate of HUNK and show that phosphorylation of Rubicon inhibits its function, promoting autophagy.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia/fisiología , Fosforilación/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Fagosomas/metabolismo
6.
J Vis Exp ; (133)2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29658916

RESUMEN

Kidney transplantation is now a mainstream therapy for end-stage renal disease. However, with approximately 96,000 people on the waiting list and only one-fourth of these patients achieving transplantation, there is a dire need for alternatives for those with failing organs. In order to decrease the harmful consequences of dialysis along with the overall healthcare costs it incurs, active investigation is ongoing in search of alternative solutions to organ transplantation. Implantable tissue-engineered renal cellular constructs are one such feasible approach to replacing lost renal functionality. Here, described for the first time, is the microdissection of murine kidneys for isolation of living corticomedullary renal segments. These segments are capable of rapid incorporation within scaffold-free endothelial-fibroblast constructs which may enable rapid connection with host vasculature once implanted. Adult mouse kidneys were procured from living donors, followed by stereoscope microdissection to obtain renal segments 200 - 300 µm in diameter. Multiple renal constructs were fabricated using primary renal segments harvested from only one kidney. This method demonstrates a procedure which could salvage functional renal tissue from organs that would otherwise be discarded.


Asunto(s)
Trasplante de Riñón/métodos , Riñón/patología , Microdisección/métodos , Ingeniería de Tejidos/métodos , Animales , Femenino , Humanos , Masculino , Ratones
7.
Int J Mol Sci ; 19(4)2018 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-29601539

RESUMEN

Over the past few decades, connexin hemichannels have become recognized as major players in modulating the inflammatory response. Chronic inflammation is documented to promote tumorigenesis and is a critical component of tumor progression. Furthermore, inflammation is strongly linked to angiogenesis, immunotolerance, invasiveness, metastasis, and resistance in breast cancers. In this review, the literature on the role of connexin hemichannels in inflammation is summarized, and the potential role for hemichannel-mediated inflammation in driving breast cancer progression is discussed. Lastly, the potential for connexin-based therapeutics to modulate the inflammatory component of the tumor microenvironment as an avenue for the treatment of breast cancer is also discussed.


Asunto(s)
Neoplasias de la Mama/metabolismo , Conexinas/metabolismo , Neoplasias de la Mama/inmunología , Conexinas/inmunología , Progresión de la Enfermedad , Femenino , Humanos , Inflamación/inmunología , Inflamación/metabolismo
8.
Am J Physiol Cell Physiol ; 313(3): C314-C326, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28701358

RESUMEN

Connexin-based therapeutics have shown the potential for therapeutic efficacy in improving wound healing. Our previous work demonstrated that the connexin43 (Cx43) mimetic peptide juxtamembrane 2 (JM2) reduced the acute inflammatory response to a submuscular implant model by inhibiting purinergic signaling. Given the prospective application in improving tissue-engineered construct tolerance that these results indicated, we sought to determine the mechanism of action for JM2 in the present study. Using confocal microscopy, a gap-FRAP cell communication assay, and an ethidium bromide uptake assay of hemichannel function we found that the peptide reduced cell surface Cx43 levels, Cx43 gap junction (GJ) size, GJ communication, and hemichannel activity. JM2 is based on the sequence of the Cx43 microtubule binding domain, and microtubules have a confirmed role in intracellular trafficking of Cx43 vesicles. Therefore, we tested the effect of JM2 on Cx43-microtubule interaction and microtubule polymerization. We found that JM2 enhanced Cx43-microtubule interaction and that microtubule polymerization was significantly enhanced. Taken together, these data suggest that JM2 inhibits trafficking of Cx43 to the cell surface by promoting irrelevant microtubule polymerization and thereby reduces the number of hemichannels in the plasma membrane available to participate in proinflammatory purinergic signaling. Importantly, this work indicates that JM2 may have therapeutic value in the treatment of proliferative diseases such as cancer. We conclude that the targeted action of JM2 on Cx43 channels may improve the tolerance of implanted tissue-engineered constructs against the innate inflammatory response.


Asunto(s)
Antiinflamatorios/inmunología , Antiinflamatorios/farmacología , Conexina 43/inmunología , Microtúbulos/efectos de los fármacos , Microtúbulos/inmunología , Péptidos/farmacología , Conexina 43/antagonistas & inhibidores , Células HeLa , Humanos , Péptidos/síntesis química , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/inmunología
9.
Cancer Lett ; 374(1): 117-126, 2016 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-26884256

RESUMEN

Connexins are a family of transmembrane proteins that are characterized by their capacity to form intercellular channels called gap junctions that directly link the cytoplasm of adjacent cells. The formation of gap junctions by connexin proteins facilitates intercellular communication between neighboring cells by allowing for the transfer of ions and small signaling molecules. Communication through gap junctions is key to cellular equilibrium, where connexins, and the gap junction intercellular communication that connexins propagate, have roles in cellular processes such as cell growth, differentiation, and tissue homeostasis. Due to their importance in maintaining cellular functions, the disruption of connexin expression and function underlies the etiology and progression of numerous pathologies, including cancer. Over the past half a century, the role of connexins and gap junction intercellular communication have been highlighted as critical areas of research in cellular malignancies, and much research effort has been geared toward understanding their dysfunction in human cancers. Although ample evidence supports the role of connexins in a variety of human cancers, detailed examination in specific cancers, such as breast cancer, is still lacking. This review highlights the most abundant gap junction connexin isoform in higher vertebrate organisms, Connexin 43, and its role in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Conexina 43/metabolismo , Animales , Femenino , Humanos , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología
10.
J Diabetes Res ; 2016: 7262680, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26788521

RESUMEN

Total pancreatectomy and islet autotransplantation is a cutting-edge technique to treat chronic pancreatitis and postoperative diabetes. A major obstacle has been low islet cell survival due largely to the innate inflammatory response. Connexin43 (Cx43) channels play a key role in early inflammation and have proven to be viable therapeutic targets. Even if cell death due to early inflammation is avoided, insufficient vascularization is a primary obstacle to maintaining the viability of implanted cells. We have invented technologies targeting the inflammatory response and poor vascularization: a Cx43 mimetic peptide that inhibits inflammation and a novel prevascularized tissue engineered construct. We combined these technologies with isolated islets to create a prevascularized bioartificial pancreas that is resistant to the innate inflammatory response. Immunoconfocal microscopy showed that constructs containing islets express insulin and possess a vascular network similar to constructs without islets. Glucose stimulated islet-containing constructs displayed reduced insulin secretion compared to islets alone. However, labeling for insulin post-glucose stimulation revealed that the constructs expressed abundant levels of insulin. This discrepancy was found to be due to the expression of insulin degrading enzyme. These results suggest that the prevascularized bioartificial pancreas is potentially a tool for improving long-term islet cell survival in vivo.


Asunto(s)
Antiinflamatorios/farmacología , Órganos Bioartificiales , Conexina 43/farmacología , Diabetes Mellitus Tipo 1/cirugía , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/efectos de los fármacos , Páncreas Artificial , Pancreatitis Crónica/cirugía , Fragmentos de Péptidos/farmacología , Ingeniería de Tejidos/métodos , Animales , Células Cultivadas , Técnicas de Cocultivo , Diabetes Mellitus Tipo 1/inmunología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/trasplante , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/trasplante , Humanos , Inmunidad Innata/efectos de los fármacos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Ratones Endogámicos C57BL , Imitación Molecular , Pancreatitis Crónica/inmunología , Factores de Tiempo
11.
FEBS Lett ; 588(8): 1349-64, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24607540

RESUMEN

Gap junctions and their connexin components are indispensable in mediating the cellular coordination required for tissue and organ homeostasis. The critical nature of their existence mandates a connection to disease while at the same time offering therapeutic potential. Therapeutic intervention may be offered through the pharmacological and molecular disruption of the pathways involved in connexin biosynthesis, gap junction assembly, stabilization, or degradation. Chemical inhibitors aimed at closing connexin channels, peptide mimetics corresponding to short connexin sequences, and gene therapy approaches have been incredibly useful molecular tools in deciphering the complexities associated with connexin biology. Recently, therapeutic potential in targeting connexins has evolved from basic research in cell-based models to clinical opportunity in the form of human trials. Clinical promise is particularly evident with regards to targeting connexin43 in the context of wound healing. The following review is aimed at highlighting novel advances where the pharmacological manipulation of connexin biology has proven beneficial in animals or humans.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Conexina 43/metabolismo , Terapia Molecular Dirigida , Neoplasias/metabolismo , Péptidos/farmacología , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Enfermedades del Sistema Nervioso Central/metabolismo , Conexina 43/antagonistas & inhibidores , Conexina 43/genética , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Uniones Comunicantes/patología , Humanos , Neoplasias/tratamiento farmacológico , Péptidos/uso terapéutico , Enfermedades de la Piel/tratamiento farmacológico , Enfermedades de la Piel/metabolismo , Cicatrización de Heridas
12.
Tissue Eng Part B Rev ; 20(5): 392-402, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24279914

RESUMEN

Purinergic signaling is a ubiquitous and vital aspect of mammalian biology in which purines--mainly adenosine triphosphate (ATP)--are released from cells through loss of membrane integrity (cell death), exocytosis, or transport/diffusion across membrane channels, and exert paracrine or autocrine signaling effects through three subclasses of well-characterized receptors: the P1 adenosine receptors, the P2X ionotropic nucleotide receptors, and the P2Y metabotropic receptors. ATP and its metabolites are released by damaged and stressed cells in injured tissues. The early events of wound healing, hemostasis, and inflammation are highly regulated by these signals through activation of purinergic receptors on platelets and neutrophils. Recent data have demonstrated that ATP signaling is of particular importance to targeting leukocytes to sites of injury. This is particularly relevant to the subject of implanted medical devices, engineered tissues, and grafts as all these technologies elicit a wound healing response with varying degrees of encapsulation, rejection, extrusion, or destruction of the tissue or device. Here, we review the biology of purinergic signaling and focus on ATP release and response mechanisms that pertain to the early inflammatory phase of wound healing. Finally, therapeutic options are explored, including a new class of peptidomimetic drugs based on the ATP-conductive channel connexin43.


Asunto(s)
Adenosina Trifosfato/metabolismo , Reacción a Cuerpo Extraño/metabolismo , Inflamación/patología , Prótesis e Implantes , Receptores Purinérgicos/metabolismo , Transducción de Señal , Ingeniería de Tejidos/métodos , Animales , Espacio Extracelular/metabolismo , Humanos
13.
Trends Cardiovasc Med ; 23(6): 222-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23490883

RESUMEN

The perinexus is a recently identified microdomain surrounding the cardiac gap junction that contains elevated levels of connexin43 and the sodium channel protein, Nav1.5. Ongoing work has established a role for the perinexus in regulating gap junction aggregation. However, recent studies have raised the possibility of a perinexal contribution at the gap junction cleft to intercellular propagation of action potential via non-electrotonic mechanisms. The latter possibility could modify the current theoretical understanding of cardiac conduction, help explain paradoxical experimental findings, and open up entirely new avenues for antiarrhythmic therapy. We review recent structural insights into the perinexus and its potential novel functional role in cardiac-excitation spread, highlighting presently unanswered questions, the evidence for ephaptic conduction in the heart and how structural insights may help complete this picture.


Asunto(s)
Arritmias Cardíacas/metabolismo , Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Microdominios de Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Transducción de Señal , Potenciales de Acción , Animales , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/fisiopatología , Uniones Comunicantes/efectos de los fármacos , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiopatología , Humanos , Microdominios de Membrana/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
14.
J Membr Biol ; 245(7): 411-22, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22811280

RESUMEN

Gap junctions (GJs) are aggregates of channels that provide for direct cytoplasmic connection between cells. Importantly, this connection is thought responsible for cell-to-cell transfer of the cardiac action potential. The GJ channels of ventricular myocytes are composed of connexin43 (Cx43). Interaction of Cx43 with zonula occludens-1 (ZO-1) is localized not only at the GJ plaque, but also to the region surrounding the GJ, the perinexus. Cx43 in the perinexus is not detectable by immunofluorescence, yet localization of Cx43/ZO-1 interaction to this region indicated the presence of Cx43. Therefore, we hypothesized that Cx43 occurs in the perinexus at a lower concentration per unit membrane than in the GJ itself, making it difficult to visualize. To overcome this, the Duolink protein-protein interaction assay was used to detect Cx43. Duolink labeling of cardiomyocytes localized Cx43 to the perinexus. Quantification demonstrated that signal in the perinexus was lower than in the GJ but significantly higher than in nonjunctional regions. Additionally, Duolink of Triton X-100-extracted cultures suggested that perinexal Cx43 is nonjunctional. Importantly, the voltage gated sodium channel Na(v)1.5, which is responsible for initiation of the action potential, was found to interact with perinexal Cx43 but not with ZO-1. This work provides a detailed characterization of the structure of the perinexus at the GJ edge and indicates that one of its potential functions in the heart may be in facilitating conduction of action potential.


Asunto(s)
Conexina 43/metabolismo , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Animales , Western Blotting , Células Cultivadas , Inmunohistoquímica , Ratas , Ratas Sprague-Dawley
15.
Biochim Biophys Acta ; 1818(8): 1831-43, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21856279

RESUMEN

The precise spatial order of gap junctions at intercalated disks in adult ventricular myocardium is thought vital for maintaining cardiac synchrony. Breakdown or remodeling of this order is a hallmark of arrhythmic disease of the heart. The principal component of gap junction channels between ventricular cardiomyocytes is connexin43 (Cx43). Protein-protein interactions and modifications of the carboxyl-terminus of Cx43 are key determinants of gap junction function, size, distribution and organization during normal development and in disease processes. Here, we review data on the role of proteins interacting with the Cx43 carboxyl-terminus in the regulation of cardiac gap junction organization, with particular emphasis on Zonula Occludens-1. The rapid progress in this area suggests that in coming years we are likely to develop a fuller understanding of the molecular mechanisms causing pathologic remodeling of gap junctions. With these advances come the promise of novel approach to the treatment of arrhythmia and the prevention of sudden cardiac death. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.


Asunto(s)
Conexina 43/química , Uniones Comunicantes/fisiología , Miocardio/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Arritmias Cardíacas/terapia , Conexina 43/metabolismo , Muerte Súbita Cardíaca/prevención & control , Ventrículos Cardíacos/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Mapeo de Interacción de Proteínas/métodos , Estructura Terciaria de Proteína , Uniones Estrechas/metabolismo
16.
Mol Biol Cell ; 22(9): 1516-28, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21411628

RESUMEN

Connexin 43 (Cx43) is a gap junction (GJ) protein widely expressed in mammalian tissues that mediates cell-to-cell coupling. Intercellular channels comprising GJ aggregates form from docking of paired connexons, with one each contributed by apposing cells. Zonula occludens-1 (ZO-1) binds the carboxy terminus of Cx43, and we have previously shown that inhibition of the Cx43/ZO-1 interaction increases GJ size by 48 h. Here we demonstrated that increases in GJ aggregation occur within 2 h (∼Cx43 half-life) following disruption of Cx43/ZO-1. Immunoprecipitation and Duolink protein-protein interaction assays indicated that inhibition targets ZO-1 binding with Cx43 in GJs as well as connexons in an adjacent domain that we term the "perinexus." Consistent with GJ size increases being matched by decreases in connexons, inhibition of Cx43/ZO-1 reduced the extent of perinexal interaction, increased the proportion of connexons docked in GJs relative to undocked connexons in the plasma membrane, and increased GJ intercellular communication while concomitantly decreasing hemichannel-mediated membrane permeance in contacting, but not noncontacting, cells. ZO-1 small interfering RNA and overexpression experiments verified that loss and gain of ZO-1 function govern the transition of connexons into GJs. It is concluded that ZO-1 regulates the rate of undocked connexon aggregation into GJs, enabling dynamic partitioning of Cx43 channel function between junctional and proximal nonjunctional domains of plasma membrane.


Asunto(s)
Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Comunicación Celular , Ciclo Celular , Membrana Celular/metabolismo , Conexina 43/química , Conexina 43/genética , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Inmunoprecipitación , Proteínas de la Membrana/genética , Fosfoproteínas/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño , Proteína de la Zonula Occludens-1
17.
J Mol Cell Cardiol ; 48(3): 550-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19560469

RESUMEN

Regenerative healing is the process by which injured tissues are restored to their original structure and function. Many species are capable of healing in this manner. However, in mammals the healing response in most tissues is marked by fibroblast proliferation and scar tissue deposition. While scarring contributes to efficient resolution of mammalian wounds and restoration of at least partial structural and functional support, the final result of scar formation can be more deleterious than the initial insult. This is especially true in the heart, which is sensitive to electrical heterogeneities and altered mechanical properties produced by scarring. Several therapeutic modalities promoting regeneration in skin wounds have been developed that modulate various aspects of the healing process. Targets include cytokine stimulation, control of fibroblast activation, modulation of gap junctions, and stem cell differentiation. Here, we review and compare mechanisms of injury, repair, and scarring in the skin and heart and discuss the promise and caveats of future therapies that may translate to improving repair of myocardial tissues.


Asunto(s)
Cicatriz/fisiopatología , Miocardio/metabolismo , Piel/patología , Animales , Cicatriz/metabolismo , Humanos , Modelos Biológicos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/patología , Piel/metabolismo , Cicatrización de Heridas/fisiología
18.
Trends Biotechnol ; 26(4): 173-80, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18295916

RESUMEN

Fibrotic scars deposited during skin wound healing can cause disfiguration and loss of dermal function. Scar differentiation involves inputs from multiple cell types in a predictable and overlapping sequence of cellular events that includes inflammation, migration/proliferation and extracellular matrix deposition. Research into the molecular mechanisms underpinning these processes in embryonic and adult wounds has contributed to the development of a growing number of novel therapeutic approaches for improving scar appearance. This review discusses some of these emerging strategies for shifting the balance of healing from scarring to regeneration in the context of non-pathological wounds. Particular focus is given to potential therapies based on transforming growth factor (TGF)-beta signaling and recent unexpected findings involving targeting of gap junctional connexins. Lessons learned in promoting scarless healing of cutaneous injuries might provide a basis for regenerative healing in other scenarios, such as spinal cord rupture or myocardial infarction.


Asunto(s)
Cicatriz/fisiopatología , Cicatriz/terapia , Piel/patología , Cicatrización de Heridas/fisiología , Adulto , Cicatriz/patología , Colágeno/inmunología , Colágeno/metabolismo , Conexinas/genética , Conexinas/metabolismo , Conexinas/uso terapéutico , Embrión de Mamíferos/inmunología , Embrión de Mamíferos/fisiopatología , Matriz Extracelular/inmunología , Matriz Extracelular/patología , Feto/inmunología , Feto/fisiopatología , Humanos , Inflamación/complicaciones , Inflamación/embriología , Inflamación/inmunología , Inflamación/fisiopatología , Ingeniería de Proteínas , Piel/metabolismo , Piel/fisiopatología , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos
19.
J Lipid Res ; 45(10): 1859-67, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15258197

RESUMEN

Humans and laboratory animals recognize human modified LDL as immunogenic. Immune complexes (ICs) isolated from human sera contain malondialdehyde-modified LDL (MDA-LDL) and N (epsilon)(carboxymethyl)lysine-modified LDL (CML-LDL) as well as antibodies reacting with MDA-LDL, copper-oxidized LDL (OxLDL), CML-LDL, and advanced glycosylation end product (AGE)-modified LDL. OxLDL and AGE-LDL antibodies isolated from human sera recognize the same LDL modifications and do not react with modified non-LDL proteins. Rabbit antibodies have different reactivity patterns: MDA-LDL antibodies react strongly with MDA-LDL and MDA-BSA but weakly with OxLDL; OxLDL antibodies react strongly with OxLDL and weakly with MDA-LDL; CML-LDL antibodies react with CML-LDL > CML-BSA > AGE-LDL > OxLDL; AGE-LDL antibodies react strongly with AGE-LDL, react weakly with OxLDL, and do not react with CML-LDL. Thus, human and rabbit antibodies seem to recognize different epitopes. Capture assays carried out with all rabbit antibodies showed binding of apolipoprotein B-rich lipoproteins isolated from ICs, suggesting that laboratory-generated epitopes are expressed by in vivo-modified LDL, although they are not necessarily recognized by the human immune system. Thus, the definition of immunogenic forms of modified LDL eliciting human autoimmune responses requires the isolation and characterization of autoantibodies and modified LDL from human samples, whereas rabbit antibodies can be used to detect in vivo-modified human LDL.


Asunto(s)
Anticuerpos Heterófilos/inmunología , Autoanticuerpos/inmunología , Lipoproteínas LDL/inmunología , Animales , Especificidad de Anticuerpos , Complejo Antígeno-Anticuerpo/análisis , Complejo Antígeno-Anticuerpo/aislamiento & purificación , Apolipoproteínas B/inmunología , Reacciones Cruzadas , Epítopos , Productos Finales de Glicación Avanzada/inmunología , Humanos , Conejos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...