Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(9): e0263021, 2023.
Article in English | MEDLINE | ID: mdl-37751438

ABSTRACT

Disease is a neurodegenerative disorder characterised by the progressive loss of dopaminergic cells of the substantia nigra pars compacta. Even though successful transplantation of dopamine-producing cells into the striatum exhibits favourable effects in animal models and clinical trials; transplanted cell survival is low. Since every transplant elicits an inflammatory response which can affect cell survival and differentiation, we aimed to study in vivo and in vitro the impact of the pro-inflammatory environment on human dopaminergic precursors. We first observed that transplanted human dopaminergic precursors into the striatum of immunosuppressed rats elicited an early and sustained activation of astroglial and microglial cells after 15 days' post-transplant. This long-lasting response was associated with Tumour necrosis factor alpha expression in microglial cells. In vitro, conditioned media from activated BV2 microglial cells increased cell death, decreased Tyrosine hydroxylase-positive cells and induced morphological alterations on human neural stem cells-derived dopaminergic precursors at two differentiation stages: 19 days and 28 days. Those effects were ameliorated by inhibition of Tumour necrosis factor alpha, a cytokine which was previously detected in vivo and in conditioned media from activated BV-2 cells. Our results suggest that a pro-inflammatory environment is sustained after transplantation under immunosuppression, providing a window of opportunity to modify this response to increase transplant survival and differentiation. In addition, our data show that the microglia-derived pro-inflammatory microenvironment has a negative impact on survival and differentiation of dopaminergic precursors. Finally, Tumour necrosis factor alpha plays a key role in these effects, suggesting that this cytokine could be an interesting target to increase the efficacy of human dopaminergic precursors transplantation in Parkinson's Disease.


Subject(s)
Microglia , Tumor Necrosis Factor-alpha , Humans , Animals , Rats , Tumor Necrosis Factor-alpha/pharmacology , Culture Media, Conditioned/pharmacology , Dopamine , Cell Differentiation , Cytokines
2.
Fly (Austin) ; 17(1): 2192457, 2023 12.
Article in English | MEDLINE | ID: mdl-36949021

ABSTRACT

In Drosophila melanogaster, several Gal4 drivers are used to direct gene/RNAi expression to different dopaminergic neuronal clusters. We previously developed a fly model of Parkinson's disease, in which dopaminergic neurons had elevated cytosolic Ca2+ due to the expression of a Plasma Membrane Ca2+ ATPase (PMCA) RNAi under the thyroxine hydroxylase (TH)-Gal4 driver. Surprisingly, TH-Gal4>PMCARNAi flies died earlier compared to controls and showed swelling in the abdominal area. Flies expressing the PMCARNAi under other TH drivers also showed such swelling and shorter lifespan. Considering that TH-Gal4 is also expressed in the gut, we proposed to suppress the expression specifically in the nervous system, while maintaining the activation in the gut. Therefore, we expressed Gal80 under the direction of the panneuronal synaptobrevin (nSyb) promoter in the context of TH-Gal4. nSyb-Gal80; TH-Gal4>PMCARNAi flies showed the same reduction of survival as TH-Gal4>PMCARNAi flies, meaning that the phenotype of abdomen swelling and reduced survival could be due to the expression of the PMCARNAi in the gut. In perimortem stages TH-Gal4>PMCARNAi guts had alteration in the proventriculi and crops. The proventriculi appeared to lose cells and collapse on itself, and the crop increased its size several times with the appearance of cellular accumulations at its entrance. No altered expression or phenotype was observed in flies expressing PMCARNAi in the dopaminergic PAM cluster (PAM-Gal4>PMCARNAi). In this work we show the importance of checking the global expression of each promoter and the relevance of the inhibition of PMCA expression in the gut.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Transcription Factors , Tyrosine 3-Monooxygenase , Animals , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Membrane/metabolism , Dopaminergic Neurons/metabolism , Down-Regulation , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Longevity/genetics , Transcription Factors/genetics , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
3.
Mult Scler Relat Disord ; 57: 103346, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35158455

ABSTRACT

BACKGROUND: Inflammation in the Central Nervous System (CNS) is associated with blood brain barrier (BBB) breakdown during the early stages of Multiple Sclerosis (MS), indicating a facilitated entry of waves of inflammatory cells from the circulation to the CNS. In the progressive forms of MS, as the lesion becomes chronic, the inflammation remains trapped within the CNS compartment forming the slow evolving lesion, characterized by low inflammation and microglia activation at the lesions edges. The chronic expression of interleukin 1ß (IL-1ß) in the cortex induces BBB breakdown, demyelination, neurodegeneration, microglial/macrophage activation and impaired cognitive performance. The latter can be improved, as long as the BBB recovers and the lesion presents low inflammation. Here, we study the effects of peripheral inflammation on cortical central lesions after the restoration of the BBB, in order to elucidate the role of the peripheral inflammation on these lesions with intact BBB, as it occurs in the progressive forms of MS. MATERIALS AND METHODS: Cortical lesions and peripheral inflammation were induced by the chronic expression of IL-1ß using an adenovector. We performed histological, immunohistochemistry on brain tissue and behavioural analyses. RESULTS: The effects of the chronic expression of IL-1ß in the cortex resolved within 56 days. However, peripheral and sustained inflammation re-opened the BBB, allowing the reappearance of the neuroinflammatory processes within the cortical lesions, increased demyelination and neurodegeneration, and an increase of the behavioral symptoms, such as cognitive impairment and anxiety-like symptoms. CONCLUSIONS: The early treatment of peripheral inflammatory processes should be considered in order to protect the brain from exacerbation of the ongoing neurodegenerative process.


Subject(s)
Blood-Brain Barrier , Multiple Sclerosis , Brain , Central Nervous System , Humans , Inflammation
4.
Eur J Neurosci ; 54(6): 5915-5931, 2021 09.
Article in English | MEDLINE | ID: mdl-34312939

ABSTRACT

The accumulation of Ca2+ and its subsequent increase in oxidative stress is proposed to be involved in selective dysfunctionality of dopaminergic neurons, the main cell type affected in Parkinson's disease. To test the in vivo impact of Ca2+ increment in dopaminergic neurons physiology, we downregulated the plasma membrane Ca2+ ATPase (PMCA), a pump that extrudes cytosolic Ca2+ , by expressing PMCARNAi in Drosophila melanogaster dopaminergic neurons. In these animals, we observed major locomotor alterations paralleled to higher cytosolic Ca2+ and increased levels of oxidative stress in mitochondria. Interestingly, although no overt degeneration of dopaminergic neurons was observed, evidences of neuronal dysfunctionality were detected such as increases in presynaptic vesicles in dopaminergic neurons and in the levels of dopamine in the brain, as well as presence of toxic effects when PMCA was downregulated in the eye. Moreover, reduced PMCA levels were found in a Drosophila model of Parkinson's disease, Parkin knock-out, expanding the functional relevance of PMCA reduction to other Parkinson's disease-related models. In all, we have generated a new model to study motor abnormalities caused by increments in Ca2+ that lead to augmented oxidative stress in a dopaminergic environment, added to a rise in synaptic vesicles and dopamine levels.


Subject(s)
Parkinson Disease , Plasma Membrane Calcium-Transporting ATPases , Animals , Calcium/metabolism , Dopaminergic Neurons/metabolism , Down-Regulation , Drosophila melanogaster , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasma Membrane Calcium-Transporting ATPases/metabolism
5.
Brain Res ; 1727: 146520, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31669283

ABSTRACT

Multiple Sclerosis (MS) is a neuroinflammatory disease affecting white and grey matter, it is characterized by demyelination, axonal degeneration along with loss of motor, sensitive and cognitive functions. MS is a heterogeneous disease that displays different clinical courses: relapsing/remitting MS (RRMS), and MS progressive forms: primary progressive (PPMS) and secondary progressive (SPMS). Cortical damage in the progressive MS forms has considerable clinical relevance due to its association with cognitive impairment and disability progression in patients. One treatment is available for the progressive forms of the disease, but none are specific for cognitive deficits. We developed an animal model that reflects most of the characteristics of the cortical damage, such as cortical neuroinflammation, demyelination, neurodegeneration and meningeal inflammation, which was associated with cognitive impairment. Cognitive rehabilitation, exercise and social support have begun to be evaluated in patients and animal models of neurodegenerative diseases. Environmental enrichment (EE) provides exercise as well as cognitive and social stimulation. EE has been demonstrated to exert positive effects on cognitive domains, such as learning and memory, and improving anxiety-like symptoms. We proposed to study the effect of EE on peripherally stimulated cortical lesion induced by the long term expression of interleukin IL-1ß (IL-1ß) in adult rats. Here, we demonstrated that EE: 1) reduces the peripheral inflammatory response to the stimulus, 2) ameliorates cognitive deficits and anxiety-like symptoms, 3) modulates neurodegeneration, demyelination and glial activation, 4) regulates neuroinflammation by reducing the expression of pro-inflammatory cytokines and enhancing the expression of anti-inflammatory ones. Our findings correlate with the fact that EE housing could be considered an effective non- pharmacological therapeutic agent that can synergistically aid in the rehabilitation of the disease.


Subject(s)
Cognitive Dysfunction/rehabilitation , Multiple Sclerosis/psychology , Multiple Sclerosis/rehabilitation , Social Interaction , Social Support , Animals , Cognition , Gray Matter/metabolism , Gray Matter/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Male , Physical Conditioning, Animal , Rats , Rats, Wistar
6.
Brain Behav Immun ; 69: 515-531, 2018 03.
Article in English | MEDLINE | ID: mdl-29378262

ABSTRACT

Multiple sclerosis (MS) is an inflammatory and demyelinating disease of unknown aetiology that causes neurological disabilities in young adults. MS displays different clinical patterns, including recurrent episodes with remission periods ("relapsing-remitting MS" (RRMS)), which can progress over several years to a secondary progressive form (SPMS). However, 10% of patients display persistent progression at the onset of disease ("primary progressive MS" (PPMS)). Currently, no specific therapeutic agents are available for the progressive forms, mainly because the underlying pathogenic mechanisms are not clear and because no animal models have been specifically developed for these forms. The development of MS animal models is required to clarify the pathological mechanisms and to test novel therapeutic agents. In the present work, we overexpressed interleukin 1 beta (IL-1ß) in the cortex to develop an animal model reflecting the main pathological hallmarks of MS. The treated animals presented with neuroinflammation, demyelination, glial activation, and neurodegeneration along with cognitive symptoms and MRI images consistent with MS pathology. We also demonstrated the presence of meningeal inflammation close to cortical lesions, with characteristics similar to those described in MS patients. Systemic pro-inflammatory stimulation caused a flare-up of the cortical lesions and behavioural symptoms, including impairment of working memory and the appearance of anxiety-like symptoms. Our work demonstrated induced cortical lesions, reflecting the main histopathological hallmarks and cognitive impairments characterizing the cortical pathology described in MS patients with progressive forms of the disease.


Subject(s)
Cerebral Cortex/pathology , Immunity, Innate/physiology , Inflammation/pathology , Multiple Sclerosis, Chronic Progressive/pathology , Animals , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/immunology , Disease Models, Animal , Disease Progression , Inflammation/diagnostic imaging , Inflammation/immunology , Magnetic Resonance Imaging , Male , Motor Activity/physiology , Multiple Sclerosis, Chronic Progressive/diagnostic imaging , Multiple Sclerosis, Chronic Progressive/immunology , Rats , Rats, Wistar
7.
Brain Res ; 1638(Pt A): 15-29, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26239914

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder, whose cardinal pathology is the loss of dopaminergic neurons in the substantia nigra. Current treatments for PD have side effects in the long term and do not halt disease progression or regenerate dopaminergic cell loss. Attempts to compensate neuronal cell loss by transplantation of dopamine-producing cells started more than 30 years ago, leading to several clinical trials. These trials showed safety and variable efficacy among patients. In addition to variability in efficacy, several patients developed graft-induced dyskinesia. Nevertheless, they have provided a proof of concept that motor symptoms could be improved by cell transplantation. Cell transplantation in the brain presents several immunological challenges. The adaptive immune response should be abolished to avoid graft rejection by the host. In addition, the innate immune response will always be present after transplanting cells into the brain. Remarkably, the innate immune response can have dramatic effects on the survival, differentiation and proliferation of the transplanted cells, but has been hardly investigated. In this review, we analyze data on the functional effects of signals from the innate immune system on dopaminergic differentiation, survival and proliferation. Then, we discussed efforts on cell transplantation in animal models and PD patients, highlighting the immune response and the immunomodulatory treatment strategies performed. The analysis of the available data lead us to conclude that the modulation of the innate immune response after transplantation can increase the success of future clinical trials in PD by enhancing cell differentiation and survival. This article is part of a Special Issue entitled SI: PSC and the brain.


Subject(s)
Dopaminergic Neurons/pathology , Dopaminergic Neurons/physiology , Neural Stem Cells/immunology , Neural Stem Cells/transplantation , Parkinson Disease/immunology , Parkinson Disease/therapy , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Embryonic Stem Cells/transplantation , Humans , Neural Stem Cells/pathology , Parkinson Disease/pathology , Pluripotent Stem Cells/physiology , Pluripotent Stem Cells/transplantation , Stem Cell Transplantation/methods
9.
Metallomics ; 7(10): 1381-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26360295

ABSTRACT

Previous studies by our group demonstrated the key role of iron in Schwann cell maturation through an increase in cAMP, PKA activation and CREB phosphorylation. These studies opened the door to further research on non-transferrin-bound iron uptake, which revealed the presence of DMT1 mRNA all along SC progeny, hinting at a constitutive role of DMT1 in ensuring the provision of iron in the PNS. In light of these previous results, the present work evaluates the participation of DMT1 in the remyelination process following a demyelinating lesion promoted by sciatic nerve crush--a reversible model of Wallerian degeneration. DMT1 was observed to colocalize with a SC marker S100ß at all survival times analyzed. In turn, the assessment of DMT1 mRNA expression exhibited an increase 7 days post-injury, while DMT1 protein levels showed an increase 14 days after crush at the lesion site and distal stump; finally, an increase in iron levels became evident as from 14 days post-injury, in parallel with DMT1 values. To sum up, the present work unveils the role of DMT1 in mediating the neuroregenerative action of iron.


Subject(s)
Cation Transport Proteins/metabolism , Iron/metabolism , Peripheral Nervous System/metabolism , Animals , Blotting, Western , Cation Transport Proteins/genetics , Myelin Sheath/metabolism , Peripheral Nervous System/pathology , Rats , Rats, Wistar , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology
10.
Mol Cell Neurosci ; 67: 75-83, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26051800

ABSTRACT

Transforming growth factor beta 1 (TGF-beta1), an anti-inflammatory cytokine, has been shown to have pro-neurogenic effects on adult Neural Stem Cells (aNSC) from the dentate gyrus and in vivo models. Here, we expanded the observation of the pro-neurogenic effect of TGF-beta1 on aNSC from the subventricular zone (SVZ) of adult rats and performed a functional genomic analysis to identify candidate genes to mediate its effect. 10 candidate genes were identified by microarray analysis and further validated by qRT-PCR. Of these, Fibulin-2 was increased 477-fold and its inhibition by siRNA blocks TGF-beta1 pro-neurogenic effect. Curiously, Fibulin-2 was not expressed by aNSC but by a GFAP-positive population in the culture, suggesting an indirect mechanism of action. TGF-beta1 also induced Fibulin-2 in the SVZ in vivo. Interestingly, 5 out of the 10 candidate genes identified are known to interact with integrins, paving the way for exploring their functional role in adult neurogenesis. In conclusion, we have identified 10 genes with putative pro-neurogenic effects, 5 of them related to integrins and provided proof that Fibulin-2 is a major mediator of the pro-neurogenic effects of TGF-beta1. These data should contribute to further exploring the molecular mechanism of adult neurogenesis of the genes identified and the involvement of the integrin pathway on adult neurogenesis.


Subject(s)
Adult Stem Cells/metabolism , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Transforming Growth Factor beta1/pharmacology , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Animals , Astrocytes/metabolism , Calcium-Binding Proteins/genetics , Cells, Cultured , Extracellular Matrix Proteins/genetics , Integrins/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/growth & development , Lateral Ventricles/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Rats , Rats, Wistar , Transforming Growth Factor beta1/metabolism
11.
Neurochem Int ; 62(2): 145-56, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23231993

ABSTRACT

The white matter (WM) represents approximately half the cerebrum volume and is profoundly affected in Alzheimer's disease (AD). However, both the WM responses to AD as well as potential influences of this compartment to dementia pathogenesis remain comparatively neglected. Neuroimaging studies have revealed WM alterations are commonly associated with AD and renewed interest in examining the pathologic basis and importance of these changes. In AD subjects, immunohistochemistry and electron microscopy revealed changes in astrocyte morphology and myelin loss as well as up to 30% axonal loss in areas of WM rarefaction when measured against non-demented control (NDC) tissue. Comparative proteomic analyses were performed on pooled samples of periventricular WM (PVWM) obtained from AD (n=4) and NDC (n=5) subjects with both groups having a mean age of death of 86 years. All subjects had an apolipoprotein E ε3/3 genotype with the exception of one NDC subject who was ε2/3. Urea-detergent homogenates were analyzed using two different separation techniques: 2-dimensional isoelectric focusing/reverse-phase chromatography and 2-dimensional difference gel electrophoresis (2D-DIGE). Proteins with different expression levels between the 2 diagnostic groups were identified using MALDI-Tof/Tof mass spectrometry. In addition, Western blots were used to quantify proteins of interest in individual AD and NDC cases. Our proteomic studies revealed that when WM protein pools were loaded at equal amounts of total protein for comparative analyses, there were quantitative differences between the 2 groups. Molecules related to cytoskeleton maintenance, calcium metabolism and cellular survival such as glial fibrillary acidic protein, vimentin, tropomyosin, collapsin response mediator protein-2, calmodulin, S100-P, annexin A1, α-internexin, α- and ß-synuclein, α-B-crystalline, fascin-1, ubiquitin carboxyl-terminal esterase and thymosine were altered between AD and NDC pools. Our experiments suggest that WM activities become globally impaired during the course of AD with significant morphological, biochemical and functional consequential implications for gray matter function and cognitive deficits. These observations may endorse the hypothesis that WM dysfunction is not only a consequence of AD pathology, but that it may precipitate and/or potentiate AD dementia.


Subject(s)
Alzheimer Disease/pathology , Proteomics , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Blotting, Western , Chromatography, Liquid , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunohistochemistry , Male , Plaque, Amyloid
12.
J Neurochem ; 121(6): 985-95, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22443207

ABSTRACT

The extension of processes of oligodendrocyte (OLG) and their precursor cells are crucial for migration, axonal contact and myelination. Here we show that a non-lethal oxidative stress induced by 3-nitropropionic acid (3-NP) elicited a rapid shortening of processes (~24%) in primary OLGs and in oligodendroglial cell line (OLN-93) cells (~36%) as compared with vehicle-exposed cells. This was reversible and prevented by antioxidants. Proteomics of OLG lysates with and without 3-NP treatment yielded collapsin response mediator protein 2 (CRMP-2) as a candidate effector molecule. Inhibition of rho kinase was sufficient to prevent process retraction in both OLGs and OLN-93 cells. Oxidative stress increased phosphorylation of CRMP-2 at T555 that was completely prevented by Y27632. Moreover, transfection of OLN-93 cells with the mutant CRMP-2 T555A which cannot be phosphorylated by rho kinase, prevented process shortening induced by 3-NP as compared with wild-type CRMP-2. Our results suggest a role for endogenous reactive oxygen species in a pathway that regulates OLG process extension. The vulnerability of late myelinated neurons in the adult brain and the presence of white matter pathology in human dementias warrant the study of this oligodendroglial pathway in the early stages of neurodegenerative conditions characterized by oxidative stress.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oxidative Stress/physiology , Animals , Antioxidants/pharmacology , Blotting, Western , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mutagenesis, Site-Directed , Oligodendroglia/drug effects , Phosphorylation , Polymerase Chain Reaction , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/toxicity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection
13.
Medicina (B Aires) ; 69(4): 466-72, 2009.
Article in Spanish | MEDLINE | ID: mdl-19770104

ABSTRACT

The global increase in life expectancy turns Alzheimer's disease (AD) into a growing problem. One of the distinctive features of AD is the excessive accumulation of amyloid-b (Ab) peptide in the brain. In recent years, a concept that has gained strength is that degradation of Ab by proteases in situ is an important mechanism that prevents cerebral peptide accumulation. Biochemical and genetic data have shown that insulin-degrading enzyme (IDE) participates in Ab and insulin homeostasis. IDE expression and activity are significantly decreased in AD brains compared to age-matched controls. Also, IDE is deposited with Ab in senile plaques and blood vessels, indicating a gross conformational change as a consequence of diverse post-translational mechanisms. These alterations in IDE distribution and activity may result in insufficient degradation of Ab and insulin, promoting the formation of Ab oligomers and hormone resistance. Both processes might play a fundamental part in neurodegeneration. The study of the clearance mechanisms of cerebral Ab will not only aid in the understanding AD pathogenesis but will also allow a better interpretation of ongoing clinical trials and the development of new therapeutic strategies.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Insulysin/metabolism , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Humans , Hydrolysis
14.
Subcell Biochem ; 38: 129-45, 2005.
Article in English | MEDLINE | ID: mdl-15709476

ABSTRACT

Amyloid beta (Abeta) accumulates in the neuropil and within the walls of cerebral vessels in association with normal aging, dementia or stroke. Abeta is released from its precursor protein as soluble monomeric species yet, under pathological conditions, it self-aggregates to form soluble oligomers or insoluble fibrils that may be toxic to neurons and vascular cells. Abeta levels could be lowered by inhibiting its generation or by promoting its clearance by transport or degradation. Here we will summarize recent findings on brain proteases capable of degrading Abeta, with a special focus on those enzymes for which there is genetic, transgenic or biochemical evidence supporting a role in the proteolysis of Abeta in vivo.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Brain/enzymology , Peptide Hydrolases/metabolism , Aspartic Acid Endopeptidases/metabolism , Endothelin-Converting Enzymes , Humans , Insulysin/metabolism , Metalloendopeptidases/metabolism , Neprilysin/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...