Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Skelet Muscle ; 14(1): 13, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38867250

RESUMEN

BACKGROUND: Adult muscle-resident myogenic stem cells, satellite cells (SCs), that play non-redundant role in muscle regeneration, are intrinsically impaired in Duchenne muscular dystrophy (DMD). Previously we revealed that dystrophic SCs express low level of anti-inflammatory and anti-oxidative heme oxygenase-1 (HO-1, HMOX1). Here we assess whether targeted induction of HMOX1 affect SC function and alleviates hallmark symptoms of DMD. METHODS: We generated double-transgenic mouse model (mdx;HMOX1Pax7Ind) that allows tamoxifen (TX)-inducible HMOX1 expression in Pax7 positive cells of dystrophic muscles. Mdx;HMOX1Pax7Ind and control mdx mice were subjected to 5-day TX injections (75 mg/kg b.w.) followed by acute exercise protocol with high-speed treadmill (12 m/min, 45 min) and downhill running to worsen skeletal muscle phenotype and reveal immediate effects of HO-1 on muscle pathology and SC function. RESULTS: HMOX1 induction caused a drop in SC pool in mdx;HMOX1Pax7Ind mice (vs. mdx counterparts), while not exaggerating the effect of physical exercise. Upon physical exercise, the proliferation of SCs and activated CD34- SC subpopulation, was impaired in mdx mice, an effect that was reversed in mdx;HMOX1Pax7Ind mice, however, both in vehicle- and TX-treated animals. This corresponded to the pattern of HO-1 expression in skeletal muscles. At the tissue level, necrotic events of selective skeletal muscles of mdx mice and associated increase in circulating levels of muscle damage markers were blunted in HO-1 transgenic animals which showed also anti-inflammatory cytokine profile (vs. mdx). CONCLUSIONS: Targeted expression of HMOX1 plays protective role in DMD and alleviates dystrophic muscle pathology.


Asunto(s)
Hemo-Oxigenasa 1 , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético , Distrofia Muscular de Duchenne , Células Satélite del Músculo Esquelético , Animales , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ratones , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Masculino , Ratones Endogámicos C57BL , Condicionamiento Físico Animal , Proteínas de la Membrana
2.
Am J Physiol Renal Physiol ; 327(1): F171-F183, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38779751

RESUMEN

Sepsis-associated acute kidney injury (SA-AKI) is a key contributor to the life-threatening sequelae attributed to sepsis. Mechanistically, SA-AKI is a consequence of unabated myeloid cell activation and oxidative stress that induces tubular injury. Iron mediates inflammatory pathways directly and through regulating the expression of myeloid-derived ferritin, an iron storage protein comprising ferritin light (FtL) and ferritin heavy chain (FtH) subunits. Previous work revealed that myeloid FtH deletion leads to a compensatory increase in intracellular and circulating FtL and is associated with amelioration of SA-AKI. We designed this study to test the hypothesis that loss of myeloid FtL and subsequently, circulating FtL will exacerbate the sepsis-induced inflammatory response and worsen SA-AKI. We generated a novel myeloid-specific FtL knockout mouse (FtLLysM-/-) and induced sepsis via cecal ligation and puncture or lipopolysaccharide endotoxemia. As expected, serum ferritin levels were significantly lower in the knockout mice, suggesting that myeloid cells dominantly contribute to circulating ferritin. Interestingly, although sepsis induction led to a marked production of pro- and anti-inflammatory cytokines, there was no statistical difference between the genotypes. There was a similar loss of kidney function, as evidenced by a rise in serum creatinine and cystatin C and renal injury identified by expression of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin. Finally, RNA sequencing revealed upregulation of pathways for cell cycle arrest and autophagy postsepsis, but no significant differences were observed between genotypes, including in key genes associated with ferroptosis, an iron-mediated form of cell death. The loss of FtL did not impact sepsis-mediated activation of NF-κB or HIF-1a signaling, key inflammatory pathways associated with dysregulated host response. Taken together, while FtL overexpression was shown to be protective against sepsis, the loss of FtL did not influence sepsis pathogenesis.NEW & NOTEWORTHY Hyperferritinemia in sepsis is often associated with a proinflammatory phenotype and poor prognosis. We previously showed the myeloid deletion of FtH results in a compensatory increase in FtL and is associated with reduced circulating cytokines and decreased rates of SA-AKI in animal sepsis models. Here, we show that myeloid deletion of FtL does not impact the severity of SA-AKI following CLP or LPS, suggesting that FtH plays the predominant role in propagating myeloid-induced proinflammatory pathways.


Asunto(s)
Lesión Renal Aguda , Apoferritinas , Ratones Noqueados , Sepsis , Animales , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Sepsis/metabolismo , Sepsis/complicaciones , Sepsis/genética , Apoferritinas/genética , Apoferritinas/metabolismo , Células Mieloides/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Riñón/metabolismo , Riñón/patología , Ratones Endogámicos C57BL , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo
4.
Clin Immunol ; 248: 109213, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36566913

RESUMEN

Ferroptosis is a druggable, iron-dependent form of cell death that is characterized by lipid peroxidation but has received little attention in lupus nephritis. Kidneys of lupus nephritis patients and mice showed increased lipid peroxidation mainly in the tubular segments and an increase in Acyl-CoA synthetase long-chain family member 4, a pro-ferroptosis enzyme. Nephritic mice had an attenuated expression of SLC7A11, a cystine importer, an impaired glutathione synthesis pathway, and low expression of glutathione peroxidase 4, a ferroptosis inhibitor. Lipidomics of nephritic kidneys confirmed ferroptosis. Using nephrotoxic serum, we induced immune complex glomerulonephritis in congenic mice and demonstrate that impaired iron sequestration within the proximal tubules exacerbates ferroptosis. Lupus nephritis patient serum rendered human proximal tubular cells susceptibility to ferroptosis which was inhibited by Liproxstatin-2, a novel ferroptosis inhibitor. Collectively, our findings identify intra-renal ferroptosis as a pathological feature and contributor to tubular injury in human and murine lupus nephritis.


Asunto(s)
Ferroptosis , Enfermedades Renales , Nefritis Lúpica , Humanos , Ratones , Animales , Hierro/metabolismo , Glomérulos Renales/metabolismo , Células Epiteliales/metabolismo
5.
Front Med (Lausanne) ; 9: 894521, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36160140

RESUMEN

Acute kidney injury (AKI) is a serious complication of rhabdomyolysis that significantly impacts survival. Myoglobin released from the damaged muscle accumulates in the kidney, causing heme iron-mediated oxidative stress, tubular cell death, and inflammation. In response to injury, myeloid cells, specifically neutrophils and macrophages, infiltrate the kidneys, and mediate response to injury. Ferritin, comprised of ferritin light chain and ferritin heavy chain (FtH), is vital for intracellular iron handling. Given the dominant role of macrophages and heme-iron burden in the pathogenesis of rhabdomyolysis, we studied the functional role of myeloid FtH in rhabdomyolysis-induced AKI and subsequent fibrosis. Using two models of rhabdomyolysis induced AKI, we found that during the acute phase, myeloid FtH deletion did not impact rhabdomyolysis-induced kidney injury, cell death or cell proliferation, suggesting that tubular heme burden is the dominant injury mechanism. We also determined that, while the kidney architecture was markedly improved after 28 days, tubular casts persisted in the kidneys, suggesting sustained damage or incomplete recovery. We further showed that rhabdomyolysis resulted in an abundance of disparate intra-renal immune cell populations, such that myeloid populations dominated during the acute phase and lymphoid populations dominated in the chronic phase. Fibrotic remodeling was induced in both genotypes at 7 days post-injury but continued to progress only in wild-type mice. This was accompanied by an increase in expression of pro-fibrogenic and immunomodulatory proteins, such as transforming growth factor-ß, S100A8, and tumor necrosis factor-α. Taken together, we found that while the initial injury response to heme burden was similar, myeloid FtH deficiency was associated with lesser interstitial fibrosis. Future studies are warranted to determine whether this differential fibrotic remodeling will render these animals more susceptible to a second AKI insult or progress to chronic kidney disease at an accelerated pace.

6.
Am J Physiol Renal Physiol ; 321(6): F675-F688, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34658261

RESUMEN

Expansion of renal lymphatic networks, or lymphangiogenesis (LA), is well recognized during development and is now being implicated in kidney diseases. Although LA is associated with multiple pathological conditions, very little is known about its role in acute kidney injury. The purpose of this study was to evaluate the role of LA in a model of cisplatin-induced nephrotoxicity. LA is predominately regulated by vascular endothelial growth factor (VEGF)-C and VEGF-D, ligands that exert their function through their cognate receptor VEGF receptor 3 (VEGFR3). We demonstrated that use of MAZ51, a selective VEGFR3 inhibitor, caused significantly worse structural and functional kidney damage in cisplatin nephrotoxicity. Apoptotic cell death and inflammation were also increased in MAZ51-treated animals compared with vehicle-treated animals following cisplatin administration. Notably, MAZ51 caused significant upregulation of intrarenal phospho-NF-κB, phospho-JNK, and IL-6. Cisplatin nephrotoxicity is associated with vascular congestion due to endothelial dysfunction. Using three-dimensional tissue cytometry, a novel approach to explore lymphatics in the kidney, we detected significant vascular autofluorescence attributed to erythrocytes in cisplatin alone-treated animals. Interestingly, no such congestion was detected in MAZ51-treated animals. We found increased renal vascular damage in MAZ51-treated animals, whereby MAZ51 caused a modest decrease in the endothelial markers endomucin and von Willebrand factor, with a modest increase in VEGFR2. Our findings identify a protective role for de novo LA in cisplatin nephrotoxicity and provide a rationale for the development of therapeutic approaches targeting LA. Our study also suggests off-target effects of MAZ51 on the vasculature in the setting of cisplatin nephrotoxicity.NEW & NOTEWORTHY Little is known about injury-associated LA in the kidney and its role in the pathophysiology of acute kidney injury (AKI). Observed exacerbation of cisplatin-induced AKI after LA inhibition was accompanied by increased medullary damage and cell death in the kidney. LA inhibition also upregulated compensatory expression of LA regulatory proteins, including JNK and NF-κB. These data support the premise that LA is induced during AKI and lymphatic expansion is a protective mechanism in cisplatin nephrotoxicity.


Asunto(s)
Indoles/toxicidad , Enfermedades Renales/inducido químicamente , Riñón/efectos de los fármacos , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Naftalenos/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Cisplatino , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Riñón/enzimología , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/enzimología , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Vasos Linfáticos/enzimología , Vasos Linfáticos/patología , Vasos Linfáticos/fisiopatología , Masculino , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Fosforilación , Transducción de Señal , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
JCI Insight ; 6(8)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33784251

RESUMEN

Despite the recent launch of tolvaptan, the search for safer polycystic kidney disease (PKD) drugs continues. Ciclopirox (CPX) or its olamine salt (CPX-O) is contained in a number of commercially available antifungal agents. CPX is also reported to possess anticancer activity. Several mechanisms of action have been proposed, including chelation of iron and inhibition of iron-dependent enzymes. Here, we show that CPX-O inhibited in vitro cystogenesis of primary human PKD cyst-lining epithelial cells cultured in a 3D collagen matrix. To assess the in vivo role of CPX-O, we treated PKD mice with CPX-O. CPX-O reduced the kidney-to-body weight ratios of PKD mice. The CPX-O treatment was also associated with decreased cell proliferation, decreased cystic area, and improved renal function. Ferritin levels were markedly elevated in cystic kidneys of PKD mice, and CPX-O treatment reduced renal ferritin levels. The reduction in ferritin was associated with increased ferritinophagy marker nuclear receptor coactivator 4, which reversed upon CPX-O treatment in PKD mice. Interestingly, these effects on ferritin appeared independent of iron. These data suggest that CPX-O can induce ferritin degradation via ferritinophagy, which is associated with decreased cyst growth progression in PKD mice. Most importantly these data indicate that CPX-O has the potential to treat autosomal dominant PKD.


Asunto(s)
Antifúngicos/farmacología , Ciclopirox/farmacología , Quistes , Ferritinas/metabolismo , Riñón/efectos de los fármacos , Enfermedades Renales Poliquísticas , Animales , Antifúngicos/uso terapéutico , Proliferación Celular , Ciclopirox/uso terapéutico , Colágeno , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Humanos , Ratones Endogámicos C57BL , Coactivadores de Receptor Nuclear/metabolismo , Tamaño de los Órganos , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/patología , Riñón Poliquístico Autosómico Dominante
8.
Nephron ; 144(12): 616-620, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32694248

RESUMEN

Sepsis associated acute kidney injury (SA-AKI) is a common clinical syndrome that occurs among hospitalized patients and significantly impacts mortality. Furthermore, survival after sepsis is intricately dependent on recovery of kidney function. In this review, we discuss the role of iron imbalance in mediating the pathogenic events during sepsis. Intracellular ferritin serves as a repository for iron and prevents iron-mediated injury and may limit the availability of iron to pathogens. Circulating levels of ferritin also increase during sepsis and often correlate with severity of sepsis. Herein, we examine preclinical and clinical data and discuss recent findings that suggest immunomodulatory roles for ferritin. We also discuss the possible mechanistic roles for ferritin in mitigating the pathogenic sequelae of sepsis and highlight current gaps in knowledge.


Asunto(s)
Lesión Renal Aguda/etiología , Ferritinas/metabolismo , Homeostasis , Hierro/metabolismo , Sepsis/complicaciones , Lesión Renal Aguda/metabolismo , Humanos
9.
Acta Neuropathol ; 140(4): 549-567, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32651669

RESUMEN

The proinflammatory cytokine interleukin 1 (IL-1) is crucially involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Herein, we studied the role of IL-1 signaling in blood-brain barrier (BBB) endothelial cells (ECs), astrocytes and microglia for EAE development, using mice with the conditional deletion of its signaling receptor IL-1R1. We found that IL-1 signaling in microglia and astrocytes is redundant for the development of EAE, whereas the IL-1R1 deletion in BBB-ECs markedly ameliorated disease severity. IL-1 signaling in BBB-ECs upregulated the expression of the adhesion molecules Vcam-1, Icam-1 and the chemokine receptor Darc, all of which have been previously shown to promote CNS-specific inflammation. In contrast, IL-1R1 signaling suppressed the expression of the stress-responsive heme catabolizing enzyme heme oxygenase-1 (HO-1) in BBB-ECs, promoting disease progression via a mechanism associated with deregulated expression of the IL-1-responsive genes Vcam1, Icam1 and Ackr1 (Darc). Mechanistically, our data emphasize a functional crosstalk of BBB-EC IL-1 signaling and HO-1, controlling the transcription of downstream proinflammatory genes promoting the pathogenesis of autoimmune neuroinflammation.


Asunto(s)
Barrera Hematoencefálica/enzimología , Encefalomielitis Autoinmune Experimental/inmunología , Células Endoteliales/enzimología , Hemo-Oxigenasa 1/metabolismo , Inflamación/inmunología , Interleucina-1/inmunología , Animales , Barrera Hematoencefálica/inmunología , Encefalomielitis Autoinmune Experimental/enzimología , Regulación de la Expresión Génica/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
10.
Semin Nephrol ; 40(2): 160-172, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32303279

RESUMEN

Ferritins are evolutionarily conserved proteins that regulate cellular iron metabolism. It is the only intracellular protein that is capable of storing large quantities of iron. Although the ratio of different subunits determines the iron content of each ferritin molecule, the exact mechanism that dictates organization of these subunits still is unclear. In this review, we address renal ferritin expression and its implication in kidney disease. Specifically, we address the role of ferritin subunits in preventing kidney injury and also promoting tolerance against infection-associated kidney injury. We describe functions for ferritin that are independent of its ability to ferroxidize and store iron. We further discuss the implications of ferritin in body fluids, including blood and urine, during inflammation and kidney disease. Although there are several in-depth review articles on ferritin in the context of iron metabolism, we chose to focus on the role of ferritin particularly in kidney health and disease and highlight unanswered questions in the field.


Asunto(s)
Lesión Renal Aguda/metabolismo , Carcinoma de Células Renales/metabolismo , Ferritinas/metabolismo , Neoplasias Renales/metabolismo , Insuficiencia Renal Crónica/metabolismo , Lesión Renal Aguda/etiología , Animales , Ferroptosis , Humanos , Sepsis/complicaciones
11.
Lab Invest ; 99(9): 1376-1388, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31019289

RESUMEN

Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected syndromes with significant attributable morbidity and mortality. The disturbing trend of increasing incidence and prevalence of these clinical disorders highlights the urgent need for better understanding of the underlying mechanisms that are involved in pathogenesis of these conditions. Lymphangiogenesis and its involvement in various inflammatory conditions is increasingly recognized while its role in AKI and CKD remains to be fully elucidated. Here, we studied lymphangiogenesis in three models of kidney injury. Our results demonstrate that the main ligands for lymphangiogenesis, VEGF-C and VEGF-D, are abundantly present in tubules at baseline conditions and the expression pattern of these ligands is significantly altered following injury. In addition, we show that both of these ligands increase in serum and urine post-injury and suggest that such increment may serve as novel urinary biomarkers of AKI as well as in progression of kidney disease. We also provide evidence that irrespective of the nature of initial insult, lymphangiogenic pathways are rapidly and robustly induced as evidenced by higher expression of lymphatic markers within the kidney.


Asunto(s)
Lesión Renal Aguda/metabolismo , Linfangiogénesis/fisiología , Insuficiencia Renal Crónica/metabolismo , Lesión Renal Aguda/patología , Animales , Modelos Animales de Enfermedad , Riñón/citología , Riñón/metabolismo , Riñón/patología , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Insuficiencia Renal Crónica/patología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor D de Crecimiento Endotelial Vascular/metabolismo
12.
Proc Natl Acad Sci U S A ; 116(12): 5681-5686, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30833408

RESUMEN

Malaria, the disease caused by Plasmodium spp. infection, remains a major global cause of morbidity and mortality. Host protection from malaria relies on immune-driven resistance mechanisms that kill Plasmodium However, these mechanisms are not sufficient per se to avoid the development of severe forms of disease. This is accomplished instead via the establishment of disease tolerance to malaria, a defense strategy that does not target Plasmodium directly. Here we demonstrate that the establishment of disease tolerance to malaria relies on a tissue damage-control mechanism that operates specifically in renal proximal tubule epithelial cells (RPTEC). This protective response relies on the induction of heme oxygenase-1 (HMOX1; HO-1) and ferritin H chain (FTH) via a mechanism that involves the transcription-factor nuclear-factor E2-related factor-2 (NRF2). As it accumulates in plasma and urine during the blood stage of Plasmodium infection, labile heme is detoxified in RPTEC by HO-1 and FTH, preventing the development of acute kidney injury, a clinical hallmark of severe malaria.


Asunto(s)
Hemo/metabolismo , Riñón/metabolismo , Malaria/fisiopatología , Animales , Apoferritinas/metabolismo , Línea Celular , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Ferritinas/metabolismo , Ferritinas/fisiología , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/fisiología , Humanos , Tolerancia Inmunológica/fisiología , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/fisiología , Oxidorreductasas , Plasmodium berghei/metabolismo , Plasmodium berghei/parasitología , Regulación hacia Arriba
13.
Front Immunol ; 10: 131, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30804939

RESUMEN

Despite the prevalence and recognition of its detrimental impact, clinical complications of sepsis remain a major challenge. Here, we investigated the effects of myeloid ferritin heavy chain (FtH) in regulating the pathogenic sequelae of sepsis. We demonstrate that deletion of myeloid FtH leads to protection against lipopolysaccharide-induced endotoxemia and cecal ligation and puncture (CLP)-induced model of sepsis as evidenced by reduced cytokine levels, multi-organ dysfunction and mortality. We identified that such protection is predominantly mediated by the compensatory increase in circulating ferritin (ferritin light chain; FtL) in the absence of myeloid FtH. Our in vitro and in vivo studies indicate that prior exposure to ferritin light chain restrains an otherwise dysregulated response to infection. These findings are mediated by an inhibitory action of FtL on NF-κB activation, a key signaling pathway that is implicated in the pathogenesis of sepsis. We further identified that LPS mediated activation of MAPK pathways, specifically, JNK, and ERK were also reduced with FtL pre-treatment. Taken together, our findings elucidate a crucial immunomodulatory function for circulating ferritin that challenges the traditional view of this protein as a mere marker of body iron stores. Accordingly, these findings will stimulate investigations to the adaptive nature of this protein in diverse clinical settings.


Asunto(s)
Apoferritinas/inmunología , Sepsis/inmunología , Animales , Ciego/cirugía , Citocinas/sangre , Escherichia coli , Femenino , Inflamación/sangre , Inflamación/etiología , Inflamación/inmunología , Ligadura , Lipopolisacáridos , Sistema de Señalización de MAP Quinasas , Macrófagos/inmunología , Masculino , Ratones , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/inmunología , Insuficiencia Multiorgánica/prevención & control , FN-kappa B/inmunología , Fagocitosis , Sepsis/sangre , Sepsis/complicaciones
14.
JCI Insight ; 4(2)2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30674729

RESUMEN

Acute kidney injury (AKI) is a devastating clinical condition affecting at least two-thirds of critically ill patients, and, among these patients, it is associated with a greater than 60% risk of mortality. Kidney mononuclear phagocytes (MPs) are implicated in pathogenesis and healing in mouse models of AKI and, thus, have been the subject of investigation as potential targets for clinical intervention. We have determined that, after injury, F4/80hi-expressing kidney-resident macrophages (KRMs) are a distinct cellular subpopulation that does not differentiate from nonresident infiltrating MPs. However, if KRMs are depleted using polyinosinic/polycytidylic acid (poly I:C), they can be reconstituted from bone marrow-derived precursors. Further, KRMs lack major histocompatibility complex class II (MHCII) expression before P7 but upregulate it over the next 14 days. This MHCII- KRM phenotype reappears after injury. RNA sequencing shows that injury causes transcriptional reprogramming of KRMs such that they more closely resemble that found at P7. KRMs after injury are also enriched in Wingless-type MMTV integration site family (Wnt) signaling, indicating that a pathway vital for mouse and human kidney development is active. These data indicate that mechanisms involved in kidney development may be functioning after injury in KRMs.

15.
JCI Insight ; 3(19)2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30282830

RESUMEN

Macrophages polarize into heterogeneous proinflammatory M1 and antiinflammatory M2 subtypes. Heme oxygenase 1 (HO-1) protects against inflammatory processes such as ischemia-reperfusion injury (IRI), organ transplantation, and atherosclerosis. To test our hypothesis that HO-1 regulates macrophage polarization and protects against IRI, we generated myeloid-specific HO-1-knockout (mHO-1-KO) and -transgenic (mHO-1-Tg) mice, with deletion or overexpression of HO-1, in various macrophage populations. Bone marrow-derived macrophages (BMDMs) from mHO-1-KO mice, treated with M1-inducing LPS or M2-inducing IL-4, exhibited increased mRNA expression of M1 (CXCL10, IL-1ß, MCP1) and decreased expression of M2 (Arg1 and CD163) markers as compared with controls, while BMDMs from mHO-1-Tg mice displayed the opposite. A similar pattern was observed in the hepatic M1/M2 expression profile in a mouse model of liver IRI. mHO-1-KO mice displayed increased hepatocellular damage, serum AST/ALT levels, Suzuki's histological score of liver IRI, and neutrophil and macrophage infiltration, while mHO-1-Tg mice exhibited the opposite. In human liver transplant biopsies, subjects with higher HO-1 levels showed lower expression of M1 markers together with decreased hepatocellular damage and improved outcomes. In conclusion, myeloid HO-1 expression modulates macrophage polarization, and protects against liver IRI, at least in part by favoring an M2 phenotype.


Asunto(s)
Rechazo de Injerto/inmunología , Hemo-Oxigenasa 1/metabolismo , Trasplante de Hígado/efectos adversos , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Daño por Reperfusión/inmunología , Adolescente , Adulto , Aloinjertos/irrigación sanguínea , Aloinjertos/citología , Aloinjertos/patología , Animales , Biopsia , Modelos Animales de Enfermedad , Femenino , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/patología , Hemo-Oxigenasa 1/genética , Humanos , Hígado/irrigación sanguínea , Hígado/citología , Hígado/patología , Pruebas de Función Hepática , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Persona de Mediana Edad , Daño por Reperfusión/diagnóstico , Daño por Reperfusión/patología , Transducción de Señal/inmunología , Adulto Joven
16.
Am J Physiol Renal Physiol ; 314(5): F702-F714, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28515173

RESUMEN

Ferroptosis is an iron-dependent form of regulated nonapoptotic cell death, which contributes to damage in models of acute kidney injury (AKI). Heme oxygenase-1 (HO-1) is a cytoprotective enzyme induced in response to cellular stress, and is protective against AKI because of its antiapoptotic and anti-inflammatory properties. However, the role of HO-1 in regulating ferroptosis is unclear. The purpose of this study was to elucidate the role of HO-1 in regulating ferroptotic cell death in renal proximal tubule cells (PTCs). Immortalized PTCs obtained from HO-1+/+ and HO-1-/- mice were treated with erastin or RSL3, ferroptosis inducers, in the presence or absence of antioxidants, an iron source, or an iron chelator. Cells were assessed for changes in morphology and metabolic activity as an indicator of cell viability. Treatment of HO-1+/+ PTCs with erastin resulted in a time- and dose-dependent increase in HO-1 gene expression and protein levels compared with vehicle-treated controls. HO-1-/- cells showed increased dose-dependent erastin- or RSL3-induced cell death in comparison to HO-1+/+ PTCs. Iron supplementation with ferric ammonium citrate in erastin-treated cells decreased cell viability further in HO-1-/- PTCs compared with HO-1+/+ cells. Cotreatment with ferrostatin-1 (ferroptosis inhibitor), deferoxamine (iron chelator), or N-acetyl-l-cysteine (glutathione replenisher) significantly increased cell viability and attenuated erastin-induced ferroptosis in both HO-1+/+ and HO-1-/- PTCs. These results demonstrate an important antiferroptotic role of HO-1 in renal epithelial cells.


Asunto(s)
Lesión Renal Aguda/enzimología , Hemo-Oxigenasa 1/metabolismo , Túbulos Renales Proximales/enzimología , Proteínas de la Membrana/metabolismo , Acetilcisteína/farmacología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Antioxidantes/farmacología , Carbolinas/toxicidad , Muerte Celular , Línea Celular , Ciclohexilaminas/farmacología , Deferoxamina/farmacología , Relación Dosis-Respuesta a Droga , Compuestos Férricos/toxicidad , Glutatión/metabolismo , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Quelantes del Hierro/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Noqueados , Fenilendiaminas/farmacología , Piperazinas/toxicidad , Compuestos de Amonio Cuaternario/toxicidad , Transducción de Señal , Factores de Tiempo
17.
Sci Rep ; 7(1): 9999, 2017 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-28855632

RESUMEN

Following myocardial infarction (MI), overactive inflammation remodels the left ventricle (LV) leading to heart failure coinciding with reduced levels of 15-epi-Lipoxin A4 (15-epi LXA4). However, the role of 15-epi LXA4 in post-MI acute inflammatory response and resolving phase is unclear. We hypothesize that liposomal fusion of 15-epi-LXA4 (Lipo-15-epi-LXA4) or free 15-epi-LXA4 will expedite the resolving phase in post-MI inflammation. 8 to 12-week-old male C57BL/6 mice were subjected to permanent coronary artery ligation. Lipo-15-epi-LXA4 or 15-epi-LXA4 (1 µg/kg/day) was injected 3 hours post-MI for (d)1 or continued daily till d5. 15-epi-LXA4 activated formyl peptide receptor (FPR2) and GPR120 on alternative macrophages but inhibited GPR40 on classical macrophages in-vitro. The 15-epi-LXA4 injected mice displayed reduced LV and lung mass to body weight ratios and improved ejection fraction at d5 post-MI. In the acute phase of inflammation-(d1), 15-epi-LXA4 primes neutrophil infiltration with a robust increase of Ccl2 and FPR2 expression. During the resolving phase-(d5), 15-epi-LXA4 initiated rapid neutrophils clearance with persistent activation of FPR2 in LV. Compared to MI-control, 15-epi-LXA4 injected mice showed reduced renal inflammation along with decreased levels of ngal and plasma creatinine. In summary, 15-epi-LXA4 initiates the resolving phase early to discontinue inflammation post-MI, thereby reducing LV dysfunction.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Lipoxinas/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Animales , Modelos Animales de Enfermedad , Ventrículos Cardíacos/patología , Pulmón/patología , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Resultado del Tratamiento , Remodelación Ventricular/efectos de los fármacos
18.
Am J Physiol Renal Physiol ; 313(3): F740-F755, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28679590

RESUMEN

Sex and age influence susceptibility to acute kidney injury (AKI), with young females exhibiting lowest incidence. In these studies, we investigated mechanisms which may underlie the sex/age-based dissimilarities. Cisplatin (Cp)-induced AKI resulted in morphological evidence of injury in all groups. A minimal rise in plasma creatinine (PCr) was seen in Young Females, whereas in Aged Females, PCr rose precipitously. Relative to Young Males, Aged Males showed significantly, but temporally, comparably elevated PCr. Notably, Aged Females showed significantly greater mortality, whereas Young Females exhibited none. Tissue KIM-1 and plasma NGAL were significantly lower in Young Females than all others. IGFBP7 levels were modestly increased in both Young groups. IGFBP7 levels in Aged Females were significantly elevated at baseline relative to Aged Males, and increased linearly through day 3, when these levels were comparable in both Aged groups. Plasma cytokine levels similarly showed a pattern of protective effects preferentially in Young Females. Expression of the drug transporter MATE2 did not explain the sex/age distinctions. Heme oxygenase-1 (HO-1) levels (~28-kDa species) showed elevation at day 1 in all groups with highest levels seen in Young Males. Exclusively in Young Females, these levels returned to baseline on day 3, suggestive of a more efficient recovery. In aggregate, we demonstrate, for the first time, a distinctive pattern of response to AKI in Young Females relative to males which appears to be significantly altered in aging. These distinctions may offer novel targets to exploit therapeutically in both females and males in the treatment of AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Envejecimiento/metabolismo , Riñón/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Factores de Edad , Envejecimiento/patología , Animales , Autofagia , Proliferación Celular , Cisplatino , Creatinina/sangre , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Hemo-Oxigenasa 1/metabolismo , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Riñón/patología , Lipocalina 2/sangre , Masculino , Proteínas de la Membrana/metabolismo , Metionina Adenosiltransferasa/metabolismo , Ratones Endogámicos C57BL , Factores Sexuales , Transducción de Señal , Factores de Tiempo
19.
Am J Kidney Dis ; 69(4): 531-545, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28139396

RESUMEN

A common clinical condition, acute kidney injury (AKI) significantly influences morbidity and mortality, particularly in critically ill patients. The pathophysiology of AKI is complex and involves multiple pathways, including inflammation, autophagy, cell-cycle progression, and oxidative stress. Recent evidence suggests that a single insult to the kidney significantly enhances the propensity to develop chronic kidney disease. Therefore, the generation of effective therapies against AKI is timely. In this context, the cytoprotective effects of heme oxygenase 1 (HO-1) in animal models of AKI are well documented. HO-1 modulates oxidative stress, autophagy, and inflammation and regulates the progression of cell cycle via direct and indirect mechanisms. These beneficial effects of HO-1 induction during AKI are mediated in part by the by-products of the HO reaction (iron, carbon monoxide, and bile pigments). This review highlights recent advances in the molecular mechanisms of HO-1-mediated cytoprotection and discusses the translational potential of HO-1 induction in AKI.


Asunto(s)
Lesión Renal Aguda/sangre , Lesión Renal Aguda/terapia , Hemo-Oxigenasa 1/sangre , Accidentes de Trabajo , Adulto , Autofagia/fisiología , Puntos de Control del Ciclo Celular/fisiología , Inducción Enzimática/fisiología , Hemo-Oxigenasa 1/fisiología , Humanos , Inflamación/sangre , Inflamación/terapia , Traumatismos de la Pierna/complicaciones , Masculino , Estrés Oxidativo/fisiología , Rabdomiólisis/sangre , Rabdomiólisis/terapia , Investigación Biomédica Traslacional
20.
Am J Physiol Renal Physiol ; 310(10): F1136-47, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26911846

RESUMEN

Acute kidney injury (AKI) is one of the leading causes of in-hospital morbidity and mortality, particularly in critically ill patients. Although our understanding of AKI at the molecular level remains limited due to its complex pathophysiology, recent advances in both quantitative and spatial mass spectrometric approaches offer new opportunities to assess the significance of renal metabolomic changes in AKI models. In this study, we evaluated lipid changes in early ischemia-reperfusion (IR)-related AKI in mice by using sequential window acquisition of all theoretical spectra (SWATH)-mass spectrometry (MS) lipidomics. We found a significant increase in two abundant ether-linked phospholipids following IR at 6 h postinjury, a plasmanyl choline, phosphatidylcholine (PC) O-38:1 (O-18:0, 20:1), and a plasmalogen, phosphatidylethanolamine (PE) O-42:3 (O-20:1, 22:2). Both of these lipids correlated with the severity of AKI as measured by plasma creatinine. In addition to many more renal lipid changes associated with more severe AKI, PC O-38:1 elevations were maintained at 24 h post-IR, while renal PE O-42:3 levels decreased, as were all ether PEs detected by SWATH-MS at this later time point. To further assess the significance of this early increase in PC O-38:1, we used matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS) to determine that it occurred in proximal tubules, a region of the kidney that is most prone to IR injury and also rich in the rate-limiting enzymes involved in ether-linked phospholipid biosynthesis. Use of SWATH-MS lipidomics in conjunction with MALDI-IMS for lipid localization will help in elucidating the role of lipids in the pathobiology of AKI.


Asunto(s)
Lesión Renal Aguda/metabolismo , Metabolismo de los Lípidos , Metabolómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Lesión Renal Aguda/etiología , Animales , Masculino , Ratones Endogámicos C57BL , Daño por Reperfusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA