Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Sci Adv ; 9(12): eadd5028, 2023 03 22.
Article En | MEDLINE | ID: mdl-36947620

Endothelial cells (ECs) grant access of disseminated cancer cells to distant organs. However, the molecular players regulating the activation of quiescent ECs at the premetastatic niche (PMN) remain elusive. Here, we find that ECs at the PMN coexpress tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its cognate death receptor 5 (DR5). Unexpectedly, endothelial TRAIL interacts intracellularly with DR5 to prevent its signaling and preserve a quiescent vascular phenotype. In absence of endothelial TRAIL, DR5 activation induces EC death and nuclear factor κB/p38-dependent EC stickiness, compromising vascular integrity and promoting myeloid cell infiltration, breast cancer cell adhesion, and metastasis. Consistently, both down-regulation of endothelial TRAIL at the PMN by proangiogenic tumor-secreted factors and the presence of the endogenous TRAIL inhibitors decoy receptor 1 (DcR1) and DcR2 favor metastasis. This study discloses an intracrine mechanism whereby TRAIL blocks DR5 signaling in quiescent endothelia, acting as gatekeeper of the vascular barrier that is corrupted by the tumor during cancer cell dissemination.


Breast Neoplasms , Endothelial Cells , Humans , Female , Endothelial Cells/metabolism , Ligands , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand , Apoptosis/genetics , Tumor Necrosis Factor-alpha/pharmacology
2.
Nat Cancer ; 3(12): 1464-1483, 2022 12.
Article En | MEDLINE | ID: mdl-36522548

Solid tumors are generally characterized by an acidic tumor microenvironment (TME) that favors cancer progression, therapy resistance and immune evasion. By single-cell RNA-sequencing analysis in individuals with pancreatic ductal adenocarcinoma (PDAC), we reveal solute carrier family 4 member 4 (SLC4A4) as the most abundant bicarbonate transporter, predominantly expressed by epithelial ductal cells. Functionally, SLC4A4 inhibition in PDAC cancer cells mitigates the acidosis of the TME due to bicarbonate accumulation in the extracellular space and a decrease in lactate production by cancer cells as the result of reduced glycolysis. In PDAC-bearing mice, genetic or pharmacological SLC4A4 targeting improves T cell-mediated immune response and breaches macrophage-mediated immunosuppression, thus inhibiting tumor growth and metastases. In addition, Slc4a4 targeting in combination with immune checkpoint blockade is able to overcome immunotherapy resistance and prolong survival. Overall, our data propose SLC4A4 as a therapeutic target to unleash an antitumor immune response in PDAC.


Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Sodium-Bicarbonate Symporters , Animals , Mice , Bicarbonates/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Immunotherapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Sodium-Bicarbonate Symporters/genetics , Tumor Microenvironment , Immune Tolerance , Pancreatic Neoplasms
3.
Cell Mol Gastroenterol Hepatol ; 14(3): 609-624, 2022.
Article En | MEDLINE | ID: mdl-35714859

BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) is a multistep process whereby abnormally proliferating cancer cells undergo extensive metabolic reprogramming. Metabolic alterations in hepatocarcinogenesis depend on the activation of specific oncogenes, thus partially explaining HCC heterogeneity. c-Myc oncogene overexpression, frequently observed in human HCCs, leads to a metabolic rewiring toward a Warburg phenotype and production of lactate, resulting in the acidification of the extracellular space, favoring the emergence of an immune-permissive tumor microenvironment. Here, we investigated whether Ldha genetic ablation interferes with metabolic reprogramming and HCC development in the mouse. METHODS: We characterized the metabolic reprogramming in tumors induced in C57BL/6J mice hydrodynamically cotransfected with c-Myc and h-Ras. Using the same experimental model, we investigated the effect of Ldha inhibition-gained through the inducible and hepatocyte-specific Ldha knockout-on cancer cell metabolic reprogramming, number and size of HCC lesions, and tumor microenvironment alterations. RESULTS: c-Myc/h-Ras-driven tumors display a striking glycolytic metabolism, suggesting a switch to a Warburg phenotype. The tumors also exhibited enhanced pentose phosphate pathway activity, the switch of glutamine to sustain glutathione synthesis instead of the tricarboxylic acid cycle, and the impairment of oxidative phosphorylation. In addition, Ldha abrogation significantly hampered tumor number and size together with an evident inhibition of the Warburg-like metabolic feature and a remarkable increase of CD4+ lymphocytes compared with Ldha wild-type livers. CONCLUSIONS: These results demonstrate that Ldha deletion significantly impairs mouse HCC development and suggest lactate dehydrogenase as a potential target to enhance the efficacy of the current therapeutic options.


Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/pathology , Humans , Lactate Dehydrogenase 5 , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Oncogenes/genetics , Tumor Microenvironment/genetics
4.
Cancer Immunol Res ; 10(1): 126-141, 2022 01.
Article En | MEDLINE | ID: mdl-34815265

Cytotoxic T cell (CTL) infiltration of the tumor carries the potential to limit cancer progression, but their exclusion by the immunosuppressive tumor microenvironment hampers the efficiency of immunotherapy. Here, we show that expression of the axon guidance molecule Plexin-A4 (Plxna4) in CTLs, especially in effector/memory CD8+ T cells, is induced upon T-cell activation, sustained in the circulation, but reduced when entering the tumor bed. Therefore, we deleted Plxna4 and observed that Plxna4-deficient CTLs acquired improved homing capacity to the lymph nodes and to the tumor, as well as increased proliferation, both achieved through enhanced Rac1 activation. Mice with stromal or hematopoietic Plxna4 deletion exhibited enhanced CTL infiltration and impaired tumor growth. In a melanoma model, adoptive transfer of CTLs lacking Plxna4 prolonged survival and improved therapeutic outcome, which was even stronger when combined with anti-programmed cell death protein 1 (PD-1) treatment. PLXNA4 abundance in circulating CTLs was augmented in melanoma patients versus healthy volunteers but decreased after the first cycle of anti-PD-1, alone or in combination with anti-cytotoxic T-Lymphocyte Associated Protein 4 (CTLA-4), in those patients showing complete or partial response to the treatment. Altogether, our data suggest that Plxna4 acts as a "checkpoint," negatively regulating CTL migration and proliferation through cell-autonomous mechanisms independent of the interaction with host-derived Plxna4 ligands, semaphorins. These findings pave the way toward Plxna4-centric immunotherapies and propose Plxna4 detection in circulating CTLs as a potential way to monitor the response to immune checkpoint blockade in patients with metastatic melanoma.


Immunotherapy/methods , Lung Neoplasms/therapy , Melanoma, Experimental/therapy , Nerve Tissue Proteins/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocyte Activation , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Cell Surface/genetics , Tumor Microenvironment/immunology
5.
Nature ; 587(7835): 626-631, 2020 11.
Article En | MEDLINE | ID: mdl-33116312

Muscle regeneration is sustained by infiltrating macrophages and the consequent activation of satellite cells1-4. Macrophages and satellite cells communicate in different ways1-5, but their metabolic interplay has not been investigated. Here we show, in a mouse model, that muscle injuries and ageing are characterized by intra-tissue restrictions of glutamine. Low levels of glutamine endow macrophages with the metabolic ability to secrete glutamine via enhanced glutamine synthetase (GS) activity, at the expense of glutamine oxidation mediated by glutamate dehydrogenase 1 (GLUD1). Glud1-knockout macrophages display constitutively high GS activity, which prevents glutamine shortages. The uptake of macrophage-derived glutamine by satellite cells through the glutamine transporter SLC1A5 activates mTOR and promotes the proliferation and differentiation of satellite cells. Consequently, macrophage-specific deletion or pharmacological inhibition of GLUD1 improves muscle regeneration and functional recovery in response to acute injury, ischaemia or ageing. Conversely, SLC1A5 blockade in satellite cells or GS inactivation in macrophages negatively affects satellite cell functions and muscle regeneration. These results highlight the metabolic crosstalk between satellite cells and macrophages, in which macrophage-derived glutamine sustains the functions of satellite cells. Thus, the targeting of GLUD1 may offer therapeutic opportunities for the regeneration of injured or aged muscles.


Glutamine/metabolism , Macrophages/metabolism , Muscle, Skeletal/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/metabolism , Aging/metabolism , Amino Acid Transport System ASC/antagonists & inhibitors , Amino Acid Transport System ASC/metabolism , Animals , Cell Differentiation , Cell Proliferation , Female , Glutamate Dehydrogenase/deficiency , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Glutamate-Ammonia Ligase/antagonists & inhibitors , Glutamate-Ammonia Ligase/metabolism , Macrophages/enzymology , Male , Mice , Minor Histocompatibility Antigens/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Oxidation-Reduction , Satellite Cells, Skeletal Muscle/cytology , TOR Serine-Threonine Kinases
6.
Sci Signal ; 12(567)2019 02 05.
Article En | MEDLINE | ID: mdl-30723174

Intratumoral hypoxia causes the formation of dysfunctional blood vessels, which contribute to tumor metastasis and reduce the efficacy of therapeutic treatments. Blood vessels are embedded in the tumor stroma of which cancer-associated fibroblasts (CAFs) constitute a prominent cellular component. We found that hypoxic human mammary CAFs promoted angiogenesis in CAF-endothelial cell cocultures in vitro. Mass spectrometry-based proteomic analysis of the CAF secretome unraveled that hypoxic CAFs contributed to blood vessel abnormalities by altering their secretion of various pro- and anti-angiogenic factors. Hypoxia induced pronounced remodeling of the CAF proteome, including proteins that have not been previously related to this process. Among those, the uncharacterized protein NCBP2-AS2 that we renamed HIAR (hypoxia-induced angiogenesis regulator) was the protein most increased in abundance in hypoxic CAFs. Silencing of HIAR abrogated the pro-angiogenic and pro-migratory function of hypoxic CAFs by decreasing secretion of the pro-angiogenic factor VEGFA and consequently reducing VEGF/VEGFR downstream signaling in the endothelial cells. Our study has identified a regulator of angiogenesis and provides a map of hypoxia-induced molecular alterations in mammary CAFs.


Cancer-Associated Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hypoxia , Neovascularization, Pathologic/genetics , Proteome/metabolism , Proteomics/methods , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics
7.
EMBO J ; 36(16): 2373-2389, 2017 08 15.
Article En | MEDLINE | ID: mdl-28694244

Tumor progression alters the composition and physical properties of the extracellular matrix. Particularly, increased matrix stiffness has profound effects on tumor growth and metastasis. While endothelial cells are key players in cancer progression, the influence of tumor stiffness on the endothelium and the impact on metastasis is unknown. Through quantitative mass spectrometry, we find that the matricellular protein CCN1/CYR61 is highly regulated by stiffness in endothelial cells. We show that stiffness-induced CCN1 activates ß-catenin nuclear translocation and signaling and that this contributes to upregulate N-cadherin levels on the surface of the endothelium, in vitro This facilitates N-cadherin-dependent cancer cell-endothelium interaction. Using intravital imaging, we show that knockout of Ccn1 in endothelial cells inhibits melanoma cancer cell binding to the blood vessels, a critical step in cancer cell transit through the vasculature to metastasize. Targeting stiffness-induced changes in the vasculature, such as CCN1, is therefore a potential yet unappreciated mechanism to impair metastasis.


Cell Communication , Endothelial Cells/physiology , Melanocytes/physiology , Cadherins/analysis , Cell Line , Cysteine-Rich Protein 61/analysis , Gene Expression Regulation , Humans , Mass Spectrometry , beta Catenin/analysis
8.
Nat Commun ; 7: 13720, 2016 12 23.
Article En | MEDLINE | ID: mdl-28008905

Various steady state and inflamed tissues have been shown to contain a heterogeneous DC population consisting of developmentally distinct subsets, including cDC1s, cDC2s and monocyte-derived DCs, displaying differential functional specializations. The identification of functionally distinct tumour-associated DC (TADC) subpopulations could prove essential for the understanding of basic TADC biology and for envisaging targeted immunotherapies. We demonstrate that multiple mouse tumours as well as human tumours harbour ontogenically discrete TADC subsets. Monocyte-derived TADCs are prominent in tumour antigen uptake, but lack strong T-cell stimulatory capacity due to NO-mediated immunosuppression. Pre-cDC-derived TADCs have lymph node migratory potential, whereby cDC1s efficiently activate CD8+ T cells and cDC2s induce Th17 cells. Mice vaccinated with cDC2s displayed a reduced tumour growth accompanied by a reprogramming of pro-tumoural TAMs and a reduction of MDSCs, while cDC1 vaccination strongly induces anti-tumour CTLs. Our data might prove important for therapeutic interventions targeted at specific TADC subsets or their precursors.


Dendritic Cells/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Immunotherapy/methods , Macrophages/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/immunology , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocyte Subsets/immunology
9.
Cell Metab ; 24(5): 701-715, 2016 11 08.
Article En | MEDLINE | ID: mdl-27773694

Hypoxic tumor-associated macrophages (TAMs) acquire angiogenic and immunosuppressive properties. Yet it remains unknown if metabolic changes influence these functions. Here, we argue that hypoxic TAMs strongly upregulate the expression of REDD1, a negative regulator of mTOR. REDD1-mediated mTOR inhibition hinders glycolysis in TAMs and curtails their excessive angiogenic response, with consequent formation of abnormal blood vessels. Accordingly, REDD1 deficiency in TAMs leads to the formation of smoothly aligned, pericyte-covered, functional vessels, which prevents vessel leakiness, hypoxia, and metastases. Mechanistically, highly glycolytic REDD1-deficient TAMs outcompete endothelial cells for glucose usage that thwarts vascular hyperactivation and promotes the formation of quiescent vascular junctions. Tuning down glycolysis in REDD1 knockout TAMs re-establishes abnormal angiogenesis and metastases. On this basis, we prove that the anti-tumor effect of mTOR inhibitors is partly countered by the deleterious outcome of these drugs on TAMs. Our data provide a functional link between TAM metabolism and tumor angiogenesis.


Blood Vessels/growth & development , Macrophages/metabolism , Morphogenesis , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Animals , Blood Vessels/metabolism , Cell Hypoxia , Disease Models, Animal , Endothelial Cells/metabolism , Gene Deletion , Glucose/metabolism , Glycolysis , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Neoplasms/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Up-Regulation
10.
Nature ; 522(7556): 349-53, 2015 Jun 18.
Article En | MEDLINE | ID: mdl-25985180

Mutations or amplification of the MET proto-oncogene are involved in the pathogenesis of several tumours, which rely on the constitutive engagement of this pathway for their growth and survival. However, MET is expressed not only by cancer cells but also by tumour-associated stromal cells, although its precise role in this compartment is not well characterized. Here we show that MET is required for neutrophil chemoattraction and cytotoxicity in response to its ligand hepatocyte growth factor (HGF). Met deletion in mouse neutrophils enhances tumour growth and metastasis. This phenotype correlates with reduced neutrophil infiltration to both the primary tumour and metastatic sites. Similarly, Met is necessary for neutrophil transudation during colitis, skin rash or peritonitis. Mechanistically, Met is induced by tumour-derived tumour necrosis factor (TNF)-α or other inflammatory stimuli in both mouse and human neutrophils. This induction is instrumental for neutrophil transmigration across an activated endothelium and for inducible nitric oxide synthase production upon HGF stimulation. Consequently, HGF/MET-dependent nitric oxide release by neutrophils promotes cancer cell killing, which abates tumour growth and metastasis. After systemic administration of a MET kinase inhibitor, we prove that the therapeutic benefit of MET targeting in cancer cells is partly countered by the pro-tumoural effect arising from MET blockade in neutrophils. Our work identifies an unprecedented role of MET in neutrophils, suggests a potential 'Achilles' heel' of MET-targeted therapies in cancer, and supports the rationale for evaluating anti-MET drugs in certain inflammatory diseases.


Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Neoplasms/immunology , Neoplasms/metabolism , Neutrophils/immunology , Proto-Oncogene Proteins c-met/metabolism , Aged , Animals , Disease Models, Animal , Disease Progression , Female , Gene Deletion , Hepatocyte Growth Factor , Humans , Inflammation/immunology , Inflammation/pathology , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Neutrophils/drug effects , Neutrophils/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/deficiency , Proto-Oncogene Proteins c-met/genetics , Solubility , Transendothelial and Transepithelial Migration , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
11.
FASEB J ; 29(3): 960-72, 2015 Mar.
Article En | MEDLINE | ID: mdl-25414484

We studied the role of matrix metalloproteinase-10 (MMP-10) during skeletal muscle repair after ischemia using a model of femoral artery excision in wild-type (WT) and MMP-10 deficient (Mmp10(-/-)) mice. Functional changes were analyzed by small animal positron emission tomography and tissue morphology by immunohistochemistry. Gene expression and protein analysis were used to study the molecular mechanisms governed by MMP-10 in hypoxia. Early after ischemia, MMP-10 deficiency resulted in delayed tissue reperfusion (10%, P < 0.01) and in increased necrosis (2-fold, P < 0.01), neutrophil (4-fold, P < 0.01), and macrophage (1.5-fold, P < 0.01) infiltration. These differences at early time points resulted in delayed myotube regeneration in Mmp10(-/-) soleus at later stages (regenerating myofibers: 30 ± 9% WT vs. 68 ± 10% Mmp10(-/-), P < 0.01). The injection of MMP-10 into Mmp10(-/-) mice rescued the observed phenotype. A molecular analysis revealed higher levels of Cxcl1 mRNA (10-fold, P < 0.05) and protein (30%) in the ischemic Mmp10(-/-) muscle resulting from a lack of transcriptional inhibition by MMP-10. This was further confirmed using siRNA against MMP-10 in vivo. Our results demonstrate an important role of MMP-10 for proper muscle repair after ischemia, and suggest that chemokine regulation such as Cxcl1 by MMP-10 is involved in muscle regeneration.


Disease Models, Animal , Hindlimb/enzymology , Ischemia/prevention & control , Matrix Metalloproteinase 10/physiology , Muscular Diseases/prevention & control , Reperfusion Injury/prevention & control , Wound Healing/physiology , Animals , Blotting, Western , Chemokine CXCL1/metabolism , Elapid Venoms/toxicity , Hindlimb/injuries , Hindlimb/pathology , Ischemia/enzymology , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Muscular Diseases/chemically induced , Muscular Diseases/enzymology , Neurotoxins/toxicity , Regeneration , Reperfusion Injury/chemically induced , Reperfusion Injury/enzymology
12.
EMBO Mol Med ; 5(6): 843-57, 2013 Jun.
Article En | MEDLINE | ID: mdl-23616286

Occlusion of the main arterial route redirects blood flow to the collateral circulation. We previously reported that macrophages genetically modified to express low levels of prolyl hydroxylase domain protein 2 (PHD2) display an arteriogenic phenotype, which promotes the formation of collateral vessels and protects the skeletal muscle from ischaemic necrosis. However, the molecular mechanisms underlying this process are unknown. Here, we demonstrate that femoral artery occlusion induces a switch in macrophage phenotype through angiopoietin-1 (ANG1)-mediated Phd2 repression. ANG blockade by a soluble trap prevented the downregulation of Phd2 expression in macrophages and their phenotypic switch, thus inhibiting collateral growth. ANG1-dependent Phd2 repression initiated a feed-forward loop mediated by the induction of the ANG receptor TIE2 in macrophages. Gene silencing and cell depletion strategies demonstrate that TIE2 induction in macrophages is required to promote their proarteriogenic functions, enabling collateral vessel formation following arterial obstruction. These results indicate an indispensable role for TIE2 in sustaining in situ programming of macrophages to a proarteriogenic, M2-like phenotype, suggesting possible new venues for the treatment of ischaemic disorders.


Procollagen-Proline Dioxygenase/metabolism , Receptor, TIE-2/metabolism , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Animals , Down-Regulation , Gene Silencing , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Ischemia/metabolism , Ischemia/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , Phenotype , Procollagen-Proline Dioxygenase/genetics , Signal Transduction
13.
Nature ; 479(7371): 122-6, 2011 Oct 09.
Article En | MEDLINE | ID: mdl-21983962

PHD2 serves as an oxygen sensor that rescues blood supply by regulating vessel formation and shape in case of oxygen shortage. However, it is unknown whether PHD2 can influence arteriogenesis. Here we studied the role of PHD2 in collateral artery growth by using hindlimb ischaemia as a model, a process that compensates for the lack of blood flow in case of major arterial occlusion. We show that Phd2 (also known as Egln1) haplodeficient (Phd2(+/-)) mice displayed preformed collateral arteries that preserved limb perfusion and prevented tissue necrosis in ischaemia. Improved arteriogenesis in Phd2(+/-) mice was due to an expansion of tissue-resident, M2-like macrophages and their increased release of arteriogenic factors, leading to enhanced smooth muscle cell (SMC) recruitment and growth. Both chronic and acute deletion of one Phd2 allele in macrophages was sufficient to skew their polarization towards a pro-arteriogenic phenotype. Mechanistically, collateral vessel preconditioning relied on the activation of canonical NF-κB pathway in Phd2(+/-) macrophages. These results unravel how PHD2 regulates arteriogenesis and artery homeostasis by controlling a specific differentiation state in macrophages and suggest new treatment options for ischaemic disorders.


Arteries/growth & development , Extremities/blood supply , Ischemia/prevention & control , Macrophages/metabolism , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/metabolism , Alleles , Animals , Disease Models, Animal , Extremities/pathology , Female , Heterozygote , Homeostasis , Hypoxia-Inducible Factor-Proline Dioxygenases , Ischemia/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Myocytes, Smooth Muscle/cytology , NF-kappa B/metabolism , Necrosis , Phenotype , Procollagen-Proline Dioxygenase/genetics
...