Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.887
Filtrar
1.
Cells ; 13(19)2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39404366

RESUMEN

OBJECTIVE: Monocytes are innate immune cells that play a central role in inflammation, an essential component during neovascularization. Our recent publication demonstrated that ischemia training by 24 h unilateral occlusion of the femoral artery (FA) can modify bone marrow-derived monocytes (BM-Mono), allowing them to improve collateral remodeling in a mouse model of hindlimb ischemia. Here, we expand on our previous findings, investigating a potential systemic effect of ischemia training and how this training can impact BM-Mono. METHODS AND RESULTS: BM-Mono from mice exposed to ischemia training (24 h) or Sham (same surgical procedure without femoral artery occlusion-ischemia training) procedures were used as donors in adoptive transfer experiments where recipients were subjected to hindlimb ischemia. Donor cells were divided corresponding to the limb from which they were isolated (left-limb previously subjected to 24 h ischemia and right-contralateral limb). Recipients who received 24 h ischemic-trained monocytes isolated from either limb had remarkable blood flow recovery compared to recipients with Sham monocytes (monocytes isolated from Sham group-no ischemia training). Since these data suggested a systemic effect of ischemic training, circulating extracellular vesicles (EVs) were investigated as potential players. EVs were isolated from both groups, 24 h-trained and Sham, and the former showed increased expression of histone deacetylase 1 (HDAC1), which is known to downregulate 24-dehydrocholesterol reductase (Dhcr24) gene expression. Since we previously revealed that ischemia training downregulates Dhcr24 in BM-Mono, we incubated EVs from 24 h-trained and Sham groups with wild-type (WT) BM-Mono and demonstrated that WT BM-Mono incubated with 24 h-trained EVs had lower gene expression of Dhcr24 and an HDAC1 inhibitor blunted this effect. Next, we repeated the adoptive transfer experiment using Dhcr24 KO mice as donors of BM-Mono for WT mice subjected to hindlimb ischemia. Recipients who received Dhcr24 KO BM-Mono had greater limb perfusion than those who received WT BM-Mono. Further, we focused on the 24 h-trained monocytes (which previously showed downregulation of Dhcr24 gene expression and higher desmosterol) to test the expression of a few genes downstream of the desmosterol pathway, confirm the Dhcr24 protein level and assess its differentiation in M2-like macrophage phenotype. We found that 24 h-trained BM-Mono had greater expression of key genes in the desmosterol pathway, such as liver X receptors (LXRs) and ATP-binding cassette transporter (ABCA1), and we confirmed low protein expression of Dhcr24. Further, we demonstrated that ischemic-trained BM-Mono polarized towards an anti-inflammatory M2 macrophage phenotype. Finally, we demonstrated that 24 h-trained monocytes adhere less to endothelial cells, and the same pattern was shown by WT BM-Mono treated with Dhcr24 inhibitor. CONCLUSIONS: Ischemia training leads to a systemic effect that, at least in part, involves circulating EVs and potential epigenetic modification in BM-Mono. These ischemic-trained BM-Mono demonstrated an anti-inflammatory phenotype towards M2 macrophage differentiation and less ability to adhere to endothelial cells, which is associated with the downregulation of Dhcr24 in those cells. These data together suggest that Dhcr24 might be an important target within monocytes to improve the outcomes of hindlimb ischemia.


Asunto(s)
Isquemia , Monocitos , Animales , Monocitos/metabolismo , Isquemia/metabolismo , Isquemia/patología , Ratones , Ratones Endogámicos C57BL , Masculino , Miembro Posterior/irrigación sanguínea , Células de la Médula Ósea/metabolismo , Condicionamiento Físico Animal
2.
PeerJ ; 12: e18189, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39399423

RESUMEN

Objective: To investigate the mid-term efficacy and patency rate of TurboHawk peripheral plaque excision system in the treatment of femoral popliteal artery lesions with different Global Limb Anatomic Staging System (GLASS) grades. Methods: The clinical data of 141 patients with femoral popliteal arteriosclerosis obliterans who were treated with TurboHawk from January 2018 to July 2022 in our institution were retrospectively analyzed. There were 109 male patients and 32 female patients. Recordings were made of the patient's symptoms of limb ischemia, technical success rate, primary patency rate of target vessels, ankle brachial index (ABI), GLASS grades, postoperative complications, and a statistical analysis with the patient's preoperative treatment was conducted. Results: All patients had improved limb ischemia symptoms to varying degrees after surgery, with a technical success rate of 100% (femoral artery puncture and superficial femoral artery recanalization) without bleeding, hematoma, pseudoaneurysm, arteriovenous fistula or other complications. The follow-up period was 1-24 months, during which the severity of claudication, resting pain, and toe ulcers significantly improved. The primary patency rate of the target vessel was 98.58% (139/141), and the ABI significantly increased on the second day, three months, and six months after surgery compared to before surgery. No major adverse events were found during follow-up. The patency rates at 1, 6, 12 and 24 months after intervention were 100%, 80%, 75% and 60% respectively. Conclusion: The mid-term efficacy and patency rate of TurboHawk in the treatment of femoral popliteal artery lesions with GLASS I patients have the best mid-term prognosis, the highest mid-term survival rate, and the highest vascular patency. The plaque removal system has proven to be an effective treatment for individual localized chronic total occlusion lesions. Additionally, the TurboHawk system provides a safe and minimally invasive treatment alternative for superficial femoral artery conditions, achieving significant therapeutic results within a brief period.


Asunto(s)
Aterectomía , Arteria Femoral , Arteria Poplítea , Humanos , Masculino , Femenino , Arteria Poplítea/cirugía , Arteria Poplítea/patología , Arteria Femoral/cirugía , Arteria Femoral/patología , Anciano , Estudios Retrospectivos , Persona de Mediana Edad , Resultado del Tratamiento , Aterectomía/métodos , Grado de Desobstrucción Vascular , Arteriosclerosis Obliterante/cirugía , Arteriosclerosis Obliterante/patología , Índice Tobillo Braquial , Isquemia/cirugía , Isquemia/patología , Complicaciones Posoperatorias , Anciano de 80 o más Años
3.
Sci Adv ; 10(40): eadn8760, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39365855

RESUMEN

Reversal of ischemia is mediated by neo-angiogenesis requiring endothelial cell (EC) and pericyte interactions to form stable microvascular networks. We describe an unrecognized role for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in potentiating neo-angiogenesis and vessel stabilization. We show that the endothelium is a major source of TRAIL in the healthy circulation compromised in peripheral artery disease (PAD). EC deletion of TRAIL in vivo or in vitro inhibited neo-angiogenesis, pericyte recruitment, and vessel stabilization, resulting in reduced lower-limb blood perfusion with ischemia. Activation of the TRAIL receptor (TRAIL-R) restored blood perfusion and stable blood vessel networks in mice. Proof-of-concept studies showed that Conatumumab, an agonistic TRAIL-R2 antibody, promoted vascular sprouts from explanted patient arteries. Single-cell RNA sequencing revealed heparin-binding EGF-like growth factor in mediating EC-pericyte communications dependent on TRAIL. These studies highlight unique TRAIL-dependent mechanisms mediating neo-angiogenesis and vessel stabilization and the potential of repurposing TRAIL-R2 agonists to stimulate stable and functional microvessel networks to treat ischemia in PAD.


Asunto(s)
Células Endoteliales , Isquemia , Microvasos , Ligando Inductor de Apoptosis Relacionado con TNF , Animales , Humanos , Masculino , Ratones , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Isquemia/metabolismo , Isquemia/patología , Microvasos/metabolismo , Microvasos/patología , Neovascularización Fisiológica , Pericitos/metabolismo , Pericitos/patología , Enfermedad Arterial Periférica/metabolismo , Enfermedad Arterial Periférica/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Adulto , Femenino
4.
Int J Mol Sci ; 25(19)2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39408774

RESUMEN

Oxidative stress triggered by testicular torsion and detorsion in young males could negatively impact future fertility. Using a rat animal model for testicular IRI (tIRI), we aim to study the induction of autophagy (ATG) during testicular ischemia and tIRI and the role of oxidative-stress-induced c-Jun N-terminal Kinase (JNK) as a cytoprotective mechanism. Sixty male Sprague-Dawley rats were divided into five groups: sham, ischemia only, ischemia+SP600125 (a JNK inhibitor), tIRI only, and tIRI+SP600125. The tIRI rats underwent an ischemic injury for 1 h followed by 4 h of reperfusion, while ischemic rats were subjected to 1 h of ischemia only without reperfusion. Testicular-ischemia-induced Beclin 1 and LC3B expression was associated with decreased p62/SQSTM1 expression, increased ATP and alkaline phosphatase (AP) activity, and slightly impaired spermatogenesis. SP600125 treatment improved p62 expression and reduced the levels of Beclin 1 and LC3B but did not affect ATP or AP levels. The tIRI-induced apoptosis lowered the expression of the three ATG proteins and AP activity, activated caspase 3, and caused spermatogenic arrest. SP600125-inhibited JNK during tIRI restored sham levels to all investigated parameters. This study emphasizes the regulatory role of JNK in balancing autophagy and apoptosis during testicular oxidative injuries.


Asunto(s)
Apoptosis , Autofagia , Proteínas Quinasas JNK Activadas por Mitógenos , Ratas Sprague-Dawley , Daño por Reperfusión , Testículo , Masculino , Animales , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratas , Testículo/metabolismo , Testículo/patología , Estrés Oxidativo , Antracenos/farmacología , Isquemia/metabolismo , Isquemia/patología , Torsión del Cordón Espermático/metabolismo , Torsión del Cordón Espermático/patología , Espermatogénesis/efectos de los fármacos , Beclina-1/metabolismo , Modelos Animales de Enfermedad
5.
Biomacromolecules ; 25(10): 6656-6665, 2024 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-39264109

RESUMEN

Critical limb ischemia (CLI) is a peripheral arterial disease resulting from chronic inflammation of vascular systems. Recent studies have shown that inhibiting macrophage inflammation has the potential to treat CLI, and extracellular vesicles (EVs) from endothelial cells can inhibit macrophage activation. However, the limited cell-targeting capabilities and rapid clearance of EVs from the injection site limit the in vivo application of the EVs. Here, we modified endothelial EVs with platelet membranes (pM/EVs) to boost the inhibition effects on macrophage inflammation and developed an injectable alginate-based collagen composite (ACC) hydrogel for localized delivery of pM/EVs (pM/EVs@ACC) for CLI treatment. We found that pM/EVs can effectively inhibit macrophage inflammation in vitro. Furthermore, pM/EVs@ACC treatment significantly promotes the recovery of limb functions, restoring the feet' blood supply and relieving inflammation. Our findings provide compelling evidence that the pM/EVs@ACC injectable system mediating delivery of pM/EVs is a promising strategy for CLI treatment.


Asunto(s)
Alginatos , Vesículas Extracelulares , Hidrogeles , Isquemia , Animales , Alginatos/química , Hidrogeles/química , Hidrogeles/farmacología , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Isquemia/terapia , Isquemia/patología , Ratones , Humanos , Células Endoteliales de la Vena Umbilical Humana , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Masculino , Inflamación , Células RAW 264.7 , Miembro Posterior/irrigación sanguínea , Ratones Endogámicos C57BL
6.
Int J Med Sci ; 21(12): 2402-2413, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39310266

RESUMEN

Lower limb ischemia is characterized by reduced arterial perfusion in the lower limbs, leading to tissue ischemia and cell death. It is primarily caused by thrombosis and the rupture of arterial plaques, resulting in damage to ischemic muscle tissues. Metabolic processes are crucial in its development. Herein we combined single-cell data with metabolomics data to explore the pathways and mechanisms influencing lower limb ischemia. We analyzed single-cell and metabolomics data. In single-cell analysis, we identified different cell subpopulations and key regulatory genes, and biological enrichment analysis was performed to understand their functions and relationships. For metabolomics, mass spectrometry and chromatography techniques were employed to analyze metabolites in clinical samples. We performed differential analysis, correlation analysis, and Mendelian randomization to determine the relationships between key metabolites and genes. Nebl, Dapl1, Igfbp4, Lef1, Klrd1, Ciita, Il17f, Cd8b1, Il17a, Cd180, Il17re, Trim7, and Slc6a19 were identified to play a crucial role in lower limb ischemia. Important metabolites included L-threonine and L-tryptophan. The metabolism of L-threonine and L-tryptophan is linked to lower limb ischemia and thrombosis. B0AT1, encoded by SLC6A19, is closely related to these metabolites and appears to play a key role in lower limb ischemia development. Our analysis revealed the roles of key genes and metabolites in lower limb ischemia. These findings enhance our understanding of the pathogenesis of lower limb ischemia and provide new insights into its prevention and treatment.


Asunto(s)
Isquemia , Extremidad Inferior , Triptófano , Humanos , Triptófano/metabolismo , Isquemia/metabolismo , Isquemia/patología , Extremidad Inferior/irrigación sanguínea , Metabolómica/métodos , Masculino
7.
N Z Vet J ; 72(6): 347-354, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39186936

RESUMEN

CASE HISTORY: In spring 2021, on a seasonally calving, pastorally based, Taranaki dairy farm, 12 first-calving heifers (≤ 30 days post-calving) developed similar dry, red to black, crusting lesions on the medial aspect of the teat udder junction extending down the medial teat. Some cows had multiple teats affected. Treatment was initially unrewarding and did not slow the progression of the disease. Overall, 8/12 cows recovered, and 4/12 cows were culled, with three of the cows culled after a teat sloughed and the fourth after surgical amputation of a teat. Outbreaks of the same condition, on the same farm but affecting fewer animals, occurred in spring 2022 (n = 6) and spring 2023 (n = 3). CLINICAL FINDINGS: An initial scab-like or crusting lesion progressed to resemble a thick eschar consisting of very dry and hard dead tissue. The unaffected areas of the teat felt normal but immediately under the dead tissue, there was a warm, firmer area consistent with an inflammatory reaction. Removing the scab led to profuse bleeding, with no visible bed of granulation underneath the scab. There was no leaking of milk in those cows that lost a teat, and no smell to the lesions themselves. Serology and virology ruled out the involvement of bovine alphaherpesvirus (BoHV-2) bovine gammaherpesvirus (BoHV-4), orthopoxviruses (cowpox) and parapoxviruses (pseudocowpox). Histopathology of an affected and surgically amputated teat showed multifocal erosion and ulceration of the epidermis, covered by a thick serocellular crust. In areas of ulceration, there were numerous neutrophils, and the dermis was expanded by granulation tissue with variable numbers of neutrophils, eosinophils, and lymphocytes around small blood vessels. DIAGNOSIS: Based on the similarity of the history, presentation, and histopathological changes to those described for a novel disease reported in the UK, a diagnosis of ischaemic teat necrosis (ITN) was made. CLINICAL RELEVANCE: If ITN is an emerging condition in New Zealand and becomes as prevalent as it has in the UK, clinicians will be confronted with a significant new welfare problem in dairy cows. Anecdotally, there have been reports of other ITN outbreaks in New Zealand, and the Ministry for Primary Industries would be interested in collating reports from other New Zealand veterinarians.


Asunto(s)
Industria Lechera , Brotes de Enfermedades , Glándulas Mamarias Animales , Necrosis , Animales , Bovinos , Femenino , Nueva Zelanda/epidemiología , Glándulas Mamarias Animales/patología , Brotes de Enfermedades/veterinaria , Necrosis/veterinaria , Mastitis Bovina/epidemiología , Mastitis Bovina/patología , Isquemia/veterinaria , Isquemia/patología , Isquemia/epidemiología , Enfermedades de los Bovinos/patología , Enfermedades de los Bovinos/epidemiología
8.
Sci Adv ; 10(32): eadp6182, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39121218

RESUMEN

Endothelial cells (ECs) are highly plastic, capable of differentiating into various cell types. Endothelial-to-mesenchymal transition (EndMT) is crucial during embryonic development and contributes substantially to vascular dysfunction in many cardiovascular diseases (CVDs). While targeting EndMT holds therapeutic promise, understanding its mechanisms and modulating its pathways remain challenging. Using single-cell RNA sequencing on three in vitro EndMT models, we identified conserved gene signatures. We validated original regulators in vitro and in vivo during embryonic heart development and peripheral artery disease. EndMT induction led to global expression changes in all EC subtypes rather than in mesenchymal clusters. We identified mitochondrial calcium uptake as a key driver of EndMT; inhibiting mitochondrial calcium uniporter (MCU) prevented EndMT in vitro, and conditional Mcu deletion in ECs blocked mesenchymal activation in a hind limb ischemia model. Tissues from patients with critical limb ischemia with EndMT features exhibited significantly elevated endothelial MCU. These findings highlight MCU as a regulator of EndMT and a potential therapeutic target.


Asunto(s)
Señalización del Calcio , Células Endoteliales , Transición Epitelial-Mesenquimal , Mitocondrias , RNA-Seq , Análisis de la Célula Individual , Animales , Humanos , Mitocondrias/metabolismo , RNA-Seq/métodos , Ratones , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Canales de Calcio/metabolismo , Canales de Calcio/genética , Isquemia/metabolismo , Isquemia/patología , Calcio/metabolismo , Análisis de Expresión Génica de una Sola Célula
9.
Cells ; 13(15)2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39120274

RESUMEN

MicroRNAs (miRs) regulate physiological and pathological processes, including ischemia-induced angiogenesis and neovascularization. They can be transferred between cells by extracellular vesicles (EVs). However, the specific miRs that are packaged in EVs released from skeletal muscles, and how this process is modulated by ischemia, remain to be determined. We used a mouse model of hindlimb ischemia and next generation sequencing (NGS) to perform a complete profiling of miR expression and determine the effect of ischemia in skeletal muscles, and in EVs of different sizes (microvesicles (MVs) and exosomes) released from these muscles. Ischemia significantly modulated miR expression in whole muscles and EVs, increasing the levels of several miRs that can have pro-angiogenic effects (angiomiRs). We found that specific angiomiRs are selectively enriched in MVs and/or exosomes in response to ischemia. In silico approaches indicate that these miRs modulate pathways that play key roles in angiogenesis and neovascularization, including HIF1/VEGF signaling, regulation of actin cytoskeleton and focal adhesion, NOTCH, PI3K/AKT, RAS/MAPK, JAK/STAT, TGFb/SMAD signaling and the NO/cGMP/PKG pathway. Thus, we show for the first time that angiomiRs are selectively enriched in MVs and exosomes released from ischemic muscles. These angiomiRs could be targeted in order to improve the angiogenic function of EVs for potential novel therapeutic applications in patients with severe ischemic vascular diseases.


Asunto(s)
Vesículas Extracelulares , Isquemia , MicroARNs , Músculo Esquelético , Neovascularización Fisiológica , Animales , Músculo Esquelético/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Vesículas Extracelulares/metabolismo , MicroARNs/metabolismo , MicroARNs/genética , Isquemia/metabolismo , Isquemia/patología , Ratones , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Ratones Endogámicos C57BL , Transducción de Señal , Masculino , Exosomas/metabolismo , Neovascularización Patológica/metabolismo , Angiogénesis
10.
Int J Mol Sci ; 25(15)2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39125579

RESUMEN

The retina is one of the highest metabolically active tissues with a high oxygen consumption, so insufficient blood supply leads to visual impairment. The incidence of related conditions is increasing; however, no effective treatment without side effects is available. Furthermore, the pathomechanism of these diseases is not fully understood. Our aim was to develop an optimal ischemic retinopathy mouse model to investigate the retinal damage in a time-dependent manner. Retinal ischemia was induced by bilateral common carotid artery occlusion (BCCAO) for 10, 13, 15 or 20 min, or by right permanent unilateral common carotid artery occlusion (UCCAO). Optical coherence tomography was used to follow the changes in retinal thickness 3, 7, 14, 21 and 28 days after surgery. The number of ganglion cells was evaluated in the central and peripheral regions on whole-mount retina preparations. Expression of glial fibrillary acidic protein (GFAP) was analyzed with immunohistochemistry and Western blot. Retinal degeneration and ganglion cell loss was observed in multiple groups. Our results suggest that the 20 min BCCAO is a good model to investigate the consequences of ischemia and reperfusion in the retina in a time-dependent manner, while the UCCAO causes more severe damage in a short time, so it can be used for testing new drugs.


Asunto(s)
Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía , Hipoxia , Isquemia , Retina , Tomografía de Coherencia Óptica , Animales , Ratones , Isquemia/metabolismo , Isquemia/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Retina/metabolismo , Retina/patología , Hipoxia/metabolismo , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Enfermedades de la Retina/etiología , Masculino , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Ratones Endogámicos C57BL , Factores de Tiempo
11.
Mol Brain ; 17(1): 50, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095918

RESUMEN

Neuroactive steroids (NASs) directly affect neuronal excitability. Despite their role in the nervous system is intimately linked to pain control, knowledge is currently limited. This study investigates the peripheral involvement of NASs in chronic ischemic pain by targeting the cytochrome P450 side-chain cleavage enzyme (P450scc). Using a rat model of hind limb thrombus-induced ischemic pain (TIIP), we observed an increase in P450scc expression in the ischemic hind paw skin. Inhibiting P450scc with intraplantar aminoglutethimide (AMG) administration from post-operative day 0 to 3 significantly reduced the development of mechanical allodynia. However, AMG administration from post-operative day 3 to 6 did not affect established mechanical allodynia. In addition, we explored the role of the peripheral sigma-1 receptor (Sig-1R) by co-administering PRE-084 (PRE), a Sig-1R agonist, with AMG. PRE reversed the analgesic effects of AMG during the induction phase. These findings indicate that inhibiting steroidogenesis with AMG alleviates peripheral ischemic pain during the induction phase via Sig-1Rs.


Asunto(s)
Modelos Animales de Enfermedad , Hiperalgesia , Isquemia , Ratas Sprague-Dawley , Receptores sigma , Animales , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/patología , Hiperalgesia/complicaciones , Masculino , Isquemia/complicaciones , Isquemia/patología , Receptores sigma/antagonistas & inhibidores , Receptores sigma/metabolismo , Receptor Sigma-1 , Dolor/tratamiento farmacológico , Dolor/complicaciones , Dolor/etiología , Dolor/patología , Miembro Posterior/efectos de los fármacos , Ratas , Sistema Enzimático del Citocromo P-450/metabolismo
12.
Nat Cardiovasc Res ; 3(6): 685-700, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39196227

RESUMEN

Sterile inflammation after injury is important for tissue restoration. In injured human and mouse tissues, macrophages were recently found to accumulate perivascularly. This study investigates if macrophages adopt a mural cell phenotype important for restoration after ischemic injury. Single-cell RNA sequencing of fate-mapped macrophages from ischemic mouse muscles demonstrates a macrophage-toward-mural cell switch of a subpopulation of macrophages with downregulated myeloid cell genes and upregulated mural cell genes, including PDGFRß. This observation was further strengthened when including unspliced transcripts in the analysis. The macrophage switch was proven functionally relevant, as induction of macrophage-specific PDGFRß deficiency prevented their perivascular macrophage phenotype, impaired vessel maturation and increased vessel leakiness, which ultimately reduced limb function. In conclusion, macrophages in adult ischemic tissue were demonstrated to undergo a cellular program to morphologically, transcriptomically and functionally resemble mural cells while weakening their macrophage identity. The macrophage-to-mural cell-like phenotypic switch is crucial for restoring tissue function and warrants further exploration as a potential target for immunotherapies to enhance healing.


Asunto(s)
Modelos Animales de Enfermedad , Isquemia , Macrófagos , Animales , Macrófagos/metabolismo , Macrófagos/inmunología , Isquemia/metabolismo , Isquemia/patología , Isquemia/genética , Fenotipo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/lesiones , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología , Ratones Endogámicos C57BL , Ratones , Masculino , Miembro Posterior/irrigación sanguínea , Neovascularización Fisiológica/genética , Regulación hacia Arriba , Transcriptoma , Análisis de la Célula Individual , Biomarcadores/metabolismo , Recuperación de la Función , Ratones Noqueados
13.
Stem Cell Res Ther ; 15(1): 225, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39075518

RESUMEN

BACKGROUND: This study explores the potential role of Thioredoxin-interacting protein (TXNIP) silencing in endothelial colony-forming cells (ECFCs) within the scope of age-related comorbidities and impaired vascular repair. We aim to elucidate the effects of TXNIP silencing on vasculogenic properties, paracrine secretion, and neutrophil recruitment under conditions of metabolic stress. METHODS: ECFCs, isolated from human blood cord, were transfected with TXNIP siRNA and exposed to a high glucose and ß-hydroxybutyrate (BHB) medium to simulate metabolic stress. We evaluated the effects of TXNIP silencing on ECFCs' functional and secretory responses under these conditions. Assessments included analyses of gene and protein expression profiles, vasculogenic properties, cytokine secretion and neutrophil recruitment both in vitro and in vivo. The in vivo effects were examined using a murine model of hindlimb ischemia to observe the physiological relevance of TXNIP modulation under metabolic disorders. RESULTS: TXNIP silencing did not mitigate the adverse effects on cell recruitment, vasculogenic properties, or senescence induced by metabolic stress in ECFCs. However, it significantly reduced IL-8 secretion and consequent neutrophil recruitment under these conditions. In a mouse model of hindlimb ischemia, endothelial deletion of TXNIP reduced MIP-2 secretion and prevented increased neutrophil recruitment induced by age-related comorbidities. CONCLUSIONS: Our findings suggest that targeting TXNIP in ECFCs may alleviate ischemic complications exacerbated by metabolic stress, offering potential clinical benefits for patients suffering from age-related comorbidities.


Asunto(s)
Proteínas Portadoras , Interleucina-8 , Infiltración Neutrófila , Estrés Fisiológico , Animales , Interleucina-8/metabolismo , Interleucina-8/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Humanos , Ratones , Infiltración Neutrófila/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/efectos de los fármacos , Isquemia/metabolismo , Isquemia/patología , ARN Interferente Pequeño/metabolismo , Tiorredoxinas/metabolismo , Tiorredoxinas/genética , Miembro Posterior/irrigación sanguínea , Ratones Endogámicos C57BL , Glucosa/metabolismo
14.
Acta Biomater ; 185: 410-428, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39029641

RESUMEN

Ischemic osteonecrosis, particularly glucocorticoid-induced osteonecrosis of the femoral head (GIONFH), is primarily due to the dysfunction of osteogenesis and angiogenesis. miRNA, as a therapeutic system with immense potential, plays a vital role in the treatment of various diseases. However, due to the unique microenvironmental structure of bone tissue, especially in the case of GIONFH, where there is a deficiency in the vascular system, it is challenging to effectively target and deliver to the ischemic osteonecrosis area. A drug delivery system assisted by genetically engineered cell membranes holds promise in addressing the challenge of targeted miRNA delivery. Herein, we leverage the potential of miR-21 in modulating osteogenesis and angiogenesis to design an innovative biomimetic nanoplatform system. First, we employed metal-organic frameworks (MOFs) as the core structure to load miR-21-m (miR-21-m@MOF). The nanoparticles were further coated with the membrane of bone marrow mesenchymal stem cells overexpressing CXCR4 (CM-miR-21-m@MOF), enhancing their ability to target ischemic bone areas via the CXCR4-SDF1 axis. These biomimetic nanocomposites possess both bone-targeting and ischemia-guiding capabilities, actively targeting GIONFH lesions to release miR-21-m into target cells, thereby silencing PTEN gene and activating the PI3K-AKT signaling pathway to regulate osteogenesis and angiogenesis. This innovative miRNA delivery system provides a promising therapeutic avenue for GIONFH and potentially other related ischemic bone diseases. STATEMENT OF SIGNIFICANCE.


Asunto(s)
Membrana Celular , Estructuras Metalorgánicas , MicroARNs , Nanopartículas , Osteonecrosis , Estructuras Metalorgánicas/química , Animales , Osteonecrosis/patología , Osteonecrosis/genética , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Membrana Celular/metabolismo , Nanopartículas/química , Osteogénesis/efectos de los fármacos , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Células Madre Mesenquimatosas/metabolismo , Ingeniería Genética , Isquemia/patología , Isquemia/terapia , Isquemia/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Conejos
15.
Kidney Int ; 106(4): 640-657, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39074555

RESUMEN

Long non-coding RNAs (lncRNAs) are a group of epigenetic regulators that have been implicated in kidney diseases including acute kidney injury (AKI). However, very little is known about the specific lncRNAs involved in AKI and the mechanisms underlying their pathologic roles. Here, we report a new lncRNA derived from the pseudogene GSTM3P1, which mediates ischemic AKI by interacting with and promoting the degradation of mir-668, a kidney-protective microRNA. GSTM3P1 and its mouse orthologue Gstm2-ps1 were induced by hypoxia in cultured kidney proximal tubular cells. In mouse kidneys, Gstm2-ps1 was significantly upregulated in proximal tubules at an early stage of ischemic AKI. This transient induction of Gstm2-ps1 depends on G3BP1, a key component in stress granules. GSTM3P1 overexpression increased kidney proximal tubular apoptosis after ATP depletion, which was rescued by mir-668. Notably, kidney proximal tubule-specific knockout of Gstm2-ps1 protected mice from ischemic AKI, as evidenced by improved kidney function, diminished tubular damage and apoptosis, and reduced kidney injury biomarker (NGAL) induction. To test the therapeutic potential, Gstm2-ps1 siRNAs were introduced into cultured mouse proximal tubular cells or administered to mice. In cultured cells, Gstm2-ps1 knockdown suppressed ATP depletion-associated apoptosis. In mice, Gstm2-ps1 knockdown ameliorated ischemic AKI. Mechanistically, both GSTM3P1 and Gstm2-ps1 possessed mir-668 binding sites and downregulated the mature form of mir-668. Specifically, GSTM3P1 directly bound to mature mir-668 to induce its decay via target-directed microRNA degradation. Thus, our results identify GSTM3P1 as a novel lncRNA that promotes kidney tubular cell death in AKI by binding mir-668 to inducing its degradation.


Asunto(s)
Lesión Renal Aguda , Apoptosis , Túbulos Renales Proximales , MicroARNs , Seudogenes , ARN Largo no Codificante , Animales , Humanos , Masculino , Ratones , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/etiología , Apoptosis/genética , Modelos Animales de Enfermedad , Isquemia/genética , Isquemia/metabolismo , Isquemia/patología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/metabolismo , MicroARNs/genética , Seudogenes/genética , Estabilidad del ARN , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
16.
Nanomedicine (Lond) ; 19(17): 1525-1539, 2024 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-39012207

RESUMEN

Aim: This study aims to investigate the effects of large extracellular vesicles (EVs) induced by pluripotent stem cell-derived mesenchymal stem cells on lower limb ischemic disease and explore its potential mechanisms. Materials & methods: The pathology of muscles was accessed by H&E staining and immunofluorescence staining. In vitro, we conducted wound-healing assay, tube formation assay, RT qPCR, ELISA, RNA sequencing and proteomic analysis. Results: iMSCs-lEVs alleviated the injury of ischemic lower limb and promoted the recovery of lower limb function. In vitro, iMSCs-lEVs promoted the proliferation, migration, and angiogenesis of HMEC-1 cells by regulating the ERK/MAPK signing pathway. Conclusion: This study demonstrated that iMSCs-lEVs promoted endothelial cell angiogenesis via the ERK/MAPK signaling pathway, thereby improving function after lower limb ischemic injury.


[Box: see text].


Asunto(s)
Vesículas Extracelulares , Células Madre Pluripotentes Inducidas , Isquemia , Sistema de Señalización de MAP Quinasas , Neovascularización Fisiológica , Vesículas Extracelulares/metabolismo , Animales , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Isquemia/terapia , Isquemia/metabolismo , Isquemia/patología , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Proliferación Celular , Extremidad Inferior/irrigación sanguínea , Movimiento Celular , Masculino , Angiogénesis
17.
Biomolecules ; 14(7)2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-39062551

RESUMEN

Acute limb ischemia (ALI) is defined as a sudden reduction in blood flow to a limb, resulting in cessation of blood flow and, therefore, cessation of the delivery of nutrients and oxygen to the tissues of the lower limb. Despite optimal treatment to restore blood flow to ischemic tissues, some patients may suffer from ischemia/reperfusion (I/R) syndrome, the most severe complication after a revascularization procedure used to restore blood flow. There are multiple molecular and cellular factors that are involved in each phase of ALI. This review focuses firstly on molecular and cellular factors of arterial thrombosis, highlighting the role of atherosclerotic plaques, smooth muscle cells (SMCs), and cytokine which may alter key components of the extracellular matrix (ECM). Then, molecular and cellular factors of arterial embolism will be discussed, highlighting the importance of thrombi composition. Molecular and cellular factors of ischemia/reperfusion syndrome are analyzed in depth, highlighting several important mechanisms related to tissue damage, such as inflammation, apoptosis, autophagy, necrosis, and necroptosis. Furthermore, local and general complications of ALI are discussed in the context of molecular alterations. Ultimately, the role of novel biomarkers and targeted therapies is discussed.


Asunto(s)
Isquemia , Humanos , Isquemia/metabolismo , Isquemia/patología , Animales , Trombosis/metabolismo , Trombosis/patología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Enfermedad Aguda , Extremidades/irrigación sanguínea , Extremidades/patología
18.
Pathol Int ; 74(8): 475-481, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38994749

RESUMEN

"Cysts of the ligamentum flavum (cysts-LF)" is the term for non-neoplastic cystic lesion involving LF. The aim of the present study was to elucidate the histopathological characteristics and pathogenesis of "cysts-LF". Herein, we defined cysts-LF as spinal cysts containing degenerative LF components. From archival cases, we investigated 18 symptomatic cysts-LF surgically removed from 18 patients (13 males and five females; median age 68.5 years [range, 42-86 years]). The elastic fibers of LF components in the wall were separated and/or torn, and cyst walls were accompanied by chondroid metaplasia (17 cases), myxoid changes (13 cases), ossification (11 cases), amyloid deposits (14 cases), hemosiderosis (six cases), granular/smudgy calcification (four cases), synovial cell linings (three cases), and severe inflammatory infiltrates (one case). These histologic features of our cysts-LF were shared by previously reported "cysts-LF." Fourteen cysts-LF demonstrated vascular stenosis/occlusion, and eight showed thick hyalinized vessels, suggesting local circulatory insufficiency. Eight cases (44%) exhibited lipomembranous fat necrosis, accompanied by hyalinized vascular changes (p = 0.003). Ischemic conditions were observed in nearly half of the present cysts-LF, and may be one of the main contributing factors for the formation of cysts-LF, via degeneration and cystic changes in the LF.


Asunto(s)
Quistes , Ligamento Amarillo , Humanos , Femenino , Masculino , Anciano , Ligamento Amarillo/patología , Persona de Mediana Edad , Adulto , Anciano de 80 o más Años , Quistes/patología , Isquemia/patología
19.
Free Radic Biol Med ; 222: 607-624, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39009244

RESUMEN

Autophagy is essential for the adaptive response to exercise and physiological skeletal muscle functionality. However, the mechanisms leading to the activation of macroautophagy and chaperone-mediated autophagy in human skeletal muscle in response to high-intensity exercise remain elusive. Our findings demonstrate that macroautophagy and chaperone-mediated autophagy are stimulated by high-intensity exercise in normoxia (PIO2: 143 mmHg) and severe acute hypoxia (PIO2: 73 mmHg) in healthy humans. High-intensity exercise induces macroautophagy initiation through AMPKα phosphorylation, which phosphorylates and activates ULK1. ULK1 phosphorylates BECN1 at Ser15, eliciting the dissociation of BECN1-BCL2 crucial for phagophore formation. Besides, high-intensity exercise elevates the LC3B-II:LC3B-I ratio, reduces total SQSTM1/p62 levels, and induces p-Ser349 SQSTM1/p62 phosphorylation, suggesting heightened autophagosome degradation. PHAF1/MYTHO, a novel macroautophagy biomarker, is highly upregulated in response to high-intensity exercise. The latter is accompanied by elevated LAMP2A expression, indicating chaperone-mediated autophagy activation regardless of post-exercise HSPA8/HSC70 downregulation. Despite increased glycolytic metabolism, severe acute hypoxia does not exacerbate the autophagy signaling response. Signaling changes revert within 1 min of recovery with free circulation, while the application of immediate post-exercise ischemia impedes recovery. Our study concludes that macroautophagy and chaperone-mediated autophagy pathways are strongly activated by high-intensity exercise, regardless of PO2, and that oxygenation is necessary to revert these signals to pre-exercise values. PHAF1/MYTHO emerges as a pivotal exercise-responsive autophagy marker positively associated with the LC3B-II:LC3B-I ratio.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia , Autofagia , Beclina-1 , Autofagia Mediada por Chaperones , Ejercicio Físico , Hipoxia , Músculo Esquelético , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ejercicio Físico/fisiología , Masculino , Fosforilación , Hipoxia/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Beclina-1/metabolismo , Beclina-1/genética , Autofagia Mediada por Chaperones/genética , Isquemia/metabolismo , Isquemia/patología , Proteína Sequestosoma-1/metabolismo , Proteína Sequestosoma-1/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adulto , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas del Choque Térmico HSC70/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Femenino
20.
Bull Exp Biol Med ; 177(1): 26-29, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38954303

RESUMEN

We present a two-stage model for the study of chronic hind limb ischemia in rats. In the area of ischemia, sclerotic changes with atrophic rhabdomyocytes and reduced vascularization were revealed. CD31 expression in the endothelium increased proportionally to the number of vessels in the ischemic zone, and at the same time, focal expression of ßIII-tubulin was detected in the newly formed nerve fibers. These histological features are equivalent to the development of peripheral arterial disease in humans, which allows using our model in the search for new therapeutic strategies.


Asunto(s)
Modelos Animales de Enfermedad , Miembro Posterior , Isquemia , Músculo Esquelético , Animales , Ratas , Músculo Esquelético/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/irrigación sanguínea , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Isquemia/patología , Isquemia/metabolismo , Isquemia/fisiopatología , Masculino , Ratas Wistar , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Tubulina (Proteína)/metabolismo , Enfermedad Arterial Periférica/patología , Enfermedad Arterial Periférica/metabolismo , Enfermedad Arterial Periférica/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA