Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 16(1): 132-157, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38177536

ABSTRACT

Thoracic aortic aneurysm and dissection (TAAD) is a life-threatening condition associated with Marfan syndrome (MFS), a disease caused by fibrillin-1 gene mutations. While various conditions causing TAAD exhibit aortic accumulation of the proteoglycans versican (Vcan) and aggrecan (Acan), it is unclear whether these ECM proteins are involved in aortic disease. Here, we find that Vcan, but not Acan, accumulated in Fbn1C1041G/+ aortas, a mouse model of MFS. Vcan haploinsufficiency protected MFS mice against aortic dilation, and its silencing reverted aortic disease by reducing Nos2 protein expression. Our results suggest that Acan is not an essential contributor to MFS aortopathy. We further demonstrate that Vcan triggers Akt activation and that pharmacological Akt pathway inhibition rapidly regresses aortic dilation and Nos2 expression in MFS mice. Analysis of aortic tissue from MFS human patients revealed accumulation of VCAN and elevated pAKT-S473 staining. Together, these findings reveal that Vcan plays a causative role in MFS aortic disease in vivo by inducing Nos2 via Akt activation and identify Akt signaling pathway components as candidate therapeutic targets.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Diseases , Aortic Dissection , Azides , Deoxyglucose , Marfan Syndrome , Animals , Humans , Mice , Aortic Aneurysm, Thoracic/complications , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Diseases/complications , Deoxyglucose/analogs & derivatives , Marfan Syndrome/complications , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Nitric Oxide Synthase Type II/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Versicans/metabolism
2.
Int J Mol Sci ; 24(9)2023 05 03.
Article in English | MEDLINE | ID: mdl-37175915

ABSTRACT

Cardiovascular disease (CVD) frequently complicates chronic kidney disease (CKD). The risk of all-cause mortality increases from 20% to 500% in patients who suffer both conditions; this is referred to as the so-called cardio-renal syndrome (CRS). Preclinical studies have described the key role of mitochondrial dysfunction in cardiovascular and renal diseases, suggesting that maintaining mitochondrial homeostasis is a promising therapeutic strategy for CRS. In this review, we explore the malfunction of mitochondrial homeostasis (mitochondrial biogenesis, dynamics, oxidative stress, and mitophagy) and how it contributes to the development and progression of the main vascular pathologies that could be affected by kidney injury and vice versa, and how this knowledge may guide the development of novel therapeutic strategies in CRS.


Subject(s)
Cardio-Renal Syndrome , Renal Insufficiency, Chronic , Humans , Kidney/metabolism , Heart , Renal Insufficiency, Chronic/metabolism , Mitochondria
3.
Sci Adv ; 8(11): eabm7322, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35294231

ABSTRACT

Pathological vascular remodeling is the underlying cause of atherosclerosis and abdominal aortic aneurysm (AAA). Here, we analyzed the role of galectin-1 (Gal-1), a ß-galactoside-binding protein, as a therapeutic target for atherosclerosis and AAA. Mice lacking Gal-1 (Lgals1-/-) developed severe atherosclerosis induced by pAAV/D377Y-mPCSK9 adenovirus and displayed higher lipid levels and lower expression of contractile markers of vascular smooth muscle cells (VSMCs) in plaques than wild-type mice. Proteomic analysis of Lgals1-/- aortas showed changes in markers of VSMC phenotypic switch and altered composition of mitochondrial proteins. Mechanistically, Gal-1 silencing resulted in increased foam cell formation and mitochondrial dysfunction in VSMCs, while treatment with recombinant Gal-1 (rGal-1) prevented these effects. Furthermore, rGal-1 treatment attenuated atherosclerosis and elastase-induced AAA, leading to higher contractile VSMCs in aortic tissues. Gal-1 expression decreased in human atheroma and AAA compared to control tissue. Thus, Gal-1-driven circuits emerge as potential therapeutic strategies in atherosclerosis and AAA.


Subject(s)
Aortic Aneurysm, Abdominal , Atherosclerosis , Animals , Aortic Aneurysm, Abdominal/etiology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Disease Models, Animal , Galectin 1/genetics , Galectin 1/metabolism , Galectin 1/pharmacology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Proteomics , Vascular Remodeling
4.
Arterioscler Thromb Vasc Biol ; 42(4): 462-469, 2022 04.
Article in English | MEDLINE | ID: mdl-35196876

ABSTRACT

BACKGROUND: The goal of this study was to determine whether boosting mitochondrial respiration prevents the development of fatal aortic ruptures triggered by atherosclerosis and hypertension. METHODS: Ang-II (angiotensin-II) was infused in ApoE (Apolipoprotein E)-deficient mice fed with a western diet to induce acute aortic aneurysms and lethal ruptures. RESULTS: We found decreased mitochondrial respiration and mitochondrial proteins in vascular smooth muscle cells from murine and human aortic aneurysms. Boosting NAD levels with nicotinamide riboside reduced the development of aortic aneurysms and sudden death by aortic ruptures. CONCLUSIONS: Targetable vascular metabolism is a new clinical strategy to prevent fatal aortic ruptures and sudden death in patients with aortic aneurysms.


Subject(s)
Aortic Rupture , Atherosclerosis , Angiotensin II , Animals , Aortic Rupture/genetics , Aortic Rupture/prevention & control , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Death, Sudden , Humans , Mice , Mitochondrial Proteins
5.
Circulation ; 143(21): 2091-2109, 2021 05 25.
Article in English | MEDLINE | ID: mdl-33709773

ABSTRACT

BACKGROUND: Marfan syndrome (MFS) is an autosomal dominant disorder of the connective tissue caused by mutations in the FBN1 (fibrillin-1) gene encoding a large glycoprotein in the extracellular matrix called fibrillin-1. The major complication of this connective disorder is the risk to develop thoracic aortic aneurysm. To date, no effective pharmacologic therapies have been identified for the management of thoracic aortic disease and the only options capable of preventing aneurysm rupture are endovascular repair or open surgery. Here, we have studied the role of mitochondrial dysfunction in the progression of thoracic aortic aneurysm and mitochondrial boosting strategies as a potential treatment to managing aortic aneurysms. METHODS: Combining transcriptomics and metabolic analysis of aortas from an MFS mouse model (Fbn1c1039g/+) and MFS patients, we have identified mitochondrial dysfunction alongside with mtDNA depletion as a new hallmark of aortic aneurysm disease in MFS. To demonstrate the importance of mitochondrial decline in the development of aneurysms, we generated a conditional mouse model with mitochondrial dysfunction specifically in vascular smooth muscle cells (VSMC) by conditional depleting Tfam (mitochondrial transcription factor A; Myh11-CreERT2Tfamflox/flox mice). We used a mouse model of MFS to test for drugs that can revert aortic disease by enhancing Tfam levels and mitochondrial respiration. RESULTS: The main canonical pathways highlighted in the transcriptomic analysis in aortas from Fbn1c1039g/+ mice were those related to metabolic function, such as mitochondrial dysfunction. Mitochondrial complexes, whose transcription depends on Tfam and mitochondrial DNA content, were reduced in aortas from young Fbn1c1039g/+ mice. In vitro experiments in Fbn1-silenced VSMCs presented increased lactate production and decreased oxygen consumption. Similar results were found in MFS patients. VSMCs seeded in matrices produced by Fbn1-deficient VSMCs undergo mitochondrial dysfunction. Conditional Tfam-deficient VSMC mice lose their contractile capacity, showed aortic aneurysms, and died prematurely. Restoring mitochondrial metabolism with the NAD precursor nicotinamide riboside rapidly reverses aortic aneurysm in Fbn1c1039g/+ mice. CONCLUSIONS: Mitochondrial function of VSMCs is controlled by the extracellular matrix and drives the development of aortic aneurysm in Marfan syndrome. Targeting vascular metabolism is a new available therapeutic strategy for managing aortic aneurysms associated with genetic disorders.


Subject(s)
Aortic Aneurysm/physiopathology , Marfan Syndrome/genetics , Mitochondria/metabolism , Animals , Disease Models, Animal , Humans , Marfan Syndrome/physiopathology , Mice
6.
Arterioscler Thromb Vasc Biol ; 40(10): 2408-2424, 2020 10.
Article in English | MEDLINE | ID: mdl-32847388

ABSTRACT

OBJECTIVE: microRNAs are master regulators of gene expression with essential roles in virtually all biological processes. miR-217 has been associated with aging and cellular senescence, but its role in vascular disease is not understood. Approach and Results: We have used an inducible endothelium-specific knock-in mouse model to address the role of miR-217 in vascular function and atherosclerosis. miR-217 reduced NO production and promoted endothelial dysfunction, increased blood pressure, and exacerbated atherosclerosis in proatherogenic apoE-/- mice. Moreover, increased endothelial miR-217 expression led to the development of coronary artery disease and altered left ventricular heart function, inducing diastolic and systolic dysfunction. Conversely, inhibition of endogenous vascular miR-217 in apoE-/- mice improved vascular contractility and diminished atherosclerosis. Transcriptome analysis revealed that miR-217 regulates an endothelial signaling hub and downregulates a network of eNOS (endothelial NO synthase) activators, including VEGF (vascular endothelial growth factor) and apelin receptor pathways, resulting in diminished eNOS expression. Further analysis revealed that human plasma miR-217 is a biomarker of vascular aging and cardiovascular risk. CONCLUSIONS: Our results highlight the therapeutic potential of miR-217 inhibitors in aging-related cardiovascular disease.


Subject(s)
Aging/metabolism , Atherosclerosis/metabolism , Endothelial Cells/metabolism , MicroRNAs/metabolism , Plaque, Atherosclerotic , Age Factors , Aged, 80 and over , Aging/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Case-Control Studies , Cells, Cultured , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Disease Models, Animal , Endothelial Cells/pathology , Female , Hemodynamics , Humans , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/blood , MicroRNAs/genetics , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Signal Transduction , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left
7.
Science ; 368(6497): 1371-1376, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32439659

ABSTRACT

The effect of immunometabolism on age-associated diseases remains uncertain. In this work, we show that T cells with dysfunctional mitochondria owing to mitochondrial transcription factor A (TFAM) deficiency act as accelerators of senescence. In mice, these cells instigate multiple aging-related features, including metabolic, cognitive, physical, and cardiovascular alterations, which together result in premature death. T cell metabolic failure induces the accumulation of circulating cytokines, which resembles the chronic inflammation that is characteristic of aging ("inflammaging"). This cytokine storm itself acts as a systemic inducer of senescence. Blocking tumor necrosis factor-α signaling or preventing senescence with nicotinamide adenine dinucleotide precursors partially rescues premature aging in mice with Tfam-deficient T cells. Thus, T cells can regulate organismal fitness and life span, which highlights the importance of tight immunometabolic control in both aging and the onset of age-associated diseases.


Subject(s)
Aging, Premature/immunology , DNA-Binding Proteins/deficiency , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Multimorbidity , T-Lymphocytes/metabolism , Transcription Factors/deficiency , Aging, Premature/genetics , Aging, Premature/prevention & control , Animals , Cytokine Release Syndrome/immunology , DNA-Binding Proteins/genetics , Female , Gene Deletion , Inflammation/genetics , Inflammation/immunology , Longevity , Male , Mice , Mice, Mutant Strains , Mitochondrial Proteins/genetics , NAD/administration & dosage , NAD/pharmacology , Physical Fitness , T-Lymphocytes/ultrastructure , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors
8.
FEBS J ; 287(16): 3350-3369, 2020 08.
Article in English | MEDLINE | ID: mdl-32255251

ABSTRACT

The inflammatory response involves the activation of several cell types to fight insults caused by a plethora of agents, and to maintain the tissue homoeostasis. On the one hand, cells involved in the pro-inflammatory response, such as inflammatory M1 macrophages, Th1 and Th17 lymphocytes or activated microglia, must rapidly provide energy to fuel inflammation, which is essentially accomplished by glycolysis and high lactate production. On the other hand, regulatory T cells or M2 macrophages, which are involved in immune regulation and resolution of inflammation, preferentially use fatty acid oxidation through the TCA cycle as a main source for energy production. Here, we discuss the impact of glycolytic metabolism at the different steps of the inflammatory response. Finally, we review a wide variety of molecular mechanisms which could explain the relationship between glycolytic metabolites and the pro-inflammatory phenotype, including signalling events, epigenetic remodelling, post-transcriptional regulation and post-translational modifications. Inflammatory processes are a common feature of many age-associated diseases, such as cardiovascular and neurodegenerative disorders. The finding that immunometabolism could be a master regulator of inflammation broadens the avenue for treating inflammation-related pathologies through the manipulation of the vascular and immune cell metabolism.


Subject(s)
Citric Acid Cycle/immunology , Glycolysis/immunology , Inflammation/immunology , Macrophage Activation/immunology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Inflammation/metabolism , Macrophages/classification , Macrophages/metabolism , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism
9.
Nat Med ; 23(8): 964-974, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28692064

ABSTRACT

Polo-like kinase 1 (PLK1), an essential regulator of cell division, is currently undergoing clinical evaluation as a target for cancer therapy. We report an unexpected function of Plk1 in sustaining cardiovascular homeostasis. Plk1 haploinsufficiency in mice did not induce obvious cell proliferation defects but did result in arterial structural alterations, which frequently led to aortic rupture and death. Specific ablation of Plk1 in vascular smooth muscle cells (VSMCs) led to reduced arterial elasticity, hypotension, and an impaired arterial response to angiotensin II in vivo. Mechanistically, we found that Plk1 regulated angiotensin II-dependent activation of RhoA and actomyosin dynamics in VSMCs in a mitosis-independent manner. This regulation depended on Plk1 kinase activity, and the administration of small-molecule Plk1 inhibitors to angiotensin II-treated mice led to reduced arterial fitness and an elevated risk of aneurysm and aortic rupture. We thus conclude that a partial reduction of Plk1 activity that does not block cell division can nevertheless impair aortic homeostasis. Our findings have potentially important implications for current approaches aimed at PLK1 inhibition for cancer therapy.


Subject(s)
Angiotensin II/metabolism , Aortic Aneurysm/genetics , Aortic Rupture/genetics , Cell Cycle Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , rho GTP-Binding Proteins/metabolism , Animals , Aorta/metabolism , Aorta/ultrastructure , Aortic Aneurysm/metabolism , Aortic Rupture/metabolism , Blood Pressure , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Proliferation/genetics , Fluorescent Antibody Technique , Gene Knockdown Techniques , Haploinsufficiency , Homeostasis/genetics , Hypotension/genetics , Immunoblotting , Mice , Microscopy, Electron, Transmission , Mitosis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Real-Time Polymerase Chain Reaction , Vascular Stiffness/genetics , rhoA GTP-Binding Protein , Polo-Like Kinase 1
10.
Nat Med ; 23(2): 200-212, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28067899

ABSTRACT

Heritable thoracic aortic aneurysms and dissections (TAAD), including Marfan syndrome (MFS), currently lack a cure, and causative mutations have been identified for only a fraction of affected families. Here we identify the metalloproteinase ADAMTS1 and inducible nitric oxide synthase (NOS2) as therapeutic targets in individuals with TAAD. We show that Adamts1 is a major mediator of vascular homeostasis, given that genetic haploinsufficiency of Adamts1 in mice causes TAAD similar to MFS. Aortic nitric oxide and Nos2 levels were higher in Adamts1-deficient mice and in a mouse model of MFS (hereafter referred to as MFS mice), and Nos2 inactivation protected both types of mice from aortic pathology. Pharmacological inhibition of Nos2 rapidly reversed aortic dilation and medial degeneration in young Adamts1-deficient mice and in young or old MFS mice. Patients with MFS showed elevated NOS2 and decreased ADAMTS1 protein levels in the aorta. These findings uncover a possible causative role for the ADAMTS1-NOS2 axis in human TAAD and warrant evaluation of NOS2 inhibitors for therapy.


Subject(s)
ADAMTS1 Protein/genetics , Aorta/metabolism , Aortic Aneurysm/genetics , Aortic Dissection/genetics , Marfan Syndrome/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide/metabolism , ADAMTS1 Protein/metabolism , Adult , Aged , Aortic Dissection/metabolism , Animals , Aorta/drug effects , Aortic Aneurysm/metabolism , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Fibrillin-1/genetics , Gene Knockdown Techniques , Haploinsufficiency , Humans , Immunoblotting , Male , Marfan Syndrome/metabolism , Mice , Middle Aged , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Real-Time Polymerase Chain Reaction
11.
Mol Cell Biol ; 35(19): 3409-22, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26217013

ABSTRACT

Emerging evidence indicates that the metalloproteinase Adamts-1 plays a significant role in the pathophysiology of vessel remodeling, but little is known about the signaling pathways that control Adamts-1 expression. We show that vascular endothelial growth factor (VEGF), angiotensin-II, interleukin-1ß, and tumor necrosis factor α, stimuli implicated in pathological vascular remodeling, increase Adamts-1 expression in endothelial and vascular smooth muscle cells. Analysis of the intracellular signaling pathways implicated in this process revealed that VEGF and angiotensin-II upregulate Adamts-1 expression via activation of differential signaling pathways that ultimately promote functional binding of the NFAT or C/EBPß transcription factors, respectively, to the Adamts-1 promoter. Infusion of mice with angiotensin-II triggered phosphorylation and nuclear translocation of C/EBPß proteins in aortic cells concomitantly with an increase in the expression of Adamts-1, further underscoring the importance of C/EBPß signaling in angiotensin-II-induced upregulation of Adamts-1. Similarly, VEGF promoted NFAT activation and subsequent Adamts-1 induction in aortic wall in a calcineurin-dependent manner. Our results demonstrate that Adamts-1 upregulation by inducers of pathological vascular remodeling is mediated by specific signal transduction pathways involving NFAT or C/EBPß transcription factors. Targeting of these pathways may prove useful in the treatment of vascular disease.


Subject(s)
ADAM Proteins/metabolism , CCAAT-Enhancer-Binding Protein-beta/physiology , NFATC Transcription Factors/metabolism , Vascular Remodeling , ADAM Proteins/genetics , ADAMTS1 Protein , Animals , Aorta/enzymology , Base Sequence , Calcineurin/metabolism , Cells, Cultured , Human Umbilical Vein Endothelial Cells/physiology , Humans , Mice, Knockout , Molecular Sequence Data , Signal Transduction , Transcriptional Activation , Vascular Endothelial Growth Factor A/physiology
12.
Arterioscler Thromb Vasc Biol ; 34(10): 2310-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25147342

ABSTRACT

OBJECTIVE: Vascular endothelial growth factor (VEGF) has been identified as a crucial regulator of physiological and pathological angiogenesis. Among the intracellular signaling pathways triggered by VEGF, activation of the calcineurin/nuclear factor of activated T cells (NFAT) signaling axis has emerged as a critical mediator of angiogenic processes. We and others previously reported a novel role for the plasma membrane calcium ATPase (PMCA) as an endogenous inhibitor of the calcineurin/NFAT pathway, via interaction with calcineurin, in cardiomyocytes and breast cancer cells. However, the functional significance of the PMCA/calcineurin interaction in endothelial pathophysiology has not been addressed thus far. APPROACH AND RESULTS: Using in vitro and in vivo assays, we here demonstrate that the interaction between PMCA4 and calcineurin in VEGF-stimulated endothelial cells leads to downregulation of the calcineurin/NFAT pathway and to a significant reduction in the subsequent expression of the NFAT-dependent, VEGF-activated, proangiogenic genes RCAN1.4 and Cox-2. PMCA4-dependent inhibition of calcineurin signaling translates into a reduction in endothelial cell motility and blood vessel formation that ultimately impairs in vivo angiogenesis by VEGF. CONCLUSIONS: Given the importance of the calcineurin/NFAT pathway in the regulation of pathological angiogenesis, targeted modulation of PMCA4 functionality might open novel therapeutic avenues to promote or attenuate new vessel formation in diseases that occur with angiogenesis.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Calcineurin/metabolism , Calcium-Transporting ATPases/metabolism , Endothelial Cells/drug effects , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Plasma Membrane Calcium-Transporting ATPases/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , Calcium-Binding Proteins , Calcium-Transporting ATPases/deficiency , Calcium-Transporting ATPases/genetics , Cell Movement , Cell Proliferation , Cyclooxygenase 2/metabolism , DNA-Binding Proteins , Disease Models, Animal , Endothelial Cells/enzymology , HEK293 Cells , Hindlimb , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ischemia/enzymology , Ischemia/physiopathology , Mice , Mice, Knockout , Muscle Proteins/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , RNA Interference , Signal Transduction , Time Factors , Transfection
13.
La Paz; 1985. 154 p. ilus.
Thesis in Spanish | LIBOCS, LIBOSP | ID: biblio-1311341

ABSTRACT

Contenido: Cap.1 Introduccion. Cap.2 Estudio de mercado.Generalidades.Estructura del mercado nacional.Especificaciones del producto.Estudio de la demanda.Oferta.Participacion del producto en la demanda futura.Balance demanda-oferta.Canales de distribucion. Cap.3 Tamano y localizacion. Cap.4 Ingenieria del proyecto.Generalidades.Procedimiento experimental.Produccion.Programa de produccion.Requerimiento de materia prima.Caracteristicas de la materia prima.Objetivos del proceso.Descripcion del proceso de produccion.Etapa de tratamiento quimico.Etapa de tratamiento mecanico.Evaluacion tecnica preliminar de la planta. Cap.5 Inversiones del proyecto. Estructura de inversiones. Cap.6 Financiamiento.Inversion requerida.Fuentes de financiamiento. Cap.7 Estimacion de costos e ingresos.Estimacion de costos.Estimacion de ingresos. Cap.8 Evaluacion del proyecto.Introduccion.Utilidades proyectadas.Determinacion del punto de equilibrio.Indice de rentabilidad.Indice de seguridad de actividad.Indice de seguridad estructural.Indice de seguridad proporcional.Rentabilidad en relacion a la inversion.Fuentes y usos de fondos.Plazo de amortizacion de la inversion.Analisis de sensibilidad. Cap.9 Conclusiones y recomendaciones. Anexos.

SELECTION OF CITATIONS
SEARCH DETAIL
...