SciELO - Scientific Electronic Library Online

 
vol.12Quimotripsina en crustáceos: estado del arteColección, vitrificación y transferencia post-calentamiento de embriones equinos producidos in vivo: una revisión de literatura índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • No hay artículos similaresSimilares en SciELO

Compartir


Abanico veterinario

versión On-line ISSN 2448-6132versión impresa ISSN 2007-428X

Abanico vet vol.12  Tepic ene./dic. 2022  Epub 31-Oct-2022

https://doi.org/10.21929/abavet2022.11 

Literature review

Systemic AA amyloidosis: a potential public health problem

1Departamento de Ciencias Veterinarias, Centro de Ciencias Agropecuarias, Universidad Autónoma de Aguascalientes, Av. Universidad #940, Ciudad Universitaria, C. P. 20131. Aguascalientes, Aguascalientes México. E-mail: rmelende@correo.uaa.mx


ABSTRACT

The objective of this review is to inform animal health professionals about systemic AA amyloidosis, a little known potential public health problem in Mexico, which can result from the consumption of animal products. It is difficult to diagnose, since clinical signs depend on the type and location of the amyloid; and its terminal phase corresponds to renal failure. It consists of the accumulation of an extracellular protein material that can originate from about 37 normal proteins in humans and more than 15 in other aquatic and terrestrial mammals and in birds. It has a peculiar fibrillar structure in a β-folded arrangement that confers great resistance to physicochemical agents, which makes difficult its elimination from food. Some amyloidosis, such as Alzheimer's disease in humans and cognitive dysfunction disorder in dogs, are localized, but AA amyloidosis is systemic and there is natural and experimental evidence that it can be transmitted intra- and interspecies when there is oral, intravenous or intraperitoneal entry of amyloid material in an individual with pre-existing chronic inflammatory disease or other risk factors, such as obesity, making it a potential public health problem.

Keywords: Public health; food safety; amyloidosis; chronic inflamation; renal insufficiency

RESUMEN

El objetivo de esta revisión es informar a los profesionales en salud animal, sobre la amiloidosis sistémica AA, un problema potencial de salud pública poco conocido en México, que puede derivarse del consumo de productos animales. Es difícil de diagnosticar, pues los signos clínicos dependen del tipo y localización del amiloide; y su fase terminal corresponde con un cuadro de insuficiencia renal. Consiste en la acumulación de un material proteico extracelular que puede originarse a partir de alrededor de 37 proteínas normales en los humanos y más de 15 en otros mamíferos acuáticos y terrestres, y en aves. Posee una estructura fibrilar característica en disposición β plegada que le confiere gran resistencia a agentes fisicoquímicos, que hace difícil su eliminación de los alimentos. Algunas amiloidosis, como el Alzheimer en humanos y el trastorno de disfunción cognitiva en los perros, son localizadas, pero la amiloidosis AA es sistémica y existe evidencia natural y experimental de que puede transmitirse intra e inter-especie cuando hay ingreso por vía oral, intravenosa o intraperitoneal de material amiloide en un individuo con una enfermedad inflamatoria crónica pre-existente u otros factores de riesgo, como la obesidad, lo que la convierte en un potencial problema de salud pública.

Palabras clave: Salud pública; inocuidad alimentaria; amiloidosis; inflamación crónica; insuficiencia renal

INTRODUCTION

Proteins, in addition to building tissues, are essential for cell metabolism and communication; their three-dimensional structure depends on how the amino acid sequence that constitutes their primary structure is folded. The structure governs the function and that is why some have certain flexibility that allows them to adjust to the required task; that makes it necessary to have mechanisms that ensure the production, folding and elimination (if applicable), of defective proteins (Jucker & Walker, 2013).

Several proteins that are soluble under physiological conditions may undergo changes or conformational changes towards a molecular structure in a β-folded arrangement and subsequently self-assemble into extremely insoluble fibers (Sipe et al., 2012). There are many diseases that arise from protein misfolding. Amyloidosis is one of them and comprises a wide range of very different human and animal disorders that have in common the infiltration of one or several tissues with an extracellular protein, amorphous accumulation with a characteristic appearance under electron microscopy; a typical X-ray diffraction pattern, affinity for Congo red staining and the production of green or yellow birefringence, under polarized light (Jucker & Walker, 2013; Sipe et al., 2016; Benson et al., 2018); although "amyloid-like" proteins are now known to be associated with important biological functions ranging from long-term memory, formation of hormone secretion granules (Maji et al., 2009; Rubel et al., 2020), membrane changes in the egg and sperm that enable fertilization and others not only in mammals; but also in plants and prokaryotic cells (Rubel et al., 2020).

The word amyloid was coined by Rudolph Virchow in 1854, after he described it as a starch-like material (because it stains with iodine), which infiltrated the liver, kidney and heart and was often found in post-mortem examinations (Gruys, 2004). Subsequently, the protein nature of the infiltrate was discovered and electron microscopy revealed its fibrillar structure.

In 1984, Glenner and Wong found the same type of fibers in the plaques of human brains with Alzheimer's disease and of hamsters experimentally infected with Scrapie (Gruys, 2004).

To date, 37 amyloidogenic proteins have been associated with approximately 70 human diseases. The best known include Alzheimer's disease, Parkinson's disease, type 2 diabetes and spongiform encephalopathies (Rubel et al., 2020). Also with some forms of cancer, such as multiple myeloma and plasmacytoma, in both humans and animals (Andrei & Wang, 2019, Kadota et al., 2020, Tamura et al., 2020).

Clinical data indicate that the proteins that constitute amyloid are different, since the deposits have been found in very different pathological contexts and have therefore been classified according to two criteria, their distribution and the precursor protein (Rubel et al., 2020). Despite their diverse origins, all amyloid deposits have a common basic structure, which also shares many cofactors; such as the P component of serum amyloid, apolipoprotein E and heparan sulfate-type proteoglycans (Martin et al., 2010; Benson et al., 2018). This isomorphism in such a heterogeneous group of proteins and diseases is unprecedented and suggests a common pathogenesis (Martin et al., 2010).

When amyloid is confined to a particular area of the body, it is known as localized amyloidosis and when it is found in more than one tissue it is known as systemic amyloidosis (Sipe et al., 2016). In localized amyloidosis, amyloid fibrils are deposited in organs where the precursor protein is produced; for example: amyloid Aβ peptide and Tau protein in the brain (Alzheimer's) and islet of Langerhans amyloid polypeptide or amylin and in the pancreas (type 2 diabetes) (Westermark et al., 2011). In systemic amyloidoses, the precursors are serum proteins, such as portions of immunoglobulins (AL amyloidosis), transthyretin (familial amyloidotic polyneuropathy) and b2-microglobulin (dialysis-related amyloidosis), which circulate in blood and polymerize to form amyloid fibers that are deposited in various organs of the body, except the brain (Sipe et al., 2012; Westermark et al., 2018).

The term amyloidosis is currently used mainly for systemic forms of disease in which the aggregates are definitely pathogenic; some types of amyloidosis have known genetic components, such as: familial amyloidoses in Abyssinian and Siamese cats (van der Linde et al., 1997; Paltrineri et al., 2015), Shar pei dogs (Segev et al., 2012) and hereditary transthyretin amyloidosis in humans (Silva-Hernández et al., 2020). Also in chickens (Gallus gallus domesticus), there is some predisposition to lineage-associated amyloid arthropathy, occurring only in brown egg layers (Ovelgöne, 2001).

There are observational studies in captive wild animals, which have reported a high prevalence of amyloidosis: Rhesus monkey (Macaca mulatta) (Blanchard et al., 1986; Rice et al., 2013, Leung et al., 2019), cheetahs (Acinonyx jubatus) (Papendick et al., 1997; Zhang et al., 2008; Franklin et al., 2016), marmosets (Callithrix jacchus), pig-tailed macaques (Macaca nemestrina) (Rice et al., 2013; Hukkanen et al., 2006), hunting falcons (Hampel et al., 2009), pronghorn (Antilocapra americana) (Martínez et al., 2019), badgers (Meles meles) (Bianco et al., 2020), caracals caracales (Caracal caracal) (Greunz et al., 2020); Japanese tree sparrows (Lonchura striata var. domestica) (Nakano & Madarame, 2020) and treeshrew (Tupaia belangeri) (Klein et al., 2021).

The same has been observed in wildlife in species such as: island foxes (Urocyon littoralis) (Gaffney et al., 2016), red foxes (Vulpes vulpes) (Rising et al., 2017), gulls (Larus argentatus) (Jansson et al., 2018) and Stejneger's beaked whales (Mesoplodon stejnegeri) (Nakagun et al., 2019; Nakagun et al., 2020), among others.

Such observations, especially in captive animals, have suggested the possibility of disease transmission. Experimental studies (Sørby et al., 2008; Murakami et al., 2011; Murakami et al., 2013; Murakami et al., 2014; Murakami et al., 2015) have allowed gathering evidence that amyloidosis known as systemic, indeed can be contagious intra- and interspecies, and thus constitutes a public health risk.

Tojo et al., (2005), found a 5% incidence of renal amyloidosis (15/302) in cattle slaughtered at the slaughterhouse, with no history of disease; although it has been found rarely; systemic AA amyloidosis has also been reported in pigs (Niewold et al., 2005). In a country where viscera are widely consumed, the study of its presence in slaughter cattle becomes more important.

Amyloidosis classification

From the amyloidogenic proteins identified, in humans at least 14 are associated with systemic disease (Westermark et al., 2018) and 19 with localized forms (Benson et al., 2018). In animals 9 have been identified, under normal conditions; all of these proteins are soluble (Zhang et al., 2008).

The accepted nomenclature is AX. Fibrillar (amyloid) proteins are designated as A proteins, followed by a suffix abbreviating the name of the precursor protein (X) (Cooper et al., 2013); for example, in AL amyloidosis, L corresponds to immunoglobulin light chains (light chains), in Alzheimer's disease amyloidosis is ATau, because the precursor protein is named Tau, IAPP is the amyloidosis located in the pancreas of people with type 2 diabetes, as the precursor is islet amyloid polypeptide (Sipe et al., 2016). In the case of systemic AA amyloidosis, it is referred to as AA, because the generating protein is serum amyloid A, a protein of the acute phase of inflammation (Brunger et al., 2020).

The types of amyloidosis that have genetic components and were previously known as "familial" are now suggested to be called hereditary; an example of the former is hereditary transthyretin amyloidosis (hATTR). The precursor protein is thyroxine-retinol transporter prealbumin whose mutation causes a rare multisystem disease in humans, consisting of amyloid deposition mainly in the peripheral nervous system and heart, causing progressive autonomic and sensorimotor peripheral neuropathy and restrictive cardiomyopathy. The eyes, kidneys and central nervous system may also be affected (Silva-Hernández et al., 2020).

In pets, familial amyloidosis (FA) is a hereditary disease that affects Abyssinian cats, and mainly the kidney; the precursor protein is serum amyloid (Paltrinieri et al., 2015). Similarly familial Shar-pei fever (FSF), is a hereditary disorder occurring in up to 23% of animals of this breed; it is characterized by recurrent episodes of fever of unknown origin, concurring with swelling and pain of the hocks, muzzle, abdominal pain, diarrhea and anorexia. It has been proposed that FSF predisposes to secondary reactive amyloidosis, as in humans suffering from Familial Mediterranean fever (Segev et al., 2012). Analysis of 255 DNA samples from Shar pei dogs revealed on chromosomes 13 and 14, areas associated with both FSF and amyloidosis (Olsson et al., 2013).

FSF is similar to Familial Mediterranean fever, an inherited autoinflammatory disease relatively common in people originating from the Eastern Mediterranean Basin, whose most severe complication is AA amyloidosis (Wekell et al., 2016). It is worth mentioning as a particular case, pharmacological amyloidosis, occasionally amyloidomas appear on the skin, around insulin injection sites in diabetics and in HIV patients, due to the administration of an antiretroviral peptide called enfurvitide (Mollee et al., 2013; D’Souza et al., 2014).

Composition, characteristics and pathogenesis

Regardless of the protein of origin, amyloid molecules have unbranched fibrillar structure, with secondary structure in a β-folded arrangement, linked by noncovalent bonds that form both in vivo and in vitro. In the amyloid fiber the repeating polypeptide chains are crosslinked by intercatenary hydrogen bridges; two or more identical ones interact through side chains to form the characteristic fibers of about 8 to 12 nm in diameter, which appear rigid and unbranched under the electron microscope (Martin et al., 2010; Benson et al., 2018).

In most amyloid deposits there are other elements, a glycoprotein called P-component and heparan sulfate-type proteoglycans; complement factors C1q and C3; in addition there may be several apolipoproteins, such as E9. (Martin et al., 2010; Benson et al., 2018; Westermark et al., 2018). It is important to emphasize that a protein is not a static structure. In a living organism or even in vitro, a protein bends, spins, and vibrates; it collides with many molecules every second, such that what is called protein structure is only the state in which the energetic interactions between amino acid groups are most stable. It is this molecular dance that gives rise to the conformation that can act as a "seed" or amyloid core, which when it interacts with the precursor proteins and attaches to them, forms a growing fiber (Martin et al., 2010).

Due to their structure, these proteins, besides being insoluble, become hydrophobic, nonfunctional and resistant to ionic detergents and some proteases; they also have affinity to stains, such as Congo red and thioflavin T (Rubel et al., 2020; Picken, 2020).

In general terms, a protein can change to its amyloid state when a segment exposes the amide-NH and C=O groups of its backbone, allowing them to dock through hydrogen bridges with other protein chains. Several conditions produce exposure of the skeleton of amide groups: denaturation of normally folded proteins, overexpression of a protein that overcomes protein repair mechanisms through chaperones and sends them to inclusion bodies, cleavage of a peptide (such as Aβ) from a folded protein, or overproduction of a disordered protein from its origin (such as Tau or IAPP) (Eisenberg & Jucker, 2012).

In vivo conditions that promote amyloid formation are:

  1. An increase in precursor protein concentration. Nucleated polymerization and fibril formation are dependent on the concentration of the protein. The pathogenesis of some of the systemic amyloidoses (AA, AL, for example); involves an increase in the plasma concentration of the precursor protein (Westermark et al., 2018). In systemic AA amyloidosis, a pre-amyloid phase is required and is characterized by elevated serum amyloid a (SAA) levels, which can result from bacterial infection, viral, autoimmune, autoinflammatory disorders, and metastasis; i.e., inflammatory conditions (Gursky, 2020).

  2. Mutations that destabilize the native forms of the protein and allow amyloidogenic segments to interact with each other. A large proportion of systemic amyloidoses are inherited usually depending on a point mutation resulting in an amino acid substitution. This may be sufficient to destabilize proteins and predispose their misfolding or, as in the case of transthyretin (TTR), make the quaternary structure less stable (Westermark et al., 2018).

  3. Exposure and/or de novo generation of amyloidogenic segments through native protein cleavage or aberrant translation.

  4. Thermodynamically destabilizing conditions, such as temperature or pH modifications. The formation of amyloidomas by insulin administration in diabetics seems to be related to the above two conditions; Amdursky et al., 2012 (Cited by D’Souza et al., 2014), showed that insulin can self-assemble to form a crystalline core, from which fibrils subsequently develop and that such process is accelerated by the acidic environment and high insulin concentration.

  5. Deterioration of the quality control of the cellular proteome (proteostasis), which occurs with aging. Many cellular functions decline, including the regulation of protein synthesis, folding, assembly and degradation. It is well known that amyloid deposits are found after the age of 50 years and that abnormal proteostasis is involved in chronic neuromuscular proteinopathies such as Alzheimer's disease and inclusion body myositis (Romani et al., 2021). Systemic amyloidoses ATTR and AAPoAIV and some localized forms (atrium and seminal vesicles) are associated with aging. ATTR is an underdiagnosed cause of heart failure in older adults. With age the TTR tetramer becomes less stable and this results in the release of misfolding intermediates that eventually form amyloid deposits mainly in the heart of men older than 75 years (Picken, 2020)..

It can be summarized that, in some proteopathies, such as Alzheimer's disease, amyloid arises from de novo misfolding and sustained modification of endogenous proteins; whereas prion diseases can be of infectious origin; although they can also be genetic or sporadic. An amyloid polymer can immobilize monomeric non-amyloid protein molecules of the same sequence and thus grow through a process of nucleic polymerization.

Similar to what occurs in prion diseases, AA amyloidosis is considered to be transmitted through a process of "seeding-nucleation" (Murakami et al., 2015). In these disorders, protein aggregates form characteristic lesions known generically as amyloid; the initial step for their formation is the aggregation of monomers into a "nest", "seed" or nucleus, which acts as the point of origin for fibrillar growth. This individual fiber grows as monomers are added to the free ends. When long fibers break, they produce a greater number of free ends. The formation of the core is the time-determining step in the process and can be shortened from weeks to days when minute amounts of preformed fibrils are added, referred to as amyloid enhancing factor (AEF) (Lundmark et al., 2013).

Infectivity involves the transfer of seeds (prions), from one organism to another; whereas genetic and idiopathic appear to be generated endogenously (Jucker & Walker, 2013).

With any of the above circumstances, the amyloid protein begins to accumulate extracellularly, either locally or systemically, in which case it is deposited in various organs, except the central nervous system; as they accumulate, they deteriorate organ function and clinical signs begin to appear according to their location.

The mechanisms of toxicity have not been completely understood, but it has been observed that in smooth myocytes they alter cell adhesion (Bobylev et al., 2021). In the case of neurodegenerative diseases such as Alzheimer's and Parkinson's, translocation of fibrils to cell membranes can generate mitochondrial damage and trigger apoptosis (Hashimoto et al., 2003). The latter has also been observed in vitro in cardiac myocytes in the case of AL amyloidosis (Shi et al., 2010; Cited by Blancas & Ramírez, 2013).

Systemic AA amyloidosis Background

Systemic amyloidoses culminate in death about 6 months after diagnosis in up to 20% of patients and cause 1/1000 deaths in developed countries where they remain an unresolved medical problem (Scarpioni et al., 2016). Fibrillar proteins are synthesized in their native form in a location such as the liver or bone marrow, released into the circulation and eventually deposited in various tissues (Rising et al., 2017).

In 1961, Benditt and Eriksen observed an "amyloid of unknown origin" protein from gel electrophoresis in cases of so-called "secondary amyloidosis" as being associated with chronic or recurrent inflammatory conditions. Antibodies prepared against these proteins identified a small serum protein of 104 amino acids that turned out to be the precursor of such amyloid deposits; being the first serum protein of a type other than immunoglobulins to be identified as an amyloid precursor, it was named serum amyloid A (SAA) (Sack et al., 2018).

Systemic AA amyloidosis, has worldwide distribution, mainly affects the kidneys and it is caused by the aggregation and deposition of AA protein, a product of the degradation of the N-terminus of serum amyloid (SAA); a protein of the acute phase of inflammation. The kidneys are the organs that are affected earliest, although clinical signs, including proteinuria, develop in very late stages of the disease (Blank et al., 2015).

This disorder affects less than 5% of patients with chronic inflammation (Gursky, 2020). In 90 % of them the kidney is affected and if the concomitant inflammatory process is not controlled, the damage progresses to renal failure. In a study by Lachmann et al., (2007) it was observed that this is more frequent when proteinuria per day is greater than 500 mg and serum creatinine concentrations greater than 1.5 mg/dL. In 20% of patients the gastrointestinal tract is involved and diarrhea and malabsorption occur. With access to hemodialysis and renal transplantation increasing life expectancy, patients with digestive disorders are more common. Amyloidotic gout, hepatomegaly, splenomegaly and polyneuropathies are less frequent clinical findings (Van der Hilst, 2011).

Causes and risk factors of systemic AA amyloidosis

The acute phase response of inflammation comprises a series of very important changes in serum protein levels, such as C-reactive protein and serum amyloid. Both are present in the blood of healthy individuals, although at very low levels (SAA: 20-50 μg/ml); however, these can increase as much as a thousandfold in 24 hours after the onset of the event, basically by their de novo hepatic synthesis.

SAA is normally secreted by the liver, although also by macrophages, endothelial and smooth muscle cells (Scarpioni et al., 2016). It circulates associated with high density lipoprotein, stimulates the release of proinflammatory cytokines such as TNF-α, IL-8 and IL-1b and in turn, its production is stimulated by IL-1, IL-5 and TNF; but also by glucocorticoids and LPS (Brunger et al., 2020).

SAA functions are not fully understood, but are associated with modulation of immune response, cholesterol homeostasis and cell signaling. It is considered to be important for its ability to mobilize cholesterol for cellular repair. It also interacts with a wide array of other diverse ligands: cholesterol, retinol, the proteoglycan heparan sulfate, metal ions, various proteins, etc. (Frame & Gursky, 2017).

In this type of amyloidosis, the existence of a pre-amyloid phase in which there is overproduction of serum amyloid is proposed; this may overcome the quality control of extracellular proteins and the degradation system (Blancas & Ramírez, 2013); however, more than elevated levels of SAA are required as a prerequisite to develop the disease.

Some of the risk factors observed are described below (Table 1); although idiopathic amyloidosis has also been observed when no underlying inflammatory disease is found and obesity and female gender have been associated.

Table 1 Underlying risk factors for systemic AA amyloidosis in humans  

Risk factor Association Presence in Mexico References
Genetics: 5 variants of SAA have been described: SAA 1 (in Caucasian patients) SAA 3 (in Japanese patients) Up to 5 times higher risk of presentation with SAA 1 variants Not determined. There is one study in which being of Mexican ancestry was a risk factor, but it was for ALEC2 renal amyloidosis. (Larsen et al., 2016) Van der Hilst, 2011 Sikora et al., 2021
Age Abnormal proteostasis after 50 years of age 12% of older adults (INEGI, 2000) ) Romani et al., 2021
Obesity Induces chronic inflammation 30% of pupulation (Flores et al., 2019) Van der Heijden et al., 2015; Westermark et al., 2018
Underlying inflammatory diseases Rheumatoid arthritis 67% of cases 0.8% of adult population (Galindo et al., 2013) 0.8% of adult population (Galindo et al., 2013) Brunger et al., (2020)
Skin infections due to chronic abuse of narcotics 2.5 million drug users (ENCODAT, 2016) Mendoza et al., 2013; Cooper et al., 2013; Brunger et al., 2020
HIV 33 new infections/day (CONASIDA 2021) Jung et al., 2012 Breillat et al., 2021 CONASIDA, 2021
Tuberculosis 16,913 cases in 2016 (Flores et al., 2019) Real de Asúa et al., 2014; Khellaf et al.,2022
Mediterranean Fever and other autoinflammatory diseases No diagnosis Brunger et al., 2020
Blood serum amyloid A levels (>155 mg/dL) 17.7 increased risk of death Van der Hilst, 2011;Lachmann et al., 2007; Blank et al., 2015,
Other markers: Asociados a procesos inflamatorios crónicos y daño renal* Van der Hilst, 2011 Van der Heijden et al., 2015 Lachmann et al., 2007
Serum creatinine levels >1.5 mg/dL
C-reactive protein
Proteinuria >500 mg/day
Consumption of animal products containing amyloid
Consumo de productos de origen animal con amiloide 5% in slaughter cattle (Japan), 1.03% (Swedish cattle) and 15.2% (Italian cattle, including muscle samples) It has not been determined Tojo et al.,(2005) Rising et al., (2021)

*Nephrotic syndrome is a terminal stage of the disease

Pathogenesis of systemic AA amyloidosis

It is considered a pre-amyloid phase in which there is an increase in serum amyloid (precursor protein), due to underlying inflammatory processes or other conditions (such as obesity) (Westermark et al., 2018). Experimentally, it has been observed that the time in which the disease develops, is dramatically shortened when animals are stimulated with injections of cells, homogeneous mixtures or extracts of any amyloid-containing tissue. The active components of these various preparations are called: amyloid facilitating factors (AEF) and it is known that they can function as "seeds" for fiber formation (Lundmark et al., 2005).

Some of the proteins naturally grow from one "seed" of another; in the case of SAA, apoprotein A-II in mice (Yan et al., 2007; Cited by Sack et al., 2018). Even preformed fibers can act as core for amyloid formation in vitro and AA amyloid and similar synthetic fibers; they can also serve for its formation in vivo. Lundmark et al., (2005) conducted a study in mice inoculated with three naturally fibrillar proteins: silk from Bombyx mori, Sup35 from Saccharomyces cerevisiae and curli from Escherichia coli. All proteins had the ability to accelerate the process of AA amyloidosis formation that is well studied in animal models in which AgNO3 is repeatedly injected.

For SAA to be incorporated into an amyloid fiber, two processes are required: first, the Cterminal end must be cleaved from the molecule to adopt the β-folded configuration, and in the second process SAA undergoes clathrin-mediated endocytosis by macrophages and is transported to their lysosomes. Structural reorganization then occurs at pH 4.3 with or without proteolysis by cathepsin B. Under normal conditions, it would be completely degraded; however, in patients with amyloidosis this does not occur. After deposition of the accumulated intermediates in the extracellular space, several elements bind to the fibril and confer resistance: glycosaminoglycans, the plasma glycoprotein known as the P-component, which is a member of the pentraxin protein family (like C-reactive protein), and lipid components (Van der Hilst, 2011; Sack et al., 2018).

Macrophages are often detected in close association with amyloid and there is evidence that they are involved in both its formation and degradation, since they synthesize a wide range of proteases (Van der Hilst, 2011; Lundmark et al., 2013).

It has also been observed that destruction of macrophages (with an injection of clodronate, a biophosphonate); in murine models for AA amyloid formation via AEF and AgNO3, delayed amyloid formation. The finding of intracellular amyloid in macrophages also suggests that the process of "seeding nucleation", indeed initiates there (Lundmark et al., 2013).

The fact that the onset of AA amyloidosis can be accelerated by fibers derived from unrelated proteins, which are normally non-pathogenic, indicates that environmental factors may be important risk factors. In addition to AEF, poor hygiene or stimuli to the immune system by repeated vaccinations could also be important (Murakami et al., 2013). As mentioned above, Westermark et al., (2018), observed similarities to prion transmission and emphasized the process of nucleus formation as central; such a mechanism could be the underlying factor for AA transmission among captive animals (Ranlov 1967; cited by Sack et al., 2018).

AA amyloidosis in animals and intra- and interspecies transmission

AA amyloidosis occurs spontaneously in many avian and mammalian species, but prevalence varies considerably even among subgroups of the same species (Rising et al., 2017). As with prions, transmission has been observed in cattle, birds, mice and cheetahs (Murakami et al., 2014).

One of the first studies suggesting the possibility of interspecies transmission was that of Papendick et al., (1997), who from necropsy data of 141 cheetahs from 38 different zoos, observed a prevalence of systemic amyloidosis AA of 20% in animals that died before 1990, which increased to 70% in 1995. Subsequently, Zhang et al. (2008) identified AA amyloid through immunohistochemistry in livers, and subsequently in feces through Western blotting. From these results, it was presumed that fecal proteins could be involved in the spread of amyloidosis; this was proven in mice inoculated with such material intravenously and it was also observed that fecal fibrils presented greater transmission capacity.

A high prevalence has been observed in many other species in captivity. Martínez et al. (2019), reported 77% in a population of pronghorn (Antilocapra americana) at the Columbus Zoo, from animals whose necropsies were performed between 1997 and 2016, and concluded that in this case the underlying inflammatory problems were haemonchosis and pneumonia.

In other species, such as badgers (Meles meles) (Bianco et al., 2020), due to its frequency, it has also been concluded that it should be included in the differential diagnoses of consumptive diseases.

In Europe, 9 cases were reviewed in caracals (Caracal caracal) (Greunz et al., 2020), all animals presented renal amyloidosis, although only one case of systemic AA amyloidosis was confirmed by immunohistochemistry. In the study they concluded that due to the kinship of the animals, the problem could be associated with genetics and that given the prevalence, it should be considered within the differential diagnosis of renal problems.

A relationship between disease and AA amyloidosis has been experimentally proven: with subcutaneous inflammation in mice and hamsters, hydatid alveolar disease in mice and infection by Opisthorchis viverrini and Enterococcus faecalis in chickens (Brunger et al., 2020).

Some research has considered risk factors. At the Oregon National Primate Research Center, data were used retrospectively, from macaques (Macaca mulatta) that died within 5 years (n=3061), with the goal of identifying risk factors that would predict the development of amyloidosis AA prior to the onset of clinical signs, and exploring potential new markers of the disease for use on a large scale (Leung et al., 2019). It was found a prevalence of systemic amyloidosis of 9.2% and observed that chronic colitis, gastrointestinal adenocarcinoma, endometriosis, and osteoarthritis, entities characterized by chronic, systemic inflammation, were the main risk factors; trauma, number of pregnancies, and diarrhea not associated with colitis followed. Circulating levels of serum amyloid A, triglycerides and triglyceride:HDL ratio were also observed, as well as lower levels of HDL and lower LDL than in healthy controls. These results are consistent with findings in chronic inflammation of altered lipoprotein metabolism and SAA.

In wildlife animals, important data have also been found, the island fox (Urocyon littoralis), which is located in the Northern Archipelago, off the coast of California; free-living and captive animals coexist on these islands. A study was conducted using necropsy data from 321 animals that died between 1987 and 2010; the prevalence of amyloidosis was 34% (109/321). Evidence of chronic inflammation was found in 83.5% of amyloidosis cases. The most common macroscopic lesions were splenomegaly, macroglossia and pallor; as well as waxy appearance of the kidneys in the most severe cases. Twenty-nine percent of the animals died from renal amyloidosis.

Some risk factors could be studied and it was found that captive animals had a significantly higher prevalence (59 vs. 27%). A higher risk was also observed in female foxes and in certain subspecies. The latter could be a factor associated with the type of SAA, as in the case of other species (Gaffney et al., 2016).

In marine mammals, a study was conducted following stranded animals off the coast of Hokkaido, Japan between 2013 and 2018; in 2 of 3 Stegnejer's whales (Mesoplodon stejnegeri) systemic AA amyloidosis was diagnosed, associated with renal parasitosis by Crassicauda sp.; slight splenomegaly and hepatomegaly were also observed and the most important involvement in kidney and gastrointestinal tract. It is considered that these cetaceans, as has been observed in other species, may have some genetic predisposition to amyloidosis (Nakagun et al., 2019).

Animal models have been developed to produce systemic AA amyloidosis. Initially, it was observed that repeated exposure of mice to inflammatory stimuli, such as subcutaneous injections of silver nitrate, complete Freund's adjuvant, casein, or lipopolysaccharide, can induce the disease within several weeks; but this period can be shortened if an amyloidenhancing factor (AEF) is administered. Amyloid AA fibrils, amyloid AL fibrils and Alzheimer's brain extract can function as AEF. The work of Murakami et al., (2011) in which systemic amyloidoisis AA was induced experimentally after observing that pododermatitis lesions in which a Staphylococcus aureus infection was frequently found, provides insight into part of the pathogenesis of the disease.

An intravenous solution of AA amyloid fibrillar material from Holstein friesian bovine kidneys with systemic amyloidosis was inoculated into rabbits previously induced with pododermatitis lesions; it was concluded that the presence of S. aureus was very efficient in the development of amyloidosis and that the presence of pododermatitis also favored it. Rabbits without lesions did not develop the disease. Similar findings have been found in waterfowl where a prevalence of amyloidosis of 78.4% was observed in swans (Cygnus olor) and a very high frequency (96.3%) of an inflammatory condition known as bumblefoot (Tanaka et al., 2008), which is a chronic inflammatory condition of the plantar metatarsal and/or digital patches of the birds' feet.

Diagnosis

Amyloidosis can present with puzzling clinical pictures, depending on the organs involved. In humans, localized amyloidosis usually presents periorbital, nasopharyngeal, pulmonary and/or bronchial, cutaneous, gastrointestinal and urinary; as well as in lymph nodes. Fatigue and thinning may also occur initially and later signs according to the organs involved. It has already been commented that in systemic amyloidosis, the final picture corresponds with nephrotic range proteinuria (Mollee et al., 2013).

In all cases, the patient's phenotype should be considered and the search for associated diseases or chronic inflammation should begin; however, association is not always evidence of causality. For the case of AA amyloidosis, since inflammation is usually present, SAA levels should ideally be measured; however, it is an assay that is not routinely available and C-reactive protein may be a suitable marker if used in serial measurements (Mollee et al., 2013). Diagnosis of amyloidosis requires identification of amyloid deposits in tissue samples; in animals, it is usually a post-mortem diagnosis. In humans, to avoid highly invasive procedures, abdominal fat aspiration is used, although its sensitivity is limited (Mollee et al., 2013). Congo Red staining with polarized light microscopy is the first histological method used to confirm its presence, followed by subtype classification through immunohistochemistry of the main precursors; however, it is sometimes not as specific and can generate false positives; in addition, it is a semi-quantitative technique. More recently, laser microdissection, liquid chromatography, followed by mass spectrophotometry have been used. This method has high sensitivity and specificity, and minimal sample quantities are required.

There is already evidence that the sensitivity and specificity of laser microdissection, liquid chromatography and mass spectrophotometry is adequate to detect amyloidosis with very small amounts of tissue and not only amyloid precursors, but also associated proteins (Kadota et al., 2020).

Although this technique is becoming the method of choice for amyloid typing, the application of immunological methods remains clinically useful. Caution and experience are required, as well as knowledge of the limitations of each method to properly interpret the results (Picken, 2020) and classify the disease to know whether or not it is treatable, both in humans and animals.

D’Souza et al., (2014), used mass spectrophotometry to identify the components of amyloid deposits reported in 52 patients who had been administered insulin or efurvitide. Similarly, Ogawa et al., (2020), were able to identify 12 of 13 precursors, including serum amyloid, transthyretin and immunoglobulins, with that technique; these were quantified even though the results by immunohistochemistry were inconclusive.

In animals, laser microdissection has also been used for sample collection and processing by liquid chromatography and mass spectrophotometry to diagnose AL amyloidosis. In a case study of 15 dogs and 2 cats; in 11 the disease could not be diagnosed by immunohistochemistry, this occurs because antibodies of human origin are sometimes used. It has been observed that the use of a combination of antibodies can improve the sensitivity of the test.

In this study, 100% of the cases could be diagnosed by liquid chromatography-mass spectrophotometry (Kadota et al., 2020).

CONCLUSIONS

There are no reports in Mexico on systemic AA amyloidosis in wild or domestic animals. Since this disease is difficult to diagnose and has unspecific clinical signs, it is probably underdiagnosed. In the review performed, only one report on ALEC2 renal amyloidosis was found in the southwestern United States, in which 54% of the diagnoses were associated with race (Mexican-Americans). Other risk factors such as chronic inflammatory diseases, obesity and/or advanced age are present in the Mexican population. Given that in Mexico a large amount of animal products are consumed, including viscera, and that interspecies transmission of the disease has been proven, it is considered very important for public health to awaken interest in the study of systemic AA amyloidosis.

REFERENCES

Andrei M, Wang JC. 2019. Cutaneous light chain amyloidosis with multiple myeloma: A concise review. Hematology/oncology and stem cell therapy. 12(2):71-81. https://doi.org/10.1016/j.hemonc.2018.09.003 [ Links ]

Benson MD, Buxbaum JN, Eisenberg DS, Merlini G, Saraiva M, Sekijima Y, Sipe JD, Westermark P. 2018. Amyloid nomenclature 2018: recommendations by the International Society of Amyloidosis (ISA) nomenclature committee. Amyloid: the international journal of experimental and clinical investigation: the official journal of the International Society of Amyloidosis. 25(4):215-219. https://doi.org/10.1080/13506129.2018.1549825 [ Links ]

Bianco C, Sánchez-Cordón PJ, Verin R, Godinho A, Weyer U, Lesellier S, Spiropoulos J, Floyd T, Everest D, Núñez A. 2020. Investigation into the Pathology of Idiopathic Systemic Amyloidosis in Four Captive Badgers (Meles meles). Journal of comparative pathology. 176:128-132. https://doi.org/10.1016/j.jcpa.2020.02.012 [ Links ]

Blancas-Mejía LM, Ramirez-Alvarado M. 2013. Systemic amyloidoses. Annual review of biochemistry. 82: 745-774. https://doi.org/10.1146/annurev-biochem-072611-130030 [ Links ]

Blanchard JL, Baskin GB, Watson EA. 1986. Generalized amyloidosis in rhesus monkeys. Veterinary pathology. 23(4): 425-430. https://doi.org/10.1177/030098588602300412 [ Links ]

Blank N, Hegenbart U, Lohse P, Beimler J, Röcken C, Ho AD, Lorenz HM, Schönland SO. 2015. Risk factors for AA amyloidosis in Germany. Amyloid. 22(1): 1-7 . https://doi.org/ 10.3109/13506129.2014.980942Links ]

Blank N, Hegenbart U, Dietrich S, Brune M, Beimler J, Röcken C, Müller-Tidow C, Lorenz HM, Schönland SO. 2018. Obesity is a significant susceptibility factor for idiopathic AA amyloidosis. Amyloid. 25(1):37-45. https://doi.org/10.1080/13506129.2018.1429391 [ Links ]

Bobylev AG, Fadeev RS, Bobyleva LG, Kobyakova MI, Shlyapnikov YM, Popov DV, Vikhlyantsev IM. 2021. Amyloid Aggregates of Smooth-Muscle Titin Impair Cell Adhesion. International journal of molecular sciences. 22(9):4579. https://doi.org/10.3390/ijms22094579 [ Links ]

BreillaT P, Pourcher V, Deshayes S, Buob D, Cez A, Michel P A, Boffa J J, Langlois V, Grateau G, Georgin-Lavialle S. 2021. AA Amyloidosis in the Course of HIV Infection: A Report of 19 Cases Including 4 New French Cases and a Comprehensive Review of Literature. Nephron. 145(6): 675-683. https://doi.org/10.1159/000516982 [ Links ]

Brunger AF, Nienhuis H, Bijzet J, Hazenberg B. 2020. Causes of AA amyloidosis: a systematic review. Amyloid. 27(1): 1-12. https://doi.org/10.1080/13506129.2019.1693359 [ Links ]

CONASIDA (Consejo Nacional para la prevención y control del SIDA). 2021. Sistema de vigilancia epidemiológica de VIH. Informe histórico Día mundial VIH 2021. https://www.gob.mx/cms/uploads/attachment/file/685220/VIHSida__D_a_Mundial_2021.pdfLinks ]

Cooper C, Bilbao J E, Said S, Alkhateeb H, Bizet J, Elfar A, Davalos O, Meza AT, Hernandez G T. 2013. Serum amyloid A renal amyloidosis in a chronic subcutaneous ("skin popping") heroin user. Journal of Nephropatholy. 2(3): 196-200. https://doi.org/10.12860/JNP.2013.31 [ Links ]

D'Souza A, Theis JD, Vrana JA, Dogan A. 2014. Pharmaceutical amyloidosis associated with subcutaneous insulin and enfuvirtide administration. Amyloid.21(2): 71-75. https://doi.org/10.3109/13506129.2013.876984 [ Links ]

Eisenberg D, Jucker M. 2012. The amyloid state of proteins in human diseases. Cell.148(6):1188-1203. https://doi.org/10.1016/j.cell.2012.02.022 [ Links ]

ENCODAT (Encuesta Nacional de consumo de drogas, alcohol y tabaco). 2016. Encuesta nacional de consumo de drogas, alcohol y tabaco 2016-2017. Pp.1. https://www.gob.mx/cms/uploads/ATTACHMENT/file/234856/CONSUMO_DE_DROGAS.pdfLinks ]

Flores Rodríguez J, Rivera Franco MM, Apodaca Cruz Á, Saldaña Montaño MC, Urbalejo Ceniceros VI, Meneses García A, Herrera Gómez A, Perez Camargo DA, Sevilla González ML. 2019. Incidence and characteristics of metabolic syndrome in patients of the National Cancer Institute of Mexico. Nutrición hospitalaria. 36 (6):1296-1299. https://dx.doi.org/10.20960/nh.02395 [ Links ]

Flores-Treviño S, Rodríguez-Noriega E, Garza-González E, González-Díaz E, Esparza-Ahumada S, Escobedo-Sánchez R, Pérez-Gómez HR,León-Garnica G, MorfínOtero R. 2019. Clinical predictors of drug-resistant tuberculosis in Mexico. PLoS ONE14(8): e0220946. https://doi.org/10.1371/journal.pone.0220946 [ Links ]

Frame NM, Gursky O. 2017. Structure of serum amyloid A suggests a mechanism for selective lipoprotein binding and functions: SAA as a hub in macromolecular interaction networks. Amyloid. 24(1): 13-14 https://doi.org/10.1080/13506129.2016.1270930 [ Links ]

Franklin AD, Schmidt-Küntzel A, Terio KA, Marker LL, Crosier AE. 2016. Serum Amyloid A Protein Concentration in Blood is Influenced by Genetic Differences in the Cheetah (Acinonyx jubatus). The Journal of heredity. 107(2): 115-121. https://doi.org/10.1093/jhered/esv089 [ Links ]

Gaffney PM, Witte C, Clifford DL, Imai DM, O'Brien TD, Trejo M, Liberta F, Annamalai K, Fändrich M, Masliah E, Munson L, Sigurdson CJ. 2016. Systemic Amyloid A Amyloidosis in Island Foxes (Urocyon littoralis): Severity and Risk Factors. Veterinary pathology. 53(3): 637-647 . https://doi.org/ 10.1177/0300985815604725Links ]

Galindo SRM, Peniche OG, Herrera RJ, Gryzbowski E. 2013. Estimación de la prevalencia de artritis reumatoide en México. Value in health. ISPOR 16TH Annual European Congress Research abstracts. 16 (7): A719. https://doi.org/10.1016/j.jval.2013.08.2235 [ Links ]

Gursky O. 2020. Structural Basis for Vital Function and Malfunction of Serum Amyloid A: an Acute-Phase Protein that Wears Hydrophobicity on Its Sleeve. Current atherosclerosis reports. 22(11 ): 69 . https://doi.org/ 10.1007/s11883-020-00888-yLinks ]

Greunz EM, Lemberger K, Catinaud J, Chenet B, Linke RP, Bräsen JH, Schmitz J, Bertelsen MF. 2020. AMYLOIDOSIS IN CARACALS (CARACAL CARACAL). Journal of zoo and wildlife medicine. 51(1):202-209. https://doi.org/10.1638/2019-0005 [ Links ]

Gruys E. 2004. Protein folding pathology in domestic animals. Journal of Zhejiang University-Science. 5(10): 1226-1238. https://doi.org/10.1631/jzus.2004.1226 [ Links ]

Hampel MR, Kinne J, Wernery U, Pospischil A, Kellermann J, Linke RP. 2009. Increasing fatal AA amyloidosis in hunting falcons and how to identify the risk: a report from the United Arab Emirates. Amyloid.16(3):122-132. https://doi.org/10.1080/13506120903090759 [ Links ]

Hashimoto M, Rockenstein E, Crews L, Masliah E. 2003. Role of protein aggregation in mitochondrial dysfunction and neurodegeneration in Alzheimer's and Parkinson's diseases. Neuromolecular medicine. 4:21-35. https://doi.org/10.1385/NMM:4:1-2:21 [ Links ]

Hukkanen RR, Liggitt HD, Anderson DM, Kelley ST 2006. Detection of systemic amyloidosis in the pig-tailed macaque (Macaca nemestrina). Comparative medicine. 56(2): 119-127. https://pubmed.ncbi.nlm.nih.gov/16639979/ [ Links ]

INEGI (Instituto Nacional de Estadística, Geografía e Informática). 2020. Comunicado de prensa número 24/21. 25 de enero de 2021. Pp. 2. https://www.inegi.org.mx/contenidos/saladeprensa/boletines/2021/EstSociodemo/Result Censo2020_Nal.pdf Links ]

Jansson DS, Bröjer C, Neimanis A, Mörner T, Murphy CL, Otman F, Westermark P. 2018. Post mortem findings and their relation to AA amyloidosis in free-ranging Herring gulls (Larus argentatus). PloS ONE.13(3): e0193265. https://doi.org/10.1371/journal.pone.0193265 [ Links ]

Jucker M, Walker LC. 2013. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature: 501(7465):45-51. https://doi.org/10.1038/nature12481 [ Links ]

Jung O, Haack HS, Buettner M, Betz C, Stephan C, Gruetzmacher P, Amann K, Bickel M. 2012. Renal AA-amyloidosis in intravenous drug users--a role for HIV-infection?. BMC nephrology. 13: 151. https://doi.org/10.1186/1471-2369-13-151 [ Links ]

Kadota A, Iwaide S, Miyazaki S, Mitsui I, Machida N, Murakami T. 2020. Pathology and Proteomics-Based Diagnosis of Localized Light-Chain Amyloidosis in Dogs and Cats. Veterinary pathology. 57(5): 658-665. https://doi.org/10.1177/0300985820934113 [ Links ]

Khellaf G, Benziane A, Kaci L, Benabadji M. 2022. AA renal amyloidosis: Clinical observations over 20 years. Clinical nephrology. 97: 167-172. https://doi.org/10.5414/CN110577 [ Links ]

Klein A, Radespiel U, Felmy F, Brezina T, Ciurkiewicz M, Schmitz J, Bräsen JH, Linke R P , Reinartz S, Distl O, Beineke A. 2021. AA-amyloidosis in captive northern tree shrews (Tupaia belangeri). Veterinary pathology. https://doi.org/10.1177/03009858211066847 [ Links ]

Lachmann HJ, Goodman HJ, Gilbertson JA, Gallimore JR, Sabin CA, Gillmore JD, Hawkins PN. 2007. Natural history and outcome in systemic AA amyloidosis. The New England journal of medicine. 356(23): 2361-2371. https://doi.org/10.1056/NEJMoa070265 [ Links ]

Larsen CP, Beggs ML, Wilson JD, Lathrop SL. 2016. Prevalence and organ distribution of leukocyte chemotactic factor 2 amyloidosis (ALECT2) among decedents in New Mexico. Amyloid.23(2): 119-123. https://doi.org/10.3109/13506129.2016.1145110 [ Links ]

Leung ET, Raboin MJ, McKelvey J, Graham A, Lewis A, Prongay K, Cohen AM, Vinson A. 2019. Modelling disease risk for amyloid A (AA) amyloidosis in non-human primates using machine learning. Amyloid. 26(3): 139-147. https://doi.org/10.1080/13506129.2019.1625038 [ Links ]

Lin X, Watanabe K, Kuragano M, Tokuraku K. 2021. Aggregation of Mouse Serum Amyloid A Protein Was Promoted by Amyloid-Enhancing Factors with the More Genetically Homologous Serum Amyloid A. International journal of molecular sciences. 22(3):1036. https://doi.org/10.3390/ijms22031036 [ Links ]

Ludlage E, Murphy>CL, Davern SM, Solomon A, Weiss DT, Glenn-Smith D, Dworkin S, Mansfield KG. 2005. Systemic AA amyloidosis in the common marmoset. Veterinary pathology. 42(2):117-124. https://doi.org/10.1354/vp.42-2-117 [ Links ]

Lundmark K, Westermark G T, Olsén A, Westermark P . 2005. Protein fibrils in nature can enhance amyloid protein A amyloidosis in mice: Cross-seeding as a disease mechanism. Proceedings of the National Academy of Sciences of the United States of America.102(17): 6098-6102. https://doi.org/10.1073/pnas.0501814102 [ Links ]

Lundmark K, Vahdat Shariatpanahi A, Westermark GT. 2013. Depletion of spleen macrophages delays AA amyloid development: a study performed in the rapid mouse model of AA amyloidosis. PloS ONE. 8(11): e79104. https://doi.org/10.1371/journal.pone.0079104 [ Links ]

Maji SK, Perrin MH, Sawaya MR, Jessberger S, Vadodaria K, Rissman RA, Singru PS, Nilsson KP, Simon R, Schubert D, Eisenberg D, Rivier J, Sawchenko P, Vale W,Riek R. 2009. Functional amyloids as natural storage of peptide hormones in pituitary secretory granules. Science. 325(5938): 328-332. https://doi.org/10.1126/science.1173155 [ Links ]

Martin DJ, Randles EG, Ramirez-Alvarado M. 2010. Fibril structure and fibrillogenesis. En: Gertz M.A. y Rajkumar S.J. Amyloidosis: Diagnosis and treatment 1ª edición. Humana Press. Pp.2. ISBN: 978-1-60761-631-3. https://doi.org/10.1007/978-1-60761-631-3 [ Links ]

Martinez ME, Zimmerman D, Seeley KE, Zhang L, Bapodra P, Cianciolo RE. 2019. Systemic amyloidosis in a population of pronghorn antelope (Antilocapra americana). Journal of zoo and wildlife medicine. 50(1):147-158. https://doi.org/10.1638/2018-0055 [ Links ]

Mendoza JM, Peev V, Ponce MA, Thomas DB, Nayer A. 2013. Amyloid A amyloidosis with subcutaneous drug abuse. Journal of renal injury prevention. 3(1): 11-16. https://doi.org/10.12861/jrip.2014.06 [ Links ]

Mollee P, Renaut P, Gottlieb D, Goodman H. 2014. How to diagnose amyloidosis. Internal medicine journal. 44(1): 7-17. https://doi.org/10.1111/imj.12288 [ Links ]

Murakami T, Inoshima Y, Watanabe K, Kobayashi Y, Matsui T, Kurazono H, Ishiguro N. 2011. Pathogenesis of experimental amyloid protein A amyloidosis in sore hocksaffected rabbits. Amyloid. 18(3): 112-118 . https://doi.org/ 10.3109/13506129.2011.582901Links ]

Murakami T, Muhammad N, Inoshima Y, Yanai T, Goryo M, Ishiguro N. 2013. Experimental induction and oral transmission of avian AA amyloidosis in vaccinated white hens. Amyloid. 20 (2): 80-85. https://doi.org/10.3109/13506129.2013.783474 [ Links ]

Murakami T, Ishiguro N, Higuchi K. 2014. Transmission of systemic AA amyloidosis in animals. Veterinary pathology. 51(2):363-371. https://doi.org/10.1177/0300985813511128 [ Links ]

Murakami T, Inoshima Y, Ishiguro N. 2015. Systemic AA amyloidosis as a prion-like disorder. Virus research. 207: 76-81. https://doi.org/10.1016/j.virusres.2014.12.019 [ Links ]

Nakagun S, Watanabe K, Matsuishi, T., Kobayashi, M., Kobayashi, Y. 2019. Surveillance of amyloidosis in stranded and bycaught cetaceans off Hokkaido, Japan. The Journal of veterinary medical science. 81(6): 897-902. https://doi.org/10.1292/jvms.180706 [ Links ]

Nakagun S, Watanabe K, Tajima Y, Yamada TK, Kobayashi Y. 2020. Systemic Amyloid A Amyloidosis in Stejneger's Beaked Whales (Mesoplodon stejnegeri).Veterinary pathology. 57 (3):437-444. https://doi.org/10.1177/0300985820914079 [ Links ]

Nakano Y, Madarame H. 2020. Systemic amyloid A (AA) amyloidosis in the Bengalese finch (Lonchura striata var. domestica).The Journal of veterinary medical science. 82(10): 1484-1487. https://doi.org/10.1292/jvms.20-0365 [ Links ]

Niewold TA, Murphy CL , Toussaint MJ, Solomon A, Gruys E. 2005. Chemical typing of porcine systemic amyloid as AA-amyloid. Amyloid. 12(3): 164-166. https://doi.org/10.1080/13506120500231806 [ Links ]

Ogawa M, Shintani-Domoto Y, Nagashima Y, Ode KL, Sato A, Shimizu Y, Ohashi K, Roehrl M, Ushiku T, Ueda HR, Fukayama M. 2020. Mass spectrometry-based absolute quantification of amyloid proteins in pathology tissue specimens: Merits and limitations. PloS ONE. 15 (7): e0235143.https://doi.org/10.1371/journal.pone.0235143 [ Links ]

Olsson M, Tintle L, Kierczak M, Perloski M, Tonomura N, Lundquist A, Murén E, Fels M, Tengvall K, Pielberg G, Dufaure de Citres C, Dorso L, Abadie J, Hanson J, Thomas A, Leegwater P, Hedhammar Å, Lindblad-Toh K, Meadows J. R. 2013. Thorough investigation of a canine autoinflammatory disease (AID) confirms one main risk locus and suggests a modifier locus for amyloidosis. PloS ONE. 8(10 ): e75242 . https://doi.org/ 10.1371/journal.pone.0075242Links ]

Ovelgönne JH, Landman WJ, Gruys E, Gielkens AL, Peeters BP. 2001. Identical amyloid precursor proteins in two breeds of chickens which differ in susceptibility to develop amyloid arthropathy. Amyloid. 8(1): 41-51. https://doi.org/10.3109/13506120108993813 [ Links ]

Paltrinieri S, Sironi G, Giori L, Faverzani S, Longeri M. 2015. Changes in serum and urine SAA concentrations and qualitative and quantitative proteinuria in Abyssinian cats with familial amyloidosis: a five-year longitudinal study (2009-2014). Journal of veterinary internal medicine. 29(2): 505-512. https://doi.org/10.1111/jvim.12561 [ Links ]

Papendick RE, Munson L, O'Brien TD, Johnson KH.1997. Systemic AA amyloidosis in captive cheetahs (Acinonyx jubatus). Veterinary pathology. 34(6): 549-556. https://doi.org/10.1177/030098589703400602 [ Links ]

Picken MM. 2020. The Pathology of Amyloidosis in Classification: A Review. Acta haematologica.143 (4): 322-334. https://doi.org/10.1159/000506696 [ Links ]

Real de Asúa D, Costa R, Galván JM, Filigheddu MT, Trujillo D, Cadiñanos, J. 2014. Systemic AA amyloidosis: epidemiology, diagnosis, and management. Clinical epidemiology. 6: 369-377. https://doi.org/10.2147/CLEP.S39981 [ Links ]

Rice KA, Chen ES, Metcalf Pate KA, Hutchinson EK, Adams RJ. 2013. Diagnosis of amyloidosis and differentiation from chronic, idiopathic enterocolitis in rhesus (Macaca mulatta) and pig-tailed (M. nemestrina) macaques. Comparative medicine. 63(3): 262- 271.https://pubmed.ncbi.nlm.nih.gov/23759529/ Links ]

Rising A, Cederlund E, Palmberg C, Uhlhorn H, Gaunitz S, Nordling K, Ågren E, Ihse E, Westermark GT, Tjernberg L, Jörnvall H, Johansson J, Westermark P . 2017. Systemic AA amyloidosis in the red fox (Vulpes vulpes). Protein science. 26(11): 2312-2318. https://doi.org/10.1002/pro.3264 [ Links ]

Rising A, Gherardi P, Chen G, Johansson J, Oskarsson ME, Westermark GT, Westermark P . 2021. AA amyloid in human food chain is a possible biohazard. Scientific reports. 11(1): 21069. https://doi.org/10.1038/s41598-021-00588-w [ Links ]

Romani M, Sorrentino V, Oh CM, Li H, de Lima TI, Zhang H, Shong M, Auwerx J. 2021. NAD+ boosting reduces age-associated amyloidosis and restores mitochondrial homeostasis in muscle. Cell reports. 34(3): 108660. https://doi.org/10.1016/j.celrep.2020.108660 [ Links ]

Rubel MS, Fedotov SA, Grizel AV, Sopova JV, Malikova OA, Chernoff YO, Rubel AA. 2020. Functional Mammalian Amyloids and Amyloid-Like Proteins. Life. 10(9):156. https://doi.org/10.3390/life10090156 [ Links ]

Sack, GH. Jr 2018. Serum amyloid A - a review. Molecular medicine. 24 (1):46. https://doi.org/10.1186/s10020-018-0047-0 [ Links ]

Scarpioni R, Ricardi M, Albertazzi V. 2016. Secondary amyloidosis in autoinflammatory diseases and the role of inflammation in renal damage. World journal of nephrology. 5 (1): 66-75. https://doi.org/10.5527/wjn.v5.i1.66 [ Links ]

Segev G, Cowgill LD, Jessen S, Berkowitz A, Mohr CF, Aroch I. 2012. Renal amyloidosis in dogs: a retrospective study of 91 cases with comparison of the disease between SharPei and non-Shar-Pei dogs. Journal of veterinary internal medicine. 26(2): 259-268. https://doi.org/10.1111/j.1939-1676.2011.00878.x [ Links ]

Sikora J, Kmochová T, Mušálková D, Pohludka M, Přikryl P, Hartmannová H, Hodaňová K, Trešlová H, Nosková L, Mrázová L, Stránecký V, Lunová M, Jirsa M, Honsová E, Dasari S, McPhail ED, Leung N, Živná M, Bleyer A J, Rychlík I, Kmoch S. 2021. A mutation in the SAA1 promoter causes hereditary amyloid A amyloidosis. Kidney international. 101(2): 349-359. https://doi.org/10.1016/j.kint.2021.09.007 [ Links ]

SIlva-Hernández L, Horga Hernández A, Valls Carbó A, Guerrero Sola A, MontalvoMoraleda MT, Galán Dávila L. 2020. «Red flags» en pacientes con polineuropatía amiloidótica familiar relacionada con transtiretina (hATTR) en el momento del diagnóstico en un área no endémica de España. Neurología. S0213-4853(20): 30212-7. https://doi.org/10.1016/j.nrl.2020.06.009 [ Links ]

Sipe JD, Benson MD, Buxbaum JN, Ikeda S, Merlini G, Saraiva MJ, Westermark P. Nomenclature Committee of the International Society of Amyloidosis .2012. Amyloid fibril protein nomenclature: 2012 recommendations from the Nomenclature Committee of the International Society of Amyloidosis. Amyloid. 19(4): 167-170. https://doi.org/10.3109/13506129.2012.734345 [ Links ]

Sipe JD, Benson MD, Buxbaum JN, Ikeda SI, Merlini G, Saraiva MJ, Westermark P. 2016. Amyloid fibril proteins and amyloidosis: chemical identification and clinical classification International Society of Amyloidosis 2016 Nomenclature Guidelines. Amyloid. 23(4): 209- 213. https://doi.org/10.1080/13506129.2016.1257986 [ Links ]

Sørby R, Espenes A, Landsverk T, Westermark G. 2008. Rapid induction of experimental AA amyloidosis in mink by intravenous injection of amyloid enhancing factor. Amyloid. 15(1): 20-28. https://doi.org/10.1080/13506120701815332 [ Links ]

Tamura Y, Chambers JK, Neo S, Goto-Koshino Y, Takagi S, Uneyama M, Uchida, K, Hisasue M. 2020. Primary duodenal plasmacytoma with associated primary (amyloid lightchain) amyloidosis in a cat. Journal of Feline Medicine and Surgery open reports.6(2): 2055116920957194. https://doi.org/10.1177/2055116920957194 [ Links ]

Tanaka S, Dan C, Kawano H, Omoto M, Ishihara T. 2008. Pathological study on amyloidosis in Cygnus olor (mute swan) and other waterfowl. Medical molecular morphology. 41(2): 99-108. https://doi.org/10.1007/s00795-008-0401-3 [ Links ]

Tojo K, Tokuda T, Hoshii Y, Fu X, Higuchi K, Matsui T, Kametani F, Ikeda S. 2005. Unexpectedly high incidence of visceral AA-amyloidosis in slaughtered cattle in Japan. Amyloid. 12(2): 103-108. https://doi.org/10.1080/13506120500107097 [ Links ]

Van der Heijden RA, Bijzet J, Meijers WC, Yakala GK, Kleemann R, Nguyen TQ, de Boer RA, Schalkwijk CG, Hazenberg BP, Tietge UJ, Heeringa P. 2015. Obesity-induced chronic inflammation in high fat diet challenged C57BL/6J mice is associated with acceleration of age-dependent renal amyloidosis. Scientific reports. 5: 16474. https://doi.org/10.1038/srep16474 [ Links ]

Van der Hilst JC. 2011. Recent insights into the pathogenesis of type AA amyloidosis. The Scientific World Journal. 11: 641-650. https://doi.org/10.1100/tsw.2011.64 [ Links ]

Van der Linde-Sipman JS, Niewold TA, Tooten PC, de Neijs-Backer M, Gruys E.1997. Generalized AA-amyloidosis in Siamese and Oriental cats. Veterinary immunology and immunopathology. 56 (1-2): 1-10. https://doi.org/10.1016/S0165-2427(96)05717-0 [ Links ]

VahdaT Shariat Panahi A, Hultman P, Öllinger K, Westermark GT, Lundmark K. 2019. Lipid membranes accelerate amyloid formation in the mouse model of AA amyloidosis. Amyloid. 26(1): 34-44. https://doi.org/10.1080/13506129.2019.1576606 [ Links ]

Wekell P, Karlsson A, Fasth A, Berg S. 2016. Familjär medelhavsfeber - viktig sjukdom i en globaliserad värld - Särskilt vanlig hos personer från östra Medelhavsområdet [Familial Mediterranean fever - an important disease in a globalised world]. Lakartidningen. 113: DZFY. https://pubmed.ncbi.nlm.nih.gov/27551868/Links ]

Westermark P , Andersson A, Westermark GT. 2011. Islet amyloid polypeptide, islet amyloid, and diabetes mellitus. Physiological reviews. 91(3):795-826. https://doi.org/10.1152/physrev.00042.2009 [ Links ]

Westermark GT, Fändrich M, Lundmark K, Westermark P. 2018. Noncerebral Amyloidoses: Aspects on Seeding, Cross-Seeding, and Transmission. Cold Spring Harbor perspectives in medicine. 8(1): a024323. https://doi.org/10.1101/cshperspect.a024323 [ Links ]

Zhang B, Une Y, Fu X, Yan J, Ge F, Yao J, Sawashita J, Mori M, Tomozawa H, Kametani, F, Higuchi K. 2008. Fecal transmission of AA amyloidosis in the cheetah contributes to high incidence of disease. Proceedings of the National Academy of Sciences of the United States of America. 105(20): 7263-7268. https://doi.org/10.1073/pnas.0800367105 [ Links ]

Code: e2021-22

Received: April 15, 2021; Accepted: March 21, 2022

Creative Commons License Este es un artículo publicado en acceso abierto bajo una licencia Creative Commons