Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Regul Integr Comp Physiol ; 326(6): R528-R551, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38497126

RESUMEN

In pilot work, we showed that somatic nerve transfers can restore motor function in long-term decentralized dogs. We continue to explore the effectiveness of motor reinnervation in 30 female dogs. After anesthesia, 12 underwent bilateral transection of coccygeal and sacral (S) spinal roots, dorsal roots of lumbar (L)7, and hypogastric nerves. Twelve months postdecentralization, eight underwent transfer of obturator nerve branches to pelvic nerve vesical branches, and sciatic nerve branches to pudendal nerves, followed by 10 mo recovery (ObNT-ScNT Reinn). The remaining four were euthanized 18 mo postdecentralization (Decentralized). Results were compared with 18 Controls. Squat-and-void postures were tracked during awake cystometry. None showed squat-and-void postures during the decentralization phase. Seven of eight ObNT-ScNT Reinn began showing such postures by 6 mo postreinnervation; one showed a return of defecation postures. Retrograde dyes were injected into the bladder and urethra 3 wk before euthanasia, at which point, roots and transferred nerves were electrically stimulated to evaluate motor function. Upon L2-L6 root stimulation, five of eight ObNT-ScNT Reinn showed elevated detrusor pressure and four showed elevated urethral pressure, compared with L7-S3 root stimulation. After stimulation of sciatic-to-pudendal transferred nerves, three of eight ObNT-ScNT Reinn showed elevated urethral pressure; all showed elevated anal sphincter pressure. Retrogradely labeled neurons were observed in L2-L6 ventral horns (in laminae VI, VIII, and IX) of ObNT-ScNT Reinn versus Controls in which labeled neurons were observed in L7-S3 ventral horns (in lamina VII). This data supports the use of nerve transfer techniques for the restoration of bladder function.NEW & NOTEWORTHY This data supports the use of nerve transfer techniques for the restoration of bladder function.


Asunto(s)
Canal Anal , Neuronas Motoras , Transferencia de Nervios , Recuperación de la Función , Uretra , Vejiga Urinaria , Animales , Transferencia de Nervios/métodos , Perros , Femenino , Vejiga Urinaria/inervación , Uretra/inervación , Canal Anal/inervación , Canal Anal/cirugía , Neuronas Motoras/fisiología , Regeneración Nerviosa/fisiología , Nervio Pudendo/cirugía , Nervio Pudendo/fisiopatología
2.
Int J Mol Sci ; 25(3)2024 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-38339140

RESUMEN

A role for substance P has been proposed in musculoskeletal fibrosis, with effects mediated through transforming growth factor beta (TGFß). We examined the in vitro effects of substance P on proliferation, collagen secretion, and collagen deposition in rat primary dermal fibroblasts cultured in medium containing 10% fetal bovine serum, with or without TGFß. In six-day cultures, substance P increased cell proliferation at concentrations from 0.0002 to 100 nM. TGFß increased proliferation at concentrations from 0.0002 to 2 pg/mL, although higher concentrations inhibited proliferation. Substance P treatment alone at concentrations of 100, 0.2, and 0.00002 nM did not increase collagen deposition per cell, yet when combined with TGFß (5 ng/mL), increased collagen deposition compared to TGFß treatment alone. Substance P treatment (100 nM) also increased smooth muscle actin (SMA) expression at 72 h of culture at a level similar to 5 ng/mL of TGFß; only TGFß increased SMA at 48 h of culture. Thus, substance P may play a role in potentiating matrix deposition in vivo when combined with TGFß, although this potentiation may be dependent on the concentration of each factor. Treatments targeting substance P may be a viable strategy for treating fibrosis where both substance P and TGFß play roles.


Asunto(s)
Sustancia P , Factor de Crecimiento Transformador beta , Ratas , Animales , Factor de Crecimiento Transformador beta/metabolismo , Sustancia P/farmacología , Sustancia P/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Colágeno/metabolismo , Fibrosis , Factor de Crecimiento Transformador beta1/metabolismo
3.
Cell ; 133(3): 415-26, 2008 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-18455983

RESUMEN

Immune homeostasis is essential for the normal functioning of the immune system, and its breakdown leads to fatal inflammatory diseases. We report here the identification of a member of the tumor necrosis factor-alpha-induced protein-8 (TNFAIP8) family, designated TIPE2, that is required for maintaining immune homeostasis. TIPE2 is preferentially expressed in lymphoid tissues, and its deletion in mice leads to multiorgan inflammation, splenomegaly, and premature death. TIPE2-deficient animals are hypersensitive to septic shock, and TIPE2-deficient cells are hyper-responsive to Toll-like receptor (TLR) and T cell receptor (TCR) activation. Importantly, TIPE2 binds to caspase-8 and inhibits activating protein-1 and nuclear factor-kappaB activation while promoting Fas-induced apoptosis. Inhibiting caspase-8 significantly blocks the hyper-responsiveness of TIPE2-deficient cells. These results establish that TIPE2 is an essential negative regulator of TLR and TCR function, and its selective expression in the immune system prevents hyperresponsiveness and maintains immune homeostasis.


Asunto(s)
Homeostasis , Inmunidad , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Apoptosis , Caspasa 8/metabolismo , Línea Celular , Encefalomielitis Autoinmune Experimental , Humanos , Inmunidad Celular , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Tejido Linfoide/inmunología , Macrófagos/inmunología , Ratones , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Choque Séptico/inmunología , Transducción de Señal , Médula Espinal/inmunología , Linfocitos T/inmunología , Factor de Transcripción AP-1/metabolismo , Receptor fas/metabolismo
4.
Int J Mol Sci ; 24(18)2023 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-37762168

RESUMEN

The matricellular protein cell communication factor 2/connective tissue growth factor (CCN2/CTGF) is critical to development of neuromuscular fibrosis. Here, we tested whether anti-CCN2 antibody treatment will reduce established forepaw fibro-degenerative changes and improve function in a rat model of overuse injury. Adult female rats performed a high repetition high force (HRHF) task for 18 weeks. Tissues were collected from one subset after 18 wks (HRHF-Untreated). Two subsets were provided 6 wks of rest with concurrent treatment with anti-CCN2 (HRHF-Rest/anti-CCN2) or IgG (HRHF-Rest/IgG). Results were compared to IgG-treated Controls. Forepaw muscle fibrosis, neural fibrosis and entheseal damage were increased in HRHF-Untreated rats, compared to Controls, and changes were ameliorated in HRHF-Rest/anti-CCN2 rats. Anti-CCN2 treatment also reduced phosphorylated-ß-catenin (pro-fibrotic protein) in muscles and distal bone/entheses complex, and increased CCN3 (anti-fibrotic) in the same tissues, compared to HRHF-Untreated rats. Grip strength declines and mechanical sensitivity observed in HRHF-Untreated improved with rest; grip strength improved further in HRHF-Rest/anti-CCN2. Grip strength declines correlated with muscle fibrosis, entheseal damage, extraneural fibrosis, and decreased nerve conduction velocity, while enhanced mechanical sensitivity (a pain-related behavior) correlated with extraneural fibrosis. These studies demonstrate that blocking CCN2 signaling reduces established forepaw neuromuscular fibrosis and entheseal damage, which improves forepaw function, following overuse injury.


Asunto(s)
Trastornos de Traumas Acumulados , Fibromialgia , Femenino , Animales , Ratas , Factor de Crecimiento del Tejido Conjuntivo , Fibrosis , Inmunoglobulina G
5.
FASEB J ; 34(5): 6554-6569, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32227398

RESUMEN

Tissue fibrosis is a hallmark of overuse musculoskeletal injuries and contributes to functional declines. We tested whether inhibition of CCN2 (cellular communication network factor 2, previously known as connective tissue growth factor, CTGF) using a specific antibody (termed FG-3019 or pamrevlumab) reduces established overuse-induced muscle fibrosis in a clinically relevant rodent model of upper extremity overuse injury. Young adult rats performed a high repetition high force (HRHF) reaching and lever-pulling task for 18 weeks, after first being shaped for 6 weeks to learn this operant task. Rats were then euthanized (HRHF-Untreated), or rested and treated for 6 weeks with FG-3019 (HRHF-Rest/FG-3019) or a human IgG as a vehicle control (HRHF-Rest/IgG). HRHF-Untreated and HRHF-Rest/IgG rats had higher muscle levels of several fibrosis-related proteins (TGFß1, CCN2, collagen types I and III, and FGF2), and higher muscle numbers of alpha SMA and pERK immunopositive cells, compared to control rats. Each of these fibrogenic changes was restored to control levels by the blocking of CCN2 signaling in HRHF-Rest/FG-3019 rats, as were HRHF task-induced increases in serum CCN2 and pro-collagen I intact N-terminal protein. Levels of cleaved CCN3, an antifibrotic protein, were lowered in HRHF-Untreated and HRHF-Rest/IgG rats, compared to control rats, yet elevated back to control levels in HRHF-Rest/FG-3019 rats. Significant grip strength declines observed in HRHF-Untreated and HRHF-Rest/IgG rats, were restored to control levels in HRHF-Rest/FG-3019 rats. These results are highly encouraging for use of FG-3019 for therapeutic treatment of persistent skeletal muscle fibrosis, such as those induced with chronic overuse.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/antagonistas & inhibidores , Trastornos de Traumas Acumulados/complicaciones , Modelos Animales de Enfermedad , Fibrosis/prevención & control , Músculo Esquelético/fisiología , Animales , Colágeno Tipo I/metabolismo , Femenino , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Músculo Esquelético/lesiones , Ratas , Ratas Sprague-Dawley
6.
Connect Tissue Res ; 62(1): 133-149, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33030055

RESUMEN

AIM: To examine the chronic effect of force on mRNA and protein expression levels of fibrosis-related genes in flexor digitorum muscles in a rat model of repetitive overuse injury that induces muscle fibrosis at high force levels. MATERIALS AND METHODS: Two groups of rats were trained to perform a voluntary repetitive lever-pulling task at either a high (HFHR) or a low force (LFHR) for 18 weeks, while a control group (FRC) performed no task. RNA and protein were prepared from forelimb flexor digitorum muscles. Fibrosis-related gene RNA transcripts were evaluated using quantitative PCR (qPCR) and analyzed using the geometric mean of three housekeeping genes or the mean of each individually as reference. Protein levels were quantified using ELISA, western blot, or immunohistofluorescence. RESULTS: Of eight fibrosis-related mRNAs examined, only FGF2 demonstrated a consistent significant increase in the HFHR group, compared to the FRC group. However, protein amounts of collagen type 1, collagen type 3, and TGFß1 were significantly higher in the HFHR, compared to the FRC and LFHR groups, while CCN2 and FGF2 were higher in both HFHR and LFHR, compared to the FRC group. CONCLUSIONS: Our results suggest that there is steady-state transcription of fibrogenic genes in muscles with established fibrosis, implying that post-transcriptional processes are responsible for the increased protein levels of fibrotic factors during muscle overuse conditions. We hypothesize that targeting such pathways represents a valid approach to treat overuse injury. Alternatively, FGF2 gene expression may represent a valid target for therapy.


Asunto(s)
Músculo Esquelético , Animales , Colágeno Tipo I , Trastornos de Traumas Acumulados/genética , Trastornos de Traumas Acumulados/patología , Factor 2 de Crecimiento de Fibroblastos , Fibrosis , Músculo Esquelético/patología , ARN , Ratas , Ratas Sprague-Dawley
7.
J Musculoskelet Neuronal Interact ; 19(4): 396-411, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31789291

RESUMEN

OBJECTIVES: Fibrosis is one contributing factor in motor dysfunction and discomfort in patients with overuse musculoskeletal disorders. We pharmacologically targeted the primary receptor for Substance P, neurokinin-1, using a specific antagonist (NK1RA) in a rat model of overuse with the goal of improving tissue fibrosis and discomfort. METHODS: Female rats performed a low repetition, high force (LRHF) grasping task for 12 weeks, or performed the task for 12 weeks before being placed on a four week rest break, with or without simultaneous NK1RA treatment. Results were compared to control rats (untreated, or treated 4 weeks with NK1RA or vehicle). RESULTS: Rest improved LRHF-induced declines in grip strength, although rest plus NK1RA treatment (Rest/NK1RA) rescued it. Both treatments improved LRHF-induced increases in muscle TGFß1 and collagen type 1 levels, forepaw mechanical hypersensitivity (Rest/NK1RA more effectively), macrophage influx into median nerves, and enhanced collagen deposition in forepaw dermis. Only Rest/NK1RA reduced muscle hypercellularity. However, LRHF+4wk Rest /NK1RA rats showed hyposensitivity to noxious hot temperatures. CONCLUSIONS: While the NK1RA induced hot temperature hyposensitivity should be taken into consideration if this or related drug were used long-term, the NK1RA more effectively reduced muscle hypercellularity and improved grip strength and forepaw mechanical hypersensitivity.


Asunto(s)
Fibrosis/metabolismo , Fuerza de la Mano/fisiología , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Antagonistas del Receptor de Neuroquinina-1/farmacología , Desempeño Psicomotor/efectos de los fármacos , Animales , Citocinas/metabolismo , Femenino , Fibrosis/patología , Fuerza Muscular/fisiología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ratas , Ratas Sprague-Dawley
8.
Immunopharmacol Immunotoxicol ; 39(6): 318-329, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28929835

RESUMEN

CONTEXT: We have reported previously that DAB389IL-2 recombinant fusion toxin targets IL-2R bearing CD4+ cells, and suppresses demyelinating disease in acute (A) - and chronic (C) - experimental autoimmune encephalomyelitis (EAE) animal models of multiple sclerosis. OBJECTIVES: The present study was undertaken to investigate the effect of DAB389IL-2 treatment on various cytokine-secreting cell populations in A-EAE and C-EAE mice. MATERIALS AND METHODS: The effects of DAB389IL-2 at doses of 200-, 800-, or 1600 kU administered i.v. on days 11-13 and 15 on the clinical score and cytokine-secreting cell populations were examined using flow cytometry. RESULTS: C-EAE mice treated with 1600kU DAB389IL-2, but not A-EAE mice treated with 800 kU had significantly reduced disease. The CD3+CD25+ sub-population in spleens and spinal cords of A-EAE mice treated with 800 kU DAB389IL-2 a was increased, whereas in C-EAE mice treated with 1600 kU this population was increased. DAB389IL-2 treatment reduced CD3+CD4+, CD3+CD8+, CD4+CD8+, CD3+IL-2+, CD3+IFN-γ+ and CD3+TNF-α+ T cell subpopulations in the spinal cord in A-EAE, and C-EAE mice on day 16. CD11b+ macrophages that were IL-2-, IFN-γ-, and TNF-α- positive were reduced in A-EAE mice. DAB389IL-2 treatment reduced CD19+ B-cells positive for IL-2 or CD11b+ in the spinal cord in acute and chronic disease. DAB389IL-2 treatment also reduced lymph node CD3+CD8+, CD4+CD8+, CD3+CD25+ populations on day 16, and lymph node CD3+IL-10+ and peripheral blood CD3+CD25+ populations on day 24. DISCUSSION AND CONCLUSIONS: Our study demonstrates that DAB389IL-2 fusion toxin suppresses EAE in a dose-dependent manner, and alters inflammatory cell sub-populations during disease development.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Toxina Diftérica/farmacología , Interleucina-2/farmacología , Macrófagos/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/metabolismo , Activación de Linfocitos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Bazo/efectos de los fármacos , Bazo/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Int J Cancer ; 136(1): 117-26, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24895110

RESUMEN

The role of STAT2 in mediating the antigrowth effects of type I interferon (IFN) is well-documented in vitro. Yet evidence of IFN-activated STAT2 as having tumor suppressor function in vivo and participation in antitumor immunity is lacking. Here we show in a syngeneic tumor transplantation model that STAT2 reduces tumor growth. Stat2(-/-) mice formed larger tumors compared to wild type (WT) mice. IFN-ß treatment of Stat2(-/-) mice did not cause tumor regression. Gene expression analysis revealed a small subset of immunomodulatory genes to be downregulated in tumors established in Stat2(-/-) mice. Additionally, we found tumor antigen cross-presentation by Stat2(-/-) dendritic cells to T cells to be impaired. Adoptive transfer of tumor antigen specific CD8(+) T cells primed by Stat2(-/-) dendritic cells into tumor-bearing Stat2(-/-) mice did not induce tumor regression with IFN-ß intervention. We observed that an increase in the number of CD4(+) and CD8(+) T cells in the draining lymph nodes of IFN-ß-treated tumor-bearing WT mice was absent in IFN-ß treated Stat2(-/-) mice. Thus our study provides evidence for further evaluation of STAT2 function in cancer patients receiving type I IFN based immunotherapy.


Asunto(s)
Antineoplásicos/farmacología , Interferón beta/farmacología , Neoplasias Pulmonares/secundario , Melanoma Experimental/patología , Factor de Transcripción STAT2/fisiología , Carga Tumoral/inmunología , Traslado Adoptivo , Animales , Línea Celular Tumoral , Medios de Cultivo Condicionados , Células Dendríticas , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Trasplante de Neoplasias , Factor de Transcripción STAT1/metabolismo , Linfocitos T/inmunología
10.
Clin Immunol ; 158(2): 231-41, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25814173

RESUMEN

Chronic inflammation is increased in patients with chronic kidney disease (CKD) and contributes to cardiovascular morbidity and mortality. Specific immune mechanisms and pathways that drive and maintain chronic inflammation in CKD are not well described. The TAM ligands (Gas6 and protein S) and receptors (Axl and Mer) have been recently recognized as playing a prominent role in immune regulation. The receptors exist in both soluble and cell-bound forms; the soluble receptors (sAxl and sMer) are believed to compete with the bound receptors and thus inhibit their function. In this study, we determined the expression of cell-bound and soluble TAM proteins in patients with CKD. CKD patients had significantly lower expression of Mer in monocytes, yet increased expression of soluble TAM receptors sAxl and sMer in plasma compared to controls. The metalloproteinase ADAM 17, responsible for cleavage of Mer to its soluble form, was increased in patient monocytes. Elevated levels of soluble TAM receptors were more evident in patients with progressive renal failure. These observations suggest that functional deficiency of TAM receptor-mediated regulation of inflammation may contribute to chronic inflammation in patients with CKD.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína S/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Insuficiencia Renal Crónica/metabolismo , Humanos , Inflamación , Péptidos y Proteínas de Señalización Intercelular/genética , Monocitos/metabolismo , Proteína S/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Insuficiencia Renal Crónica/inmunología , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
11.
Exp Mol Pathol ; 96(1): 108-17, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23872438

RESUMEN

In multiple sclerosis (MS) and its rodent model, experimental autoimmune encephalomyelitis (EAE), activated CD4(+) T cells with upregulated IL-2R mediate inflammation and demyelination in the central nervous system (CNS). DAB(389)IL-2, a chimeric fusion protein of IL-2 and diphtheria toxin, inhibits human and rodent IL-2 activated T cells that express the high affinity interleukin-2 receptor. In the present study, DAB(389)IL-2 was used to treat rats with EAE. We wanted to investigate the possibility that DAB(389)IL-2 could prevent tissue destruction within the CNS. We used a suboptimal dose of DAB(389)IL-2 that allowed substantial transmigration of inflammatory cells across the blood-brain barrier. DAB(389)IL-2 inhibited infiltration of CD4(+), CD8(+), CD25(+) and TCR αß(+) associated mononuclear cells and inflammatory macrophages in the spinal cord on day 13 post-immunization, at the peak of disease. Gene expression study showed that DAB(389)IL-2 treatment suppressed TNF-α and IFN-γ as well as IL-10 cytokine gene expression in the spinal cord of rats with EAE on day 13. DAB(389)IL-2 in vitro treatment suppressed cytotoxicity of MBP-activated T cells from rats with EAE against oligodendrocytes in culture by 66%. Astrocytes were less targeted by MBP activated T cells in vitro. This study suggests that DAB(389)IL-2 directly targets CD4(+) and CD25(+) (IL-2R) T cells and effector T cell function and also indirectly suppresses the activation of macrophage CD169(+) (ED3(+)) and microglia CD11b/c (OX42(+)) populations in the CNS.


Asunto(s)
Antineoplásicos/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Sistema Nervioso Central/efectos de los fármacos , Toxina Diftérica/uso terapéutico , Encefalomielitis Autoinmune Experimental/prevención & control , Inflamación/prevención & control , Interleucina-2/uso terapéutico , Neuroglía/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Astrocitos/patología , Western Blotting , Linfocitos T CD4-Positivos/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Técnicas para Inmunoenzimas , Inflamación/inmunología , Inflamación/patología , Interferón gamma/metabolismo , Activación de Linfocitos/efectos de los fármacos , Neuroglía/inmunología , Neuroglía/patología , ARN Mensajero/genética , Ratas , Ratas Endogámicas Lew , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes de Fusión/uso terapéutico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
J Immunol ; 189(7): 3508-20, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22942426

RESUMEN

Mer tyrosine kinase (MerTK) is a major macrophage apoptotic cell (AC) receptor. Its functional impairment promotes autoimmunity and atherosclerosis, whereas overexpression correlates with poor prognosis in cancer. However, little is known about mechanisms regulating MerTK expression in humans. We found that MerTK expression is heterogenous among macrophage subsets, being mostly restricted to anti-inflammatory M2c (CD14(+)CD16(+)CD163(+)CD204(+)CD206(+)CD209(-)) cells, differentiated by M-CSF or glucocorticoids. Small numbers of MerTK(+) "M2c-like" cells are also detectable among circulating CD14(bright)CD16(+) monocytes. MerTK expression levels adapt to changing immunologic environment, being suppressed in M1 and M2a macrophages and in dendritic cells. Remarkably, although glucocorticoid-induced differentiation is IL-10 independent, M-CSF-driven M2c polarization and related MerTK upregulation require IL-10. However, neither IL-10 alone nor TGF-ß are sufficient to fully differentiate M2c (CD16(+)CD163(+)MerTK(+)) macrophages. M-CSF and IL-10, both released by T lymphocytes, may thus be required together to promote regulatory T cell-mediated induction of anti-inflammatory monocytes-macrophages. MerTK enables M2c macrophages to clear early ACs more efficiently than other macrophage subsets, and it mediates AC clearance by CD14(bright)CD16(+) monocytes. Moreover, M2c cells release Gas6, which in turn amplifies IL-10 secretion via MerTK. IL-10-dependent induction of the Gas6/MerTK pathway may, therefore, constitute a positive loop for M2c macrophage homeostasis and a critical checkpoint for maintenance of anti-inflammatory conditions. Our findings give new insight into human macrophage polarization and favor a central role for MerTK in regulation of macrophage functions. Eliciting M2c polarization can have therapeutic utility for diseases such as lupus, in which a defective AC clearance contributes to initiate and perpetuate the pathological process.


Asunto(s)
Apoptosis/inmunología , Movimiento Celular/inmunología , Polaridad Celular/inmunología , Macrófagos/enzimología , Macrófagos/inmunología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Apoptosis/genética , Comunicación Autocrina/genética , Comunicación Autocrina/inmunología , Diferenciación Celular/inmunología , Movimiento Celular/genética , Polaridad Celular/genética , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Humanos , Inmunofenotipificación , Macrófagos/citología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Activación Transcripcional/inmunología , Tirosina Quinasa c-Mer
13.
Brain Behav Immun Health ; 35: 100714, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38111687

RESUMEN

Poor sleep is thought to enhance pain via increasing peripheral and/or central sensitization. Aerobic exercise, conversely, relives pain via reducing sensitization, among other mechanisms. This raises two clinical questions: (1) does poor sleep contribute to the transition from acute-to-persistent pain, and (2) can exercise protect against this transition? This study tested these questions and explored underlying mechanisms in a controlled injury model. Twenty-nine adult female Sprague-Dawley rats performed an intensive lever-pulling task for 4 weeks to induce symptoms consistent with clinical acute-onset overuse injury. Rats were then divided into three groups and exposed for 4 weeks to either: voluntary exercise via access to a running wheel, sleep disturbance, or both. Pain-related behaviours (forepaw mechanical sensitivity, reflexive grip strength), systemic levels of brain derived neurotrophic factor (BDNF), estradiol and corticosterone, and white blood cells (WBC) were assessed pre-injury, post-injury and post-intervention. Mechanical sensitivity increased post-injury and remained elevated with sleep disturbance alone, but decreased to pre-injury levels with exercise both with and without sleep disturbance. Reflexive grip strength decreased post-injury but recovered post-intervention-more with exercise than sleep disturbance. BDNF increased with sleep disturbance alone, remained at pre-injury levels with exercise regardless of sleep, and correlated with mechanical sensitivity. WBCs and estradiol increased with exercise alone and together with sleep disturbance, respectively. Corticosterone was not impacted by injury/intervention. Findings provide preliminary evidence for a role of poor sleep in the transition from acute-to-persistent pain, and the potential for aerobic exercise to counter these effects. BDNF might have a role in these relationships.

14.
Front Pain Res (Lausanne) ; 5: 1394017, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38770243

RESUMEN

Introduction: Low back pain is the most common type of chronic pain. We examined pain-related behaviors across 18 weeks in rats that received injury to one or two lumbar intervertebral discs (IVD) to determine if multi-level disc injuries enhance/prolong pain. Methods: Twenty-three Sprague-Dawley adult female rats were used: 8 received disc puncture (DP) of one lumbar IVD (L5/6, DP-1); 8 received DP of two lumbar IVDs (L4/5 & L5/6, DP-2); 8 underwent sham surgery. Results: DP-2 rats showed local (low back) sensitivity to pressure at 6- and 12-weeks post-injury, and remote sensitivity to pressure (upper thighs) at 12- and 18-weeks and touch (hind paws) at 6, 12 and 18-weeks. DP-1 rats showed local and remote pressure sensitivity at 12-weeks only (and no tactile sensitivity), relative to Sham DP rats. Both DP groups showed reduced distance traveled during gait testing over multiple weeks, compared to pre-injury; only DP-2 rats showed reduced distance relative to Sham DP rats at 12-weeks. DP-2 rats displayed reduced positive interactions with a novel adult female rat at 3-weeks and hesitation and freezing during gait assays from 6-weeks onwards. At study end (18-weeks), radiological and histological analyses revealed reduced disc height and degeneration of punctured IVDs. Serum BDNF and TNFα levels were higher at 18-weeks in DP-2 rats, relative to Sham DP rats, and levels correlated positively with remote sensitivity in hind paws (tactile) and thighs (pressure). Discussion: Thus, multi-level disc injuries resulted in earlier, prolonged and greater discomfort locally and remotely, than single-level disc injury. BDNF and TNFα may have contributing roles.

15.
JBMR Plus ; 7(9): e10783, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37701153

RESUMEN

We have an operant model of reaching and grasping in which detrimental bone remodeling is observed rather than beneficial adaptation when rats perform a high-repetition, high-force (HRHF) task long term. Here, adult female Sprague-Dawley rats performed an intense HRHF task for 18 weeks, which we have shown induces radial trabecular bone osteopenia. One cohort was euthanized at this point (to assay the bone changes post task; HRHF-Untreated). Two other cohorts were placed on 6 weeks of rest while being simultaneously treated with either an anti-CCN2 (FG-3019, 40 mg/kg body weight, ip; twice per week; HRHF-Rest/anti-CCN2), or a control IgG (HRHF-Rest/IgG), with the purpose of determining which might improve the trabecular bone decline. Results were compared with food-restricted control rats (FRC). MicroCT analysis of distal metaphysis of radii showed decreased trabecular bone volume fraction (BV/TV) and thickness in HRHF-Untreated rats compared with FRCs; responses improved with HRHF-Rest/anti-CCN2. Rest/IgG also improved trabecular thickness but not BV/TV. Histomorphometry showed that rest with either treatment improved osteoid volume and task-induced increases in osteoclasts. Only the HRHF-Rest/anti-CCN2 treatment improved osteoblast numbers, osteoid width, mineralization, and bone formation rate compared with HRHF-Untreated rats (as well as the latter three attributes compared with HRHF-Rest/IgG rats). Serum ELISA results were in support, showing increased osteocalcin and decreased CTX-1 in HRHF-Rest/anti-CCN2 rats compared with both HRHF-Untreated and HRHF-Rest/IgG rats. These results are highly encouraging for use of anti-CCN2 for therapeutic treatment of bone loss, such as that induced by chronic overuse. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

16.
Nephrol Dial Transplant ; 27(11): 4166-72, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22907951

RESUMEN

BACKGROUND: The TAM receptors (tyro3, axl and mer) and their ligands (vitamin K-dependent proteins-Gas6 and Protein S) are crucial modulators of inflammation, which may be relevant in chronic kidney disease (CKD). Gas6 and axl have multiple roles in mediating vascular atherosclerosis and injury, thrombosis and inflammation, yet nothing is known about the Gas6-axl pathway in humans with CKD. Given the prevalence of chronic inflammation and vascular disease in this population, we measured TAM ligands in patients with various levels of renal function. METHODS: Gas6 and protein S were quantified in the plasma by ELISA in three patient groups: end-stage renal disease on chronic hemodialysis (HD), CKD and normal controls. RESULTS: Significantly increased levels of Gas6 and protein S were found in CKD patients compared with normal controls (P < 0.01 and <0.001, respectively). In HD patients, Gas6 levels were elevated compared with controls (P < 0.001) and positively associated with low albumin (r = 0.33; P = 0.01), dialysis vintage (r = 0.36; P = 0.008) and IV iron administration (r = 0.33; P = 0.01). The levels of Gas6 rose with CKD stage and were inversely associated with estimated GFR (P < 0.0001). CONCLUSIONS: Dysregulation of circulating Gas6 is associated with renal disease and inversely proportional to renal function. Low albumin and higher IV iron administration were associated with higher Gas6 levels, suggesting a possible connection between inflammation and oxidative stress mediated by iron. Protein S levels were also elevated in CKD patients, but the relevance of this finding needs to be further investigated.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/sangre , Fallo Renal Crónico/sangre , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Precursores de Proteínas/sangre , Proteína S/análisis , Protrombina , Diálisis Renal
17.
J Exp Med ; 201(4): 567-77, 2005 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-15710649

RESUMEN

Decay-accelerating factor ([DAF] CD55) is a glycosylphosphatidylinositol-anchored membrane inhibitor of complement with broad clinical relevance. Here, we establish an additional and unexpected role for DAF in the suppression of adaptive immune responses in vivo. In both C57BL/6 and BALB/c mice, deficiency of the Daf1 gene, which encodes the murine homologue of human DAF, significantly enhanced T cell responses to active immunization. This phenotype was characterized by hypersecretion of interferon (IFN)-gamma and interleukin (IL)-2, as well as down-regulation of the inhibitory cytokine IL-10 during antigen restimulation of lymphocytes in vitro. Compared with wild-type mice, Daf1(-/-) mice also displayed markedly exacerbated disease progression and pathology in a T cell-dependent experimental autoimmune encephalomyelitis (EAE) model. However, disabling the complement system in Daf1(-/-) mice normalized T cell secretion of IFN-gamma and IL-2 and attenuated disease severity in the EAE model. These findings establish a critical link between complement and T cell immunity and have implications for the role of DAF and complement in organ transplantation, tumor evasion, and vaccine development.


Asunto(s)
Antígenos CD55/inmunología , Proteínas del Sistema Complemento/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD55/genética , Antígenos CD55/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Inmunización , Interferón gamma/biosíntesis , Interleucina-10/metabolismo , Interleucina-2/biosíntesis , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/citología , Bazo/inmunología
18.
Mol Pharm ; 8(5): 1980-4, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21732666

RESUMEN

Intrathecal delivery of gene therapeutics is a route of administration that overcomes several of the limitations that plague current immunosuppressive treatments for autoimmune diseases of the central nervous system (CNS). Here we report intrathecal delivery of small amounts (3 µg) of plasmid DNA that codes for an immunomodulatory fusion protein, OX40-TRAIL, composed of OX40, a tumor necrosis factor receptor, and tumor necrosis factor related apoptosis inducing ligand (TRAIL). This DNA was delivered in a formulated nucleic acid-lipid complex (lipoplexes) with an asymmetric two-chain cationic lipid myristoyl (14:0) and lauroyl (12:1) rosenthal inhibitor-substituted compound (MLRI) formed from the tetraalkylammonium glycerol-based compound N-(1-(2,3-dioleoyloxy)-propyl-N-1-(2-hydroxy)ethyl)-N,N-dimethyl ammonium iodide. Delivery and expression in the CNS of OX40-TRAIL in the mouse prior to onset of experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, decreased the severity of clinical disease. We believe this preclinical demonstration of rapid, widespread, and biologically therapeutic nonviral gene delivery to the CNS is important in further development of clinical lipid-based therapeutics for CNS disorders.


Asunto(s)
ADN/química , Encefalomielitis Autoinmune Experimental/terapia , Técnicas de Transferencia de Gen , Terapia Genética , Lípidos/química , Animales , Sistema Nervioso Central/metabolismo , Cisterna Magna , ADN/administración & dosificación , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Expresión Génica , Genes Reporteros , Inyecciones Espinales , Lauratos/química , Lípidos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/terapia , Miristatos/química , Plásmidos/administración & dosificación , Plásmidos/química , Receptores OX40/genética , Receptores OX40/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Estearatos/administración & dosificación , Estearatos/química , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
19.
Cell Immunol ; 261(2): 144-52, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20042183

RESUMEN

Previously we have shown that DAB(389)IL-2, a recombinant fusion toxin targeting IL-2R bearing cells, suppressed disease in the rat experimental autoimmune encephalomyelitis (EAE) model of acute multiple sclerosis (MS). Our present study demonstrates that DAB(389)IL-2 can also effectively suppress acute (A)-EAE, relapsing (R)-EAE and chronic (C)-EAE in mouse demyelinating models. DAB(389)IL-2 significantly suppressed mitogenic proliferation of spleen cells while mutant fusion proteins DA(glu53)B(389)IL-2 and DAB(389)IL-2(8-10) did not. EAE was successfully suppressed when DAB(389)IL-2 was administered in various regimens between days 1 and 15 post immunization in all three models. CD4(+)IL-2R(+) cells were reduced in the spleen but not in the lymph nodes of DAB(389)IL-2-treated mice during A-EAE while the number of CD8(+) cells was unchanged. DAB(389)IL-2 also significantly reduced the number of CD4(+), CD8(+), CD25(+), TCRgammadelta(+) phenotype and CD11b(+) macrophages/microglia within spinal cord lesions. These data strongly suggest that DAB(389)IL-2 specifically targeted myelin protein-activated CD4(+) T cells and strengthens the argument for the use of DAB(389)IL-2 in treatment strategies for MS.


Asunto(s)
Toxina Diftérica/inmunología , Toxina Diftérica/uso terapéutico , Encefalomielitis Autoinmune Experimental , Interleucina-2/inmunología , Interleucina-2/uso terapéutico , Receptores de Interleucina-2 , Animales , Proliferación Celular , Células Cultivadas , Toxina Diftérica/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Humanos , Interleucina-2/genética , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Ratas , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Médula Espinal/citología , Médula Espinal/inmunología , Médula Espinal/patología , Linfocitos T/inmunología
20.
Am J Pathol ; 174(2): 460-74, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19147815

RESUMEN

Hallmarks of the pathogenesis of autoimmune encephalomyelitis include perivascular infiltration of inflammatory cells into the central nervous system, multifocal demyelination in the brain and spinal cord, and focal neuronal degeneration. Optimal treatment of this complex disease will ultimately call for agents that target the spectrum of underlying pathogenic processes. In the present study, Fn14-TRAIL is introduced as a unique immunotherapeutic fusion protein that is designed to exchange and redirect intercellular signals within inflammatory cell networks, and, in so doing, to impact multiple pathogenic events and yield a net anti-inflammatory effect. In this soluble protein product, a Fn14 receptor component (capable of blocking the pro-inflammatory TWEAK ligand) is fused to a TRAIL ligand (capable of inhibiting activated, pathogenic T cells). Sustained Fn14-TRAIL expression was obtained in vivo using a transposon-based eukaryotic expression vector. Fn14-TRAIL expression effectively prevented chronic, nonremitting, paralytic disease in myelin oligodendrocyte glycoprotein-challenged C57BL/6 mice. Disease suppression in this model was reflected by decreases in the clinical score, disease incidence, nervous tissue inflammation, and Th1, Th2, and Th17 cytokine responses. Significantly, the therapeutic efficacy of Fn14-TRAIL could not be recapitulated simply by administering its component parts (Fn14 and TRAIL) as soluble agents, either alone or in combination. Its functional pleiotropism was manifest in its additional ability to attenuate the enhanced permeability of the blood-brain barrier that typically accompanies autoimmune encephalomyelitis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Células CHO , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cricetinae , Cricetulus , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Factores Inmunológicos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores del Factor de Necrosis Tumoral/inmunología , Proteínas Recombinantes de Fusión/inmunología , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Receptor de TWEAK
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA