Open access peer-reviewed chapter - ONLINE FIRST

Clinical and Welfare Aspects of Immunosuppression in Poultry Farming

Written By

Amra Alispahic, Adis Softic, Aida Kustura, Jasmin Omeragic and Teufik Goletic

Submitted: 15 April 2024 Reviewed: 07 May 2024 Published: 10 June 2024

DOI: 10.5772/intechopen.115072

From Farm to Zoo - The Quest for Animal Welfare IntechOpen
From Farm to Zoo - The Quest for Animal Welfare Edited by Jaco Bakker

From the Edited Volume

From Farm to Zoo - The Quest for Animal Welfare [Working Title]

Dr. Jaco Bakker and Dr. Melissa Delagarza

Chapter metrics overview

15 Chapter Downloads

View Full Metrics

Abstract

Immunosuppression refers to a condition in which the body’s immune system becomes weakened or suppressed, making them more susceptible to infections, diseases and other health problems. Immunosuppression in poultry can be caused by a variety of factors, including infectious activity (viruses, bacteria, parasites, and fungi), environmental stress, malnutrition, and poor management. Viruses have the most dominant immunosuppressive effect on the poultry population, especially infectious bursitis virus, infectious anemia virus and Marek’s disease virus. Immunosuppression in poultry can have significant consequences on their health, welfare, and overall productivity. The biggest losses in poultry production are reflected in reduced growth and performance of individuals, reduced egg production and shell quality, increased morbidity and mortality rates, and greater susceptibility of immunocompromised individuals to secondary infections. Effective management strategies are key to minimizing the impact of immunosuppression in poultry. Biosecurity measures, strict hygiene protocols, immunoprophylaxis, control, and reduced transport of poultry and people, adequate diet, and correct husbandry and housing conditions are some of the factors that result in prevention and/or solvation of this problem.

Keywords

  • immunosuppression
  • poultry
  • welfare
  • immunity
  • tropism
  • infectious agents
  • stressors

1. Introduction

Animal welfare stands as a cornerstone principle across a multitude of domains, with particular significance in agricultural operations and food production systems [1]. At its core, ensuring the well-being of animals not only resonates with ethical imperatives but also serves as a linchpin for sustaining the health and productivity of livestock populations. Within the intricate tapestry of agricultural landscapes, where animals are raised to supply meat, dairy, and other essential products, maintaining optimal welfare conditions emerges as a linchpin for success [2]. These conditions foster an environment conducive to heightened animal health, diminished stress levels, and accelerated growth rates, culminating in more robust and resilient livestock populations. Furthermore, the promotion of poultry welfare assumes a critical role in mitigating the risk of immunosuppression within poultry populations. Optimal welfare conditions, characterized by spacious living environments, adequate nutrition, and minimal stressors, are paramount for maintaining robust immune function in poultry [3]. By prioritizing poultry welfare, farmers can bolster the resilience of their flocks against immunosuppressive factors, thereby reducing the incidence of infectious diseases and enhancing overall flock health. Additionally, promoting poultry welfare aligns with broader societal values regarding ethical animal treatment and sustainable food production. Consumers increasingly expect poultry products to be sourced from farms that prioritize animal welfare, underscoring the interconnectedness between animal welfare, immune health, and consumer preferences. Ultimately, by prioritizing both poultry welfare and immune health, agricultural practitioners not only safeguard the well-being of their flocks but also meet the expectations of conscientious consumers and uphold the integrity of the poultry industry [4]. Consumers are increasingly attuned to the conditions under which their food is produced, demanding transparency, accountability, and adherence to humane standards. By prioritizing animal welfare in agricultural practices and food production, producers not only align with evolving consumer preferences but also fortify the ethical foundation of their operations [5]. In essence, the pursuit of animal welfare in agricultural contexts transcends mere industry norms; it embodies a collective commitment to ethical stewardship, sustainable husbandry, and societal well-being. By championing the welfare of animals and elevating standards of care, agricultural stakeholders not only honor their moral obligation to sentient beings but also fortify the resilience and viability of our food systems. In doing so, they forge a path toward a future where compassion, sustainability, and societal values converge, ensuring the welfare and prosperity of both animals and consumers alike [6].

In poultry farming, the control of infectious diseases stands as a paramount concern in ensuring the production of healthy poultry flocks. This objective is typically achieved through comprehensive vaccination programs complemented by effective management practices, including rigorous biosecurity measures aimed at minimizing the risk of infection. The efficacy of vaccination programs hinges upon the birds’ capacity to mount a robust immune response, underscoring the foundational importance of immune function in poultry health. However, beyond the intrinsic capability of individual birds to initiate an immune response to natural antigens or those introduced via vaccination, numerous external factors exert influence over the level of immunity conferred [7]. It is the delicate interplay of these myriad factors, whether in excess or deficiency, that can precipitate the onset of immunosuppression.

The term ‘immunosuppression’ was first defined by Dohms and Saif as ‘a state of temporary or permanent dysfunction of the immune response resulting from an insult to the immune system and resulting in increased susceptibility to disease’ [8]. Lutticken further elaborated on this definition, suggesting that immunosuppression often manifests as a suboptimal antibody response, to which we add ‘suboptimal innate and cell-mediated responses’ [9]. In essence, poultry immunosuppression denotes a condition wherein the avian immune system becomes weakened or suppressed, rendering birds more susceptible to infections, diseases, and other health complications. This phenomenon holds profound significance within both the poultry industry and wild bird populations, as it can precipitate substantial economic losses and harbor detrimental biological and ecological ramifications. Avian immunosuppression can arise from a myriad of sources, including infectious agents, environmental stressors, malnutrition, and suboptimal management practices. Understanding the mechanisms underlying immunosuppression in birds, as well as its attendant consequences, is paramount for the formulation and implementation of effective prevention and control strategies [10]. Viral infections such as infectious bursal disease virus (IBDV), chicken infectious anemia virus (CIAV), and Marek’s disease virus (MDV) exemplify prominent immunosuppressive agents. These viruses exhibit a pronounced tropism toward the avian immune system, targeting the lymphoid tissue/cells and impeding the normal development and function of immune cells. Consequently, the bird’s capacity to mount an effective immune response against pathogens becomes compromised, heightening susceptibility to infections [11]. Environmental stressors, such as temperature fluctuations, overcrowding, poor ventilation, and hygiene deficiencies, also contribute significantly to the manifestation of immunosuppression in birds. Additionally, malnutrition, particularly deficiencies in essential vitamins and minerals, can compromise immune function by impairing cellular and humoral immunity [10]. The repercussions of impaired immune function in birds extend beyond mere health implications; they encompass reduced production efficiency, elevated mortality rates in commercial settings, and heightened antibiotic usage. While antibiotics may offer therapeutic benefits, their excessive use can engender antibiotic resistance, posing a formidable challenge to public health. Immunosuppression in wild bird populations can engender ecological ramifications, influencing disease dynamics and population dynamics within ecosystems [12]. A weakened immune system renders birds more susceptible to infections, resulting in elevated disease prevalence and severity, potential population declines, and altered interspecies interactions. Addressing avian immunosuppression necessitates a comprehensive and multifaceted approach encompassing robust biosecurity measures, enhanced management practices, and optimized nutrition to bolster immune function. Key interventions include ensuring optimal housing conditions, implementing stringent biosecurity protocols, and adhering to the ‘all in, all out’ principle in poultry production [13]. The recognition of the causes and consequences of immunosuppression in domestic poultry and wild birds is paramount to the implementation of proactive measures aimed at promoting bird health, welfare, and sustainable production practices. Resolving the issue of immunosuppression confers myriad benefits, ranging from the preservation of indigenous poultry and wild bird species to the mitigation of economic losses attributable to disease outbreaks. By fostering a deeper understanding of immunosuppression and its implications, stakeholders can work collaboratively to safeguard the well-being of birds across diverse environments. The aim of this book chapter is to elucidate the mechanisms of poultry immunosuppression and its profound implications for both poultry welfare and production.

Advertisement

2. Avian immune system

The evolution of birds and mammals from a common reptilian ancestor over 250 million years ago has resulted in distinct immune system adaptations reflecting their divergent ecological niches. While sharing fundamental immune protection mechanisms, avian and mammalian immune systems exhibit notable differences crucial for understanding avian immunology [14]. Mammals possess discrete lymphoid organs such as the thymus, spleen, and lymph nodes, facilitating adaptive immune responses. In contrast, birds lack distinct lymph nodes, with lymphoid tissue distributed peripherally and associated with mucosal surfaces, such as the conjunctiva and gastrointestinal tract [15]. In mammals, bone marrow serves as the primary site for B cell production, whereas birds rely on the bursa of Fabricius for B cell maturation, akin to mammalian bone marrow [16]. Birds exhibit a broader repertoire of antimicrobial peptides (AMPs) compared to mammals, including species-specific defensins that aid in pathogen neutralization [17]. Additionally, variations in the complement system, such as unique complement proteins in birds, contribute to pathogen recognition and elimination [18]. Birds produce IgY antibodies in the bursa of Fabricius, providing passive immunity to offspring through the yolk. Unlike mammals, birds lack IgA and IgE isotypes but possess γδ T cells instead of CD4+ T cells [19]. Birds exhibit cellular memory primarily mediated by γδ T cells and NK cells, ensuring rapid immune responses upon reinfection. However, humoral memory in birds is less pronounced compared to mammals [20]. Birds maintain higher body temperatures than mammals, creating less favorable conditions for pathogen growth and enhancing immune responses. The unique adaptations of avian immune systems reflect their specialized physiological and ecological requirements. Understanding these differences is pivotal for advancing knowledge in immunology and disease management across avian species [21].

Advertisement

3. Immunosuppression in poultry

3.1 Mechanisms of immunosuppression

Immunosuppression poses a significant challenge in the poultry industry, yet quantifying its prevalence remains elusive due to the complex interplay of various factors spanning physiological, environmental, and managerial stressors. Both acute and chronic stressors, including transportation, handling procedures, crowding, and abrupt environmental changes, disrupt the delicate equilibrium of the avian immune system, thereby impeding its ability to mount effective immune responses against pathogens and impacting the profitability of broiler flocks [22]. Similarly, pathogen-induced immunosuppression can occur independently of clinical disease, emphasizing the need for focused investigation into subclinical infections [22]. Infectious diseases constitute another pivotal determinant of immunosuppression in poultry. Pathogens such as viruses, bacteria, fungi, and parasites possess mechanisms to directly target and compromise immune cells, thereby impairing their functionality and rendering birds more susceptible to secondary infections [13]. Chronic infections exacerbate immunosuppression by perpetuating persistent inflammatory responses and depleting the host’s immune resources. Environmental variables, encompassing temperature fluctuations, inadequate ventilation, high humidity, and exposure to pollutants, significantly impact poultry immune competence. Suboptimal environmental conditions impose physiological stress on birds, disrupt metabolic homeostasis, and compromise immune function. For instance, extremes of temperature elevate metabolic demands, predisposing birds to thermal stress and concomitant immunosuppression [10]. Management practices exert a profound influence on the immunological integrity of poultry flocks. Poor nutrition, suboptimal vaccination strategies, inadequate biosecurity measures, and overcrowding contribute to immunosuppression. Nutritional deficiencies impede the synthesis of vital immune mediators, such as antibodies and cytokines, while substandard vaccination regimens leave birds vulnerable to preventable diseases. Furthermore, overcrowded housing conditions foster pathogen transmission and exacerbate stressors, exacerbating immunosuppression. Some of the key viral pathogens implicated in immunosuppression include Marek’s disease virus (MDV), although their immunosuppressive effects may be confounded by interactions with other pathogens such as Chicken Infectious Anemia Virus (CIAV) [7]. For instance, while CIAV primarily causes clinical disease in young chickens, its subclinical effects can result in profound immunosuppression, compromising the host’s ability to mount effective immune responses [7]. Infectious bursal disease virus (IBDV) also poses a threat by damaging immune tissues, particularly the bursa of Fabricius and associated lymphocytes, thereby impairing subsequent immunity [7]. However, caution is warranted when interpreting immunosuppressive effects attributed to certain viruses, as older studies may not delineate primary immunosuppression from synergistic effects with other pathogens [7]. For instance, early observations of hematopoietic system destruction in chickens with Marek’s disease were later found to be influenced by concurrent CIAV infection [23]. The exploration of immunosuppressive mechanisms has traditionally focused on well-documented factors such as stress-induced elevation of corticosteroids, programmed cell death pathways including apoptosis, necrosis, and pyroptosis, and alterations in immune response regulation mediated by viral infections. However, despite significant advancements, the intricate molecular interactions between viral proteins and host cells remain inadequately characterized in current scientific literature. This subchapter delves into the complexities of these mechanisms, shedding light on their nuanced roles in immunosuppression.

3.1.1 Immunosuppression induced by corticosteroids

Stress, whether acute or chronic, triggers a complex cascade of physiological and immunological responses aimed at adapting to challenging conditions and ensuring survival. In mammals, this response involves neuroendocrine signaling pathways, including the hypothalamus-pituitary-adrenal axis and the sympathetic nervous system, resulting in the secretion of glucocorticoids such as corticosterone [24]. These hormones modulate immune cell activities and cytokine production, influencing immune function [25]. However, the avian stress response differs due to unique immune system characteristics, including the absence of lymph nodes and a distinct repertoire of cytokines and chemokines [26]. In chickens, stress induces increased plasma corticosterone levels, impacting immune function through alterations in immune organ mass and cellular activity [27]. While stress is often associated with immunosuppression, some instances, particularly acute stress, may enhance immune functions, resulting in improved resistance to certain pathogens [28, 29]. Therefore, experiments with exogenous corticosterone administration in chickens reveal dynamic immune responses, including leukocyte redistribution and altered cytokine expression [30]. These observations underscore the intricate interplay between stress, corticosterone, and immune function in poultry, highlighting the need for further investigation into the nuanced effects of stress on avian immunity.

3.1.2 Apoptosis, necrosis, and pyroptosis

Viral infections often trigger programmed cell death (PCD), which can either benefit or harm the host depending on the timing of cell death relative to the viral replication cycle [31]. In lymphoid cells, such as those infected with infectious bursal disease virus (IBDV), chicken infectious anemia virus (CIAV), or Marek’s disease virus (MDV), viral replication-induced PCD can lead to (sub)clinical immunosuppression. PCD encompasses three main mechanisms: apoptosis, necroptosis, and pyroptosis, each with distinct characteristics [32]. Apoptosis, the best-understood form of PCD, can be initiated via intrinsic or extrinsic pathways, resulting in controlled cell death. While necroptosis and pyroptosis are inflammatory processes triggered by external and internal stimuli, they differ in their cellular and molecular mechanisms [32]. Although necroptosis and pyroptosis have not yet been identified as primary mechanisms of viral insult in avian infections, apoptosis induced by viral replication remains a significant contributor to cell death in avian viral infections, as observed in studies of IBDV and chicken infectious anemia virus [33, 34]. Understanding the intricacies of viral-induced PCD is crucial for deciphering the pathogenesis of avian viral infections and developing targeted therapeutic interventions [35].

3.2 Causes of immunosuppression in poultry farming

Immunosuppression in poultry farming arises from a complex interplay of factors spanning management practices, environmental conditions, infectious agents, and physiological stressors [36]. These causes, individually and collectively, contribute to the impairment of the avian immune systems function, thereby impacting the health and welfare of poultry populations. Suboptimal management practices, including overcrowding, inadequate ventilation, and lapses in biosecurity, create stressful conditions within poultry production systems. Overcrowding fosters social stress and competition for resources, while poor ventilation facilitates the accumulation of airborne pathogens and pollutants. Inadequate biosecurity measures heighten the risk of introducing infectious agents into the flock, further compromising immune health [37]. Poor nutrition is a significant contributor to immunosuppression in poultry. Diets lacking essential nutrients, such as vitamins, minerals, and amino acids, impair immune function and compromise the birds ability to mount effective immune responses against pathogens. Imbalances or deficiencies in the diet exacerbate the susceptibility of poultry to infectious diseases [38]. Poultry are susceptible to a wide array of infectious agents, including viruses, bacteria, fungi, and parasites, which directly target immune cells, compromising their function and resulting in immunosuppression. Disease outbreaks within flocks can significantly impact immune health and productivity [22]. Environmental stressors, such as temperature fluctuations, high humidity, and exposure to toxins, induce physiological stress in poultry. Chronic stress alters hormone levels and cytokine production, suppressing immune function and increasing susceptibility to infections. Environmental stressors exacerbate the impact of infectious diseases on poultry health [37]. Inappropriate vaccination protocols can compromise immune function in poultry [36]. Failure to administer vaccines effectively or the use of vaccines with limited efficacy leaves birds vulnerable to infections. Conversely, over-vaccination or vaccination during periods of stress can induce immunosuppression and compromise immune responses. Genetic selection for traits such as rapid growth and high productivity can inadvertently impact immune function in poultry. Birds bred for these traits may exhibit compromised immune systems, increasing their susceptibility to infections and immunosuppression [36].

3.2.1 Heat stress

Heat stress poses a significant challenge in the poultry industry, exerting profound effects on both the health and reproductive performance of poultry [39]. With annual economic losses ranging from 128 to 165 million dollars since 2003, exacerbated by escalating global temperatures, its impact is poised to escalate further [40]. Defined as the inability of chickens to maintain thermal equilibrium amidst environmental heat loads, heat stress arises from a complex interplay of factors including ambient temperature, humidity, heat radiation, and air velocity, with elevated ambient temperature playing a pivotal role [41]. Chickens exhibit optimal growth within a thermoneutral temperature range of 18–21°C, with any ambient temperature surpassing 25°C inducing heat stress [42]. Beyond compromising immune function, heat stress disrupts various physiological processes, manifesting in increased feed intake coupled with reduced growth rates and egg production [43]. In addition, heat stress triggers the activation of the sympathetic pituitary-adrenal axis, resulting in elevated plasma corticosterone levels via the hypothalamic-pituitary-adrenal (HPA) axis [41, 43]. Immune suppression due to thermal stress primarily manifests as regression in immune organs such as the spleen, thymus, and lymphatic tissues. This decline is evident in reduced total white blood cell (WBC) counts and antibody levels, along with elevated heterophils to lymphocytes ratio (H/L) in immunocompromised poultry [44].

3.2.2 Oxidative stress

Oxidative stress arises from an imbalance between the production and elimination of reactive oxygen species (ROS), which are free radicals and peroxides generated within cells during routine metabolism. While ROS plays vital roles in cellular functions like cytokine transcription, immunomodulation, and ion transport, their surplus can overwhelm cellular detoxification mechanisms, resulting in oxidative stress [45]. Excessive ROS inflicts damage upon various cellular components, including proteins, lipids, and DNA, resulting in reversible alterations or, under severe oxidative stress, apoptosis and cell demise [46].

3.2.3 Acid-base disbalance

The absence of sebaceous glands and feather covering in birds compromises their thermoregulation, necessitating active heat dissipation methods such as panting [47]. Panting involves increased respiratory frequency and evaporative cooling through open-beak breathing. This rapid exhalation of carbon dioxide (CO2) during panting alters the standard bicarbonate buffering system in the blood. Hypocapnia results in reduced concentrations of carbonic acid (H2CO3) and hydrogen ions (H+) in the bloodstream, while bicarbonate ion (HCO3−) concentrations increase, causing blood alkalinity [48]. To counteract this, birds eliminate excess bicarbonate ions and retain hydrogen ions through renal mechanisms to maintain normal blood pH. Elevated H+ ion levels disrupt the acid-base balance, resulting in respiratory alkalosis and metabolic acidosis, ultimately contributing to decreased production performance due to resultant immunosuppression [47].

3.2.4 Coccidia-induced immunosuppression

Avian coccidiosis, caused by Eimeria spp., poses a significant threat to the poultry industry due to its adverse economic impact. These intracellular protozoan parasites target intestinal epithelial tissues, resulting in a range of clinical manifestations, including bloody diarrhea and reduced body weight, resulting in substantial economic losses [49]. The complex life cycle of coccidia involves both intracellular and extracellular stages, with rapid reproduction resulting in the production of infective oocysts that enhance parasite spread [50]. Poultry develop immunity against coccidiosis through both innate and adaptive responses, with innate immunity triggered by pattern recognition receptors like toll-like receptors, resulting in immune cell activation and cytokine production [51]. Adaptive immunity, involving B and T lymphocytes, plays a crucial role in antigen-specific responses to prevent pathogen colonization [51]. While B cell depletion studies suggest antibodies may not be essential for anticoccidial immunity, passively transferred humoral immunity remains significant [52]. Cell-mediated immunity, mediated by T cells, particularly CD4+ T helper cells and CD8+ cytotoxic T lymphocytes, is vital for protection against Eimeria spp. infections [53]. Further research is needed to elucidate the role of various T cell subpopulations in avian coccidiosis for the development of effective control strategies.

Treg cells, a subset of T lymphocytes, play a crucial role in immunosuppression. In mammals, Treg cells exhibit a CD4+CD25+FoxP3+ phenotype [54]. Although the avian FoxP3 ortholog has not been identified, CD4+CD25+ T lymphocytes in chickens act as Treg cells, suppressing activated immune cells and producing IL-10, TGF-β, CTLA-4, and LAG-3. IL-10, significantly upregulated in CD4+CD25+ cells, plays a pivotal role in evading the host’s immune response during coccidiosis pathogenesis, aiding parasite invasion and survival by inhibiting the IFN-γ-associated Th1 response. Differential expression of IL-10 in chicken genetic lines suggests its crucial role in modulating immune responses to Eimeria spp. infection [55]. Neutralization of IL-10 in Eimeria-infected poultry improves growth rates, highlighting its involvement in coccidiosis pathogenesis. Treg cells also mitigate pathological intestinal changes induced by Eimeria spp. by suppressing Th17 cells, reducing parasite pathogenicity [56]. The interplay between Treg and Th17 cells during immunoinflammatory events is critical in determining the outcome of Eimeria parasite infestation [57].

Two groups of coccidial species have been associated with immunosuppression, namely Cryptosporidium baileyi and several species of the genus Eimeria, although the evidence for the immunosuppressive effect of these parasites is rather limited. Cryptosporidium baileyi replicates in the epithelial cells of the bursa of Fabricius and the respiratory tract of chickens, and the prevalence of this disease is probably significantly higher than the number of diagnosed cases indicates [57]. Oral inoculation of high infectious doses of C. baileyi to young commercially bred poultry caused histopathological changes in the epithelium and interfollicular tissue of the bursa of Fabricius and reduced the production of specific antibodies, but also the production of antigen-dependent and independent T lymphocytes. Antibody titers in the serum of chickens after vaccination against infectious bronchitis virus (IBV), Newcastle disease virus (NDV), and avian influenza virus (AIV) were reduced after the experimental inoculation of C. baileyi chickens [58]. However, infection with C. baileyi did not increase the incidence of Marek’s disease or affect the efficacy of the Marek’s disease virus (MDV) vaccine strain CVI988. Also, these authors found that Marek’s disease, which appeared before or after infection/infestation with C. baileyi, leads to its worsening. In addition, MDV vaccination administered 4 days post-infection with C. baileyi caused respiratory lesions at 6 days post-vaccination.

3.2.5 Virus-induced immunosuppression

Infectious bursal disease virus (IBDV), a member of the Birnaviridae family, comprises two serotypes, with only serotype 1 inducing immunosuppression and disease in chickens. Various pathotypes within serotype 1 exhibit differing virulence levels. Genetic grouping of serotype 1 strains reveals distinct variations based on sequencing techniques. The IBDV genome consists of double-stranded (ds)RNA segments A and B, encoding viral proteins crucial for pathogenicity [59]. Serotype 1 strains predominantly replicate in B cells expressing chB6 and surface immunoglobulin, resulting in bursal atrophy and apoptosis [60]. Early infection reduces B cell populations, affecting primary antibody responses, while recovery may take up to seven weeks post-infection [61]. IBDV-induced immunosuppression compromises vaccine efficacy against avian influenza A virus and Campylobacter jejuni bacterial colonization. Furthermore, highly virulent IBDV strains (serotype 1) alter cecal microbiota, although the significance of this alternation remains unclear [62]. The VP5 protein of IBDV plays a role in pathogenesis, modulating apoptosis and survival in infected cells [31, 62]. IBDV infection induces complex interactions between viral proteins and B cells, resulting in apoptosis and subsequent recovery [63]. Post-recovery, two types of follicles emerge, indicating diverse immune responses [64]. Besides B cells, IBDV targets macrophages and T cells, contributing to immunopathogenesis [65]. Vaccination with live attenuated strains controls IBDV, but residual pathogenicity and vaccine-induced immunosuppression pose challenges [66]. Overall, IBDV infection triggers intricate immune responses involving B cells, macrophages, and T cells, resulting in bursal destruction and long-term immune suppression.

Chicken infectious anemia virus (CIAV), a small DNA virus approximately 25 nm in size, belongs to the genus Gyrovirus, family Anelloviridae, boasting a single-stranded covalently closed DNA genome encoding three proteins [67]. It has remarkable resistance to disinfectants and heat treatment, making it a common contaminant in chicken flocks, complicating studies on the immunosuppressive properties of other pathogens, especially in vaccine production [68]. Vertical transmission during egg production or horizontal transmission within the first few weeks of age can result in clinical disease, although most chicks are protected by maternal antibodies. Infections occurring in animals older than three weeks are often subclinical but can induce significant immunosuppression [69]. CIAV primarily targets rapidly dividing cells, utilizing them for viral DNA replication. Infected cells include hemocytoblasts in the bone marrow, T cell precursors in the thymus, or antigen-stimulated T cells. Hemocytoblast infection results in decreased erythrocyte, platelet, and granulocyte numbers, increasing susceptibility to secondary bacterial infections [67]. CIAV-induced immunosuppression is associated with reduced cytotoxic T lymphocyte (CTL) responses, facilitating prolonged viral replication [70]. The virus also affects CD4+ and CD8+ cell populations, particularly during co-infections with other pathogens [71, 72]. Furthermore, CIAV has been linked to reduced local antibody responses to other viral infections, such as infectious bronchitis virus (IBV) and avian influenza viruses [73, 74]. Studies on cytokine modulation during CIAV infection remain limited but suggest alterations in Th1, Th2, and Treg-associated cytokines [75]. Early changes in cytokine expression are crucial for understanding subsequent immunosuppression and require further investigation [69].

Marek’s disease virus (MDV), scientifically known as Gallid herpesvirus 2 and classified under the genus Mardivirusin the subfamily Alphaherpesvirinae, induces T lymphocyte tumors in chickens. The advent of highly effective vaccines against MDV in the early 1970s marked a significant milestone in veterinary virology, but subsequent evolution of the virus into highly virulent strains has posed new challenges [76]. These highly virulent (vv) pathotypes not only increase tumor incidence but also induce severe damage to lymphoid organs, potentially exacerbating immunosuppression [77]. The immunosuppression associated with MDV manifests in two phases: an early phase characterized by lymphocyte destruction within lymphoid organs and a late phase marked by virus reactivation with or without tumor development [78]. During the early phase, severe atrophy of the thymus and bursa of Fabricius occurs within the first 2 weeks of infection, with implications for long-term immunity [78]. The pathogenesis model of Marek’s disease elucidates how the virus targets B and T cells, causing depletion in lymphoid populations [79, 80]. The cytokine IFN-γ is upregulated as early as 3 days post-infection, possibly enhancing IL-8 receptor expression and facilitating viral transfer from B to T cells [72, 81]. Enhanced vIL-8 production by vv+ strains promotes cytolytic infection, exacerbating lymphoid damage [82]. Notably, vv+ MDV induces a significant upregulation of proinflammatory cytokines, potentially exacerbating immunosuppression [83]. Apoptosis plays a pivotal role in lymphocyte depletion during MDV infection, affecting both thymocytes and peripheral T cells [84, 85]. While MDV primarily infects B cells during early cytolytic infection, subsequent effects on thymocytes remain incompletely understood [86]. Several MDV gene products, including LORF4, VP23, and Meq, contribute to lymphoid atrophy, underscoring the multifaceted nature of MDV-induced immunosuppression [87]. MDV-induced immunosuppression extends beyond lymphoid tissue, affecting macrophage activation and cytokine regulation [88]. The inhibition of CTL migration and reduction of CD8 glycoprotein levels in feather follicle epithelium highlight the complex interplay between the virus and host immunity [89]. Moreover, MDV vaccine strains, while crucial for disease prevention, can paradoxically induce immunosuppression, impacting host responses to secondary infections [89]. Understanding these dynamics is essential for optimizing vaccine strategies and mitigating the impact of MDV-induced immunosuppression in commercial poultry production [88].

3.2.6 Mycotoxin-induced immunosuppression

Derived from ‘mykes’ (fungi) and ‘toxicon’ (poison), mycotoxins are toxic byproducts of mold metabolism with a low molecular weight [90]. Produced by a limited number of molds, some molds can synthesize multiple mycotoxin types, while others produce a single type [91]. Commonly found in mold-contaminated animal feed, mycotoxins pose a significant risk to animal health due to their widespread presence in feed, including poultry feed [92]. Approximately 25% of cereal, rice, and nut crops worldwide are contaminated by molds and fungi, raising concerns for human and animal health [93]. Therefore, mycotoxins such as aflatoxins, ochratoxins, and deoxynivalenol (DON) have detrimental effects on the immune system of farm animals.

Mycotoxins, derived from mold metabolism, exhibit a peculiar dual effect on immunity. At low concentrations (<5 mg/kg of food), they stimulate immune responses, while higher concentrations suppress immunity [92]. Chronic exposure to elevated levels of mycotoxins like DON damages immune organs and gastrointestinal mucosa [94]. This dual immune modulation, termed hormesis, involves mechanisms such as oxidative stress, apoptosis, and autophagy of immune cells, alongside regulation of immune signaling pathways including ERK 1/2, P38, mitogen-activated protein kinases (MAPK), JNK-STAT1, and MyD88-dependent toll-like receptor (TLR) pathways [91]. In addition, mycotoxins impact dendritic cells, T- and B-lymphocytes, and monocytes, influencing the secretion of pro- and anti-inflammatory cytokines and the differentiation of macrophages. Additionally, they directly activate JNK-STAT1 and MyD88-dependent TLR signaling pathways, inducing immunosuppressive and inflammatory responses [91].

Aflatoxin, primarily produced by Aspergillus molds like A. flavus and A. parasiticus, encompasses various types, with AFB1 being the most toxic and carcinogenic. Widely found in moldy agricultural products, especially animal feed ingredients, AFB1 poses significant health risks to livestock and poultry [95]. Its mechanism of action involves DNA damage, oxidative stress, apoptosis, hepatotoxicity, and nephrotoxicity [95, 96]. Bioactivation by cytochrome P450 in the liver transforms AFB1 into its highly toxic component, aflatoxin-8,9-epoxide, which binds to mitochondrial or nucleic DNA, resulting in cytotoxicity upon hydrolysis [96]. In poultry, AFB1 induces cell death in primary lymphoid organs, compromises intestinal integrity, and reduces production performance, with effects on both humoral and cell-mediated immunity [31, 97]. While AFB1 may initially elevate antibody levels, chronic exposure results in reduced immune function in hatchlings [98]. AFB1 can bypass primary immune responses, penetrating poultry organisms and compromising intestinal innate immunity [97]. The discovery of aflatoxin and ochratoxin elevated awareness among both the scientific community and the public about the potential dangers posed by mycotoxins [91]. Ochratoxins, produced by Penicillium and Aspergillus fungi, particularly ochratoxin A (OTA), emerged as significant contributors to diseases in human and veterinary medicine [99]. OTA exhibits nephrotoxic, hepatotoxic, teratogenic, and immunotoxic effects in poultry [99, 100]. Absorption of OTA primarily occurs in the glandular stomach and proximal jejunum of poultry, with unabsorbed OTA interacting with intestinal microbiota in the large intestine [101]. OTA, even present in chicken eggs from OTA-fed hens, reduces both innate and acquired immune responses, increasing susceptibility to diseases. Notably, OTA supplementation in feed significantly increases chicken mortality caused by Salmonella enterica subsp. enterica serovar Gallinarum [91].

DON, a significant trichothecene mycotoxin produced by Fusarium molds, is prevalent in various grains like wheat, rye, barley, and oats. Experimental studies in pigs, poultry, and cell culture models have documented DON’s adverse effects on immune function [101]. DON’s impact on the immune system varies from immunosuppression to immunostimulation, influenced by concentration, duration, and timing of exposure [94]. Like other trichothecenes, DON inhibits host protein biosynthesis by binding to the 60S ribosomal subunit, inducing metabolic stress and activating mitogen-activated protein kinases (MAPK) in poultry. Elevated prostaglandin levels following cyclooxygenase 2 (COX2) induction further exacerbate immune responses via increased expression of nuclear factor κB (NF-κB) and proinflammatory cytokines [94].

Experimental evidence suggests DON’s role in dysregulating cell signaling pathways and immune reaction genes, potentially predisposing poultry (and wild birds) to infectious diseases [102]. DON suppresses antibody responses to infectious bronchitis vaccine (IBV) and Newcastle disease virus in broilers and layers, while also reducing tumor necrosis factor-alpha (TNF-α) concentration in broiler plasma, indicative of weakened immune function and increased susceptibility to infectious diseases [103]. Additionally, DON adversely affects intestinal morphology, electrophysiology, absorption, and barrier function in chickens [104].

Beyond DON, the Fusarium genus produces several mycotoxins, including fumonisin B1, T-2 toxin, and zearalenone (ZEA), each with immunotoxic implications for chickens, turkeys, and ducks [105]. Fumonisin B1, even at low concentrations, reduces macrophage activity, antibody responses to vaccines, and gene expression of interleukins in broilers without affecting growth performance [106]. T-2 toxin administration reduces vaccine-induced protection against Marek’s disease virus in chickens. DON inhibits DNA, RNA, and protein synthesis, affecting tissues with rapid turnover, like intestinal epithelial cells and immune organs [104]. While low doses of T-2 and DON enhance antibody titers against Newcastle disease virus (NDV), high doses induce immunosuppression [107]. Zearalenone, a nonsteroidal estrogenic mycotoxin, binds to estrogen receptors, potentially interacting with chicken infectious anemia virus due to shared receptor affinity [104]. Interactions between trichothecenes and poultry immune responses are intricate, with low doses often stimulating cytokine production while high doses induce apoptosis in immune cells. Combined mycotoxin exposure exacerbates immunosuppressive effects, complicating the interpretation of immune dysfunctions in commercial flocks [108].

Advertisement

4. Impacts of intensive farming on poultry welfare and immunosuppression

Intensive farming methods wield a significant influence on poultry welfare, exacerbating immunosuppression and compromising the health and well-being of birds [109]. Central to this issue are the stressors and constraints inherent in high-density production systems. Overcrowding, a hallmark of intensive farming, imposes profound social stress on poultry populations. Densely packed housing conditions foster heightened competition for resources, escalating aggression among birds and inducing physiological responses that undermine immune function, thereby heightening susceptibility to infectious diseases [110]. Inadequate ventilation compounds these challenges, compromising air quality within intensive production facilities. The accumulation of airborne pollutants, including dust, ammonia, and microbial contaminants, irritates the respiratory tract and compromises lung function. Respiratory distress, a consequence of poor ventilation, predisposes birds to respiratory infections, exacerbating the burden of immunosuppression within poultry populations [111]. Moreover, the high stocking densities characteristic of intensive farming environments foster rapid pathogen transmission. Close confinement facilitates the spread of infectious agents through various modes, including direct contact, airborne transmission, and contaminated surfaces. Consequently, disease outbreaks are not only frequent but also severe, imposing significant immunosuppressive burdens on poultry populations [112]. The stressful environment further compounds these challenges, as chronic stressors associated with confinement, artificial lighting, and limited behavioral opportunities disrupt physiological homeostasis and compromise immune function. Stress-induced immunosuppression renders birds more susceptible to infections, exacerbating welfare concerns in intensive production systems. Additionally, intensive farming practices may prioritize production efficiency over optimal nutrition, potentially compromising immune function due to inadequate nutrient provision. Diets formulated to meet basic nutritional requirements may lack essential immune-supporting nutrients, further exacerbating immunosuppression in intensive rearing of poultry.

Advertisement

5. Evaluation techniques for assessing and monitoring immunosuppression in poultry

Assessing and monitoring immunosuppression in poultry is paramount for ensuring the health and welfare of flocks. A diverse array of scientific methodologies is employed to comprehensively evaluate the immune status of poultry populations, encompassing diagnostic tests, clinical observation, and welfare assessment. Laboratory assays are pivotal in assessing immune function in poultry. Parameters such as leukocyte counts and serum protein levels provide valuable insights into immune status. Additionally, specific assays like ELISA and PCR enable the detection of antibodies or pathogens associated with immunosuppressive diseases [4]. Observation of clinical signs serves as a primary method for gauging immunosuppression in poultry. Indicators such as increased morbidity and mortality rates, reduced growth rates, and poor feed conversion efficiency signify compromised immune function. Monitoring temporal changes in these parameters offers valuable insights into flock immune status [113]. Evaluating welfare indicators yields crucial information about the overall health and well-being of poultry. Behavioral alterations, including heightened lethargy and altered feeding behaviors, may indicate immunosuppression. Furthermore, monitoring environmental parameters such as air quality, temperature, and humidity aids in identifying stressors that compromise immune function [114]. Post-mortem examination and histopathological analysis of tissues provide valuable insights into the immunological status of poultry. Histopathological lesions such as lymphoid depletion and inflammation serve as indicators of underlying immune dysfunction [115]. Specialized immunological assays, including lymphocyte proliferation assays and cytokine profiling, offer quantitative assessments of cellular and humoral immune responses in poultry. These assays elucidate the functional capacity of immune cells and the production of immune mediators, providing comprehensive insights into immune function [116].

Advertisement

6. Strategies for preventing and mitigating immunosuppression in poultry

Mitigating immunosuppression in poultry is paramount for safeguarding flock health and welfare. Employing effective strategies grounded in scientific principles can substantially reduce the prevalence and severity of immunosuppressive conditions. Improving housing environments is crucial for minimizing stressors that contribute to immunosuppression. This involves optimizing ventilation systems to ensure optimal air quality and reducing stocking densities to alleviate social stress. Providing adequate space allocation, comfortable bedding materials, and environmental enrichment are also vital for promoting physiological and behavioral well-being [117]. Proper nutrition is fundamental for supporting immune function in poultry. Formulating diets tailored to meet the specific nutritional requirements of birds at various life stages and production phases is essential. Supplementation with micronutrients, antioxidants, and immunomodulatory additives can enhance immune responses and mitigate immunosuppressive effects. Quality assurance in feed formulation and ensuring access to clean water sources are also critical components of nutritional optimization strategies [118]. Robust biosecurity measures are indispensable for preventing the introduction and spread of pathogens associated with immunosuppression. This includes stringent control of personnel, equipment, and vehicle movements, as well as the implementation of disinfection and pest control protocols. Comprehensive biosecurity programs encompassing flock isolation, quarantine procedures, and regular health monitoring are essential for safeguarding flock health and mitigating disease risks [119]. Vaccination plays a pivotal role in disease prevention in poultry production. Tailoring vaccination programs to the specific disease challenges faced by the flock is crucial. Regular evaluation of vaccine efficacy, adjustment of vaccination protocols based on epidemiological data, and adherence to strict vaccination schedules are critical for ensuring robust immune protection and reducing immunosuppressive risks [120]. Regular monitoring and surveillance of flock health are essential for early detection of immunosuppressive conditions. This encompasses routine health assessments, diagnostic testing, and surveillance for emerging diseases. Timely identification of immunosuppressive factors facilitates prompt intervention and mitigation strategies, minimizing disease outbreaks and welfare impacts [121].

Advertisement

7. Conclusion

This chapter provides a comprehensive exploration of the intricate relationship between poultry welfare, immunosuppression, and alternative production systems. Key points discussed underscored the profound impact of housing conditions, nutrition, stressors, and disease exposure on immune function and overall welfare in poultry farming. Additionally, the potential benefits of alternative production systems, such as free-range and organic farming, in promoting poultry welfare and reducing immunosuppression are elucidated, highlighting their alignment with principles of animal welfare and sustainable production practices. Throughout the discourse, emphasis was placed on scientific evidence and best practices, emphasizing the necessity of evidence-based approaches in addressing poultry welfare and immunosuppression challenges. By integrating scientific knowledge with practical management strategies, producers can optimize flock health, welfare, and productivity while minimizing adverse environmental impacts. Moreover, the discussion touched upon the growing consumer demand for ethically produced poultry products and its influence on market trends. Consumer preferences for products from alternative production systems underscore the economic viability of sustainable poultry production practices and the importance of aligning with consumer values to ensure market competitiveness. The insights gleaned extend beyond poultry production to encompass the broader spectrum of animal agriculture and welfare across diverse settings, including zoos and captive animal facilities. Prioritizing animal welfare, implementing evidence-based practices, and fostering consumer awareness are paramount in promoting a more sustainable and ethical approach to animal husbandry, from farm to zoo. This holistic perspective emphasizes the interconnectedness of animal welfare considerations and underscores the importance of collaborative efforts to enhance the well-being of animals across various contexts.

Advertisement

Acknowledgments

We would like to acknowledge that this book chapter is based on the graduation thesis of the first author (Amra Alispahic, DVM). In addition, we acknowledge the use of ChatGPT, an AI language model developed by OpenAI, which capabilities were useful in enhancing the clarity of the manuscript.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Phillips CJC. Farm animal welfare-from the farmers’ perspective. Animals. 2024;14:671. DOI: 10.3390/ani14050671
  2. 2. Buller H, Blokhuis H, Jensen P, Keeling L. Towards farm animal welfare and sustainability. Animals. 2018;8:81. DOI: 10.3390/ani8060081
  3. 3. Abo-Al-Ela HG, El-Kassas S, El-Naggar K, Abdo SE, Jahejo AR, Al Wakeel RA. Stress and immunity in poultry: Light management and nanotechnology as effective immune enhancers to fight stress. Cell Stress & Chaperones. 2021;26:457-472. DOI: 10.1007/s12192-021-01204-6
  4. 4. Wlaźlak S, Pietrzak E, Biesek J, Dunislawska A. Modulation of the immune system of chickens a key factor in maintaining poultry production-a review. Poultry Science. 2023;102:102785. DOI: 10.1016/j.psj.2023.102785
  5. 5. Sinclair M, Fryer C, Phillips CJC. The benefits of improving animal welfare from the perspective of livestock stakeholders across Asia. Animals. 2019;9:123. DOI: 10.3390/ani9040123
  6. 6. Alonso ME, González-Montaña JR, Lomillos JM. Consumers’ concerns and perceptions of farm animal welfare. Animals. 2020;10:385. DOI: 10.3390/ani10030385
  7. 7. Schat KA, Skinner MA. Chapter 16-Avian immunosuppressive diseases and immunoevasion. In: Schat KA, Bernd K, Pete K, editors. Avian Immunology. 2nd edn. Academic Press; 2014. pp. 275-297. ISBN 9780123969651. doi: 10.1016/B978-0-12-396965-1.00016-9. Available from: https://www.sciencedirect.com/science/article/pii/B9780123969651000169
  8. 8. Dohms JE, Saif YM. Criteria for evaluating immunosuppression. Avian Diseases. 1984;28:305-310
  9. 9. Lütticken D. Viral diseases of the immune system and strategies to control infectious bursal disease by vaccination. Acta Veterinaria Hungarica. 1997;45:239-249
  10. 10. Wasti S, Sah N, Mishra B. Impact of heat stress on poultry health and performances, and potential mitigation strategies. Animals. 2020;10:1266. DOI: 10.3390/ani10081266
  11. 11. Gimeno IM, Schat KA. Virus-induced immunosuppression in chickens. Avian Diseases. 2018;62:272-285. DOI: 10.1637/11841-041318-Review.1
  12. 12. Fritzsche McKay A, Hoye BJ. Are migratory animals superspreaders of infection? Integrative and Comparative Biology. 2016;56:260-267. DOI: 10.1093/icb/icw054
  13. 13. Davison TF, Kaspers B, Schat KA. Avian Immunology. United Kingdom, San Diego, United States, Kidlington, Oxford: Elsevier/Academic Press; 2008. DOI: 10.1016/B978-0-12-370634-8.50032-9
  14. 14. Kaspers B, Schat AK, Göbel T, Vervelde L. Avian Immunology. United Kingdom, San Diego, United States, Kidlington, Oxford: Elsevier/Academic Press; 2021. 978-0-12-818708-1
  15. 15. Hegde SN, Rolls BA, Turvey A, Coates ME. Influence of gut microflora on the lymphoid tissue of the chicken (Gallus domesticus) and Japanese quail (Coturnix coturnix japonica). Composite Biochemical Physiology A Molecular Integrated Physiology. 1982;72:205-209. DOI: 10.1016/0300-9629(82)90034-2
  16. 16. Glick B, Chang TS, George JR. The Bursa of Fabricius and antibody production. Poultry Science. 1956;35:224-225. DOI: 10.3382/ps.0350224
  17. 17. Ebers KL, Zhang CY, Zhang MZ, Bailey RH, Zhang S. Transcriptional profiling avian beta-defensins in chicken oviduct epithelial cells before and after infection with Salmonella enterica serovar Enteritidis. BMC Microbiology. 2009;9:153. DOI: 10.1186/1471-2180-9-153
  18. 18. Farsang A, Bódi I, Fölker O, Minko K, Beneyda Z, Balint A, et al. Avian coronavirus infection induces mannose-binding lectin production in dendritic cell precursors of chicken lymphoid organs. Acta Veterinaria Hungarica. 2019;67:183-196. DOI: 10.1556/004.2019.020
  19. 19. Walliser I, Göbel TW. Chicken IL-17A is expressed in αβ and γδ T cell subsets and binds to a receptor present on macrophages, and T cells. Developmental and Comparative Immunology. 2018;81:44-53. DOI: 10.1016/j.dci.2017.11.004
  20. 20. Bourgeon S, Guindre-Parker S, Williams TD. Effects of sibling competition on growth, oxidative stress, and humoral immunity: A two-year brood-size manipulation. Physiological and Biochemical Zoology. 2011;84(4):429-437. DOI: 10.1086/661080
  21. 21. Kobayashi KS, Chamaillard M, Ogura Y, Henegariu O, Inohara N, Nuñez G, et al. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science. 2005;307:731-734. DOI: 10.1126/science.1104911
  22. 22. Hoerr FJ. Clinical aspects of immunosuppression in poultry. Avian Diseases. 2010;54:2-15. DOI: 10.1637/8909-043009-Review.1
  23. 23. Jakowski RM, Fredrickson TN, Chomiak TW, Luginbuhl RE. Hematopoietic destruction in Marek’s disease. Avian Diseases. 1970;14(2):374-385
  24. 24. Lin H, Sui SJ, Jiao HC, Buyse J, Decuypere E. Impaired development of broiler chickens by stress mimicked by corticosterone exposure. Composite Biochemical Physiology A Molecular Integrated Physiology. 2006;143(3):400-405. DOI: 10.1016/j.cbpa.2005.12.030
  25. 25. Besedovsky H, del Rey A. Regulating inflammation by glucocorticoids. Nature Immunology. 2006;7:537. DOI: 10.1038/ni0606-537
  26. 26. Kaiser P, Poh TY, Rothwell L, Avery S, Balu S, Pathania US, et al. A genomic analysis of chicken cytokines and chemokines. Journal of Interferon & Cytokine Research. 2005;25(8):467-484. DOI: 10.1089/jir.2005.25.467
  27. 27. Post J, Rebel JM, ter Huurne AA. Physiological effects of elevated plasma corticosterone concentrations in broiler chickens. An alternative means by which to assess the physiological effects of stress. Poultry Science. 2003;82(8):1313-1318. DOI: 10.1093/ps/82.8.1313
  28. 28. Mumma JO, Thaxton JP, Vizzier-Thaxton Y, Dodson WL. Physiological stress in laying hens. Poultry Science. 2006;85(4):761-769. DOI: 10.1093/ps/85.4.761
  29. 29. Puvadolpirod S, Thaxton JP. Model of physiological stress in chickens 1. Response parameters. Poultry Science. 2000;79(3):363-369. DOI: 10.1093/ps/79.3.363
  30. 30. Shini S, Shini A, Kaiser P. Cytokine and chemokine gene expression profiles in heterophils from chickens treated with corticosterone. Stress. 2010;13(3):185-194. DOI: 10.3109/10253890903144639
  31. 31. Zhou X, Jiang W, Liu Z, Liu S, Liang X. Virus infection and death receptor-mediated apoptosis. Viruses. 2017;9(11):316. DOI: 10.3390/v9110316
  32. 32. Orzalli MH, Kagan JC. Apoptosis and necroptosis as host defense strategies to prevent viral infection. Trends in Cell Biology. 2017;27(11):800-809. DOI: 10.1016/j.tcb.2017.05.007
  33. 33. Liu M, Vakharia VN. Nonstructural protein of infectious bursal disease virus inhibits apoptosis at the early stage of virus infection. Journal of Virology. 2006;80(7):3369-3377. DOI: 10.1128/JVI.80.7.3369-3377.2006
  34. 34. Noteborn MH. Chicken anemia virus induced apoptosis: Underlying molecular mechanisms. Veterinary Microbiology. 2004;98(2):89-94. DOI: 10.1016/j.vetmic.2003.10.003
  35. 35. Schat KA, Nair V. Marek’s disease. In: Swayne DE, Glisson JR, McDougald LR, Nolan LK, Suarez DL, Nair V, editors. Diseases of Poultry. 13th ed. Wiley-Blackwell: Iowa State Press, Ames, Iowa, USA; 2013. pp. 515-552
  36. 36. Birhane N, Fesseha H. Vaccine failure in poultry production and its control methods: A review. Biomedical Journal of Science and Technology Research. 2020;29:22588-22596. DOI: 10.26717/BJSTR.2020.29.004827
  37. 37. Christe P, de Lope F, Gonzalez G, Saino N, Møller AP. The influence of environmental conditions on immune responses, morphology and recapture probability of nestling house martins (Delichonurbica). Oecologia. 2001;126:333-338. DOI: 10.1007/s004420000527A
  38. 38. Schat KA, Skinner MA. Avian Immunosuppressive Diseases and Immune Evasion. 3rd ed. Boston, MA: Academic Press; 2022. pp. 387-417. DOI: 10.1016/B978-0-12-818708-1.00018-XA
  39. 39. Das S, Palai TK, Mishra SR, Das D, Jena B. Nutrition in relation to diseases and heat stress in poultry. Veterinary World. 2011;4:429-432. DOI: 10.5455/vetworld.2011.429-432
  40. 40. St-Pierre NR, Cobanov B, Schnitkey G. Economic losses from heat stress by US livestock industries. Journal of Dairy Science. 2003;86:52-77. DOI: 10.3168/jds.S0022-0302(03)74040-5
  41. 41. Lara LJ, Rostagno MH. Impact of heat stress on poultry production. Animals. 2013;3(2):356-369. DOI: 10.3390/ani3020356
  42. 42. Naga Raja Kumari K, Narendra ND. Ameliorative measures to counter heat stress in poultry. World’s Poultry Science Journal. 2018;74:117-130. DOI: 10.1017/S0043933917001003
  43. 43. Nawab A, Ibtisham F, Li G, Kieser B, Wu J, Liu W, et al. Heat stress in poultry production: Mitigation strategies to overcome the future challenges facing the global poultry industry. Journal of Thermal Biology. 2018;78:131-139. DOI: 10.1016/j.jtherbio.2018.08.010
  44. 44. Badruzzaman ATM, Noor M, Mamun M, Husna A, Islam KM. Prevalence of diseases in commercial chickens at sylhet division of Bangladesh. International Clinical Pathology Journal. 2015;1(5):104-108. DOI: 10.15406/icpjl.2015.01.00023
  45. 45. Mishra B, Jha R. Oxidative stress in the poultry gut: Potential challenges and interventions. Frontiers in Veterinary Science. 2019;6:60. Published 2019 Mar 4. DOI: 10.3389/fvets.2019.00060
  46. 46. Surai PF, Kochish II, Fisinin VI, Kidd MT. Antioxidant defence systems and oxidative stress in poultry biology: An update. Antioxidants. 2019;8:235. Published 2019 Jul 22. DOI: 10.3390/antiox8070235s
  47. 47. Goo D, Kim JH, Park GH, Delos Reyes JB, Kil DY. Effect of heat stress and stocking density on growth performance, breast meat quality, and intestinal barrier function in broiler chickens. Animals. 2019;9(3):107. DOI: 10.3390/ani9030107
  48. 48. Richards SA. The biology and comparative physiology of thermal panting. Biological Review. 1970;45(2):223-264. DOI: 10.1111/j.1469-185x.1970.tb01631.x
  49. 49. Lal K, Bromley E, Oakes R, Prieto JH, Sanderson SJ, Kurian D, et al. Proteomic comparison of four Eimeria tenella life-cycle stages: Unsporulated oocyst, sporulated oocyst, sporozoite and second-generation merozoite. Proteomics. 2009;9:4566-4576. DOI: 10.1002/pmic.200900305
  50. 50. Kumar H, Kawai T, Akira S. Pathogen recognition by the innate immune system. International Reviews of Immunology. 2011;30:16-34. DOI: 10.3109/08830185.2010.529976
  51. 51. Lee SH, Lillehoj HS, Park DW, et al. Protective effect of hyperimmune egg yolk IgY antibodies against Eimeria tenella and Eimeria maxima infections. Veterinary Parasitology. 2009;163:123-126. DOI: 10.1016/j.vetpar.2009.04.020
  52. 52. Wallach M. Role of antibody in immunity and control of chicken coccidiosis. Trends in Parasitology. 2010;26:382-387. DOI: 10.1016/j.pt.2010.04.004
  53. 53. Lillehoj HS, Lillehoj EP. Avian coccidiosis. A review of acquired intestinal immunity and vaccination strategies. Avian Diseases. 2000;44:408-425
  54. 54. Tiemessen MM, Jagger AL, Evans HG, van Herwijnen MJ, John S, Taams LS. CD4+CD25+Foxp3+ regulatory T cells induce alternative activation of human monocytes/macrophages. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:19446-19451. DOI: 10.1073/pnas.0706832104
  55. 55. Rothwell L, Young JR, Zoorob R, Whittaker CA, Hesketh P, Archer A, et al. Cloning and characterization of chicken IL-10 and its role in the immune response to Eimeria maxima. Journal of Immunology. 2004;173:2675-2682. DOI: 10.4049/jimmunol.173.4.2675
  56. 56. Sand JM, Arendt MK, Repasy A, Deniz G, Cook ME. Oral antibody to interleukin-10 reduces growth rate depression due to Eimeria spp. infection in broiler chickens. Poultry Science. 2016;95:439-446. DOI: 10.3382/ps/pev352
  57. 57. Sehrawat S, Rouse BT. Interplay of regulatory T cell and Th17 cells during infectious diseases in humans and animals. Frontiers in Immunology. 2017;8:341. DOI: 10.3389/fimmu.2017.00341
  58. 58. Hao YX, Yang JM, He C, Liu Q , McAllister TA. Reduced serologic response to avian influenza vaccine in specific-pathogen-free chicks inoculated with Cryptosporidium baileyi. Avian Diseases. 2008;52(4):690-693. DOI: 10.1637/8370-052608-Reg.1
  59. 59. Eterradossi N, Saif YM. Infectious bursal disease. In: Swayne DE, editor. Diseases of Poultry. 14th ed. Chichester: Wiley; 2019. pp. 219-246. DOI: 10.1002/9781119421481.ch7
  60. 60. Kim IJ, Gagic M, Sharma JM. Recovery of antibody-producing ability and lymphocyte repopulation of bursal follicles in chickens exposed to infectious bursal disease virus. Avian Diseases. 1999;43:401-413
  61. 61. Li L, Pielsticker C, Han Z, et al. Infectious bursal disease virus inoculation infection modifies Campylobacter jejuni-host interaction in broilers. Gut Pathogens. 2018;10:13. Published 2018 Mar 30. DOI: 10.1186/s13099-018-0241-1
  62. 62. Yao K, Goodwin MA, Vakharia VN. Generation of a mutant infectious bursal disease virus that does not cause bursal lesions. Journal of Virology. 1998;72(4):2647-2654. DOI: 10.1128/JVI.72.4.2647-2654.1998
  63. 63. Cubas-Gaona LL, Diaz-Beneitez E, Ciscar M, Rodríguez JF, Rodríguez D. Exacerbated apoptosis of cells infected with infectious bursal disease virus upon exposure to interferon alpha. Journal of Virology. 2018;92:e00364-e00318. DOI: 10.1128/JVI.00364-18
  64. 64. Withers DR, Davison TF, Young JR. Diversified bursal medullary B cells survive and expand independently after depletion following neonatal infectious bursal disease virus infection. Immunology. 2006;117:558-565. DOI: 10.1111/j.1365-2567.2006.02332.x
  65. 65. Khatri M, Sharma JM. Susceptibility of chicken mesenchymal stem cells to infectious bursal disease virus. Journal of Virological Methods. 2009;160:197-199. DOI: 10.1016/j.jviromet.2009.05.008
  66. 66. Bashir K, Kappala D, Singh Y, Dar JA, Mariappan AK, Kumar A, et al. Combination of TLR2 and TLR3 agonists derepress infectious bursal disease virus vaccine-induced immunosuppression in the chicken. Scientific Reports. 2019;9(1):8197. Published 2019 Jun 3. DOI: 10.1038/s41598-019-44578-5
  67. 67. Schat KA. Chicken infectious anemia. In: Samal SK, editor. Avian Virology: Current Research and Future Trends. 1st ed. Norfolk, UK: Caister Academic Press; 2019. p. 24187
  68. 68. Su Q , Li Y, Meng F, Cui Z, Chang S, Zhao P. Newcastle disease virus-attenuated vaccine co-contaminated with fowl adenovirus and chicken infectious anemia virus results in inclusion body hepatitis-hydropericardium syndrome in poultry. Veterinary Microbiology. 2018;218:52-59. DOI: 10.1016/j.vetmic.2018.03.019
  69. 69. Smyth JA, Moffett DA, McNulty MS, Todd D, Mackie DP. A sequential histopathologic and immunocytochemical study of chicken anemia virus infection at one day of age. Avian Diseases. 1993;37:324-338
  70. 70. Adair BM, McNeilly F, McConnell CD, McNulty MS. Characterization of surface markers present on cells infected by chicken anemia virus in experimentally infected chickens. Avian Diseases. 1993;37:943-950
  71. 71. Haridy M, Sasaki J, Ikezawa M, Okada K, Goryo M. Pathological and immunohistochemical studies of subclinical infection of chicken anemia virus in 4-week-old chickens. The Journal of Veterinary Medical Science. 2012;74:757-764. DOI: 10.1292/jvms.11-0374
  72. 72. Markowski-Grimsrud CJ, Schat KA. Infection with chicken anaemia virus impairs the generation of pathogen-specific cytotoxic T lymphocytes. Immunology. 2003;109:283-294. DOI: 10.1046/j.1365-2567.2003.01643.x
  73. 73. Erfan AM, Selim AA, Helmy SA, Eriksson P, Naguib MM. Chicken anaemia virus enhances and prolongs subsequent avian influenza (H9N2) and infectious bronchitis viral infections. Veterinary Microbiology. 2019;230:123-129. DOI: 10.1016/j.vetmic.2019.01.024
  74. 74. van Ginkel FW, van Santen VL, Gulley SL, Toro H. Infectious bronchitis virus in the chicken Harderian gland and lachrymal fluid: Viral load, infectivity, immune cell responses, and effects of viral immunodeficiency. Avian Diseases. 2008;52(4):608-617. DOI: 10.1637/8349-050908-Reg.1
  75. 75. Giotis ES, Scott A, Rothwell L, Hu T, Talbot R, Todd D, et al. Chicken anaemia virus evades host immune responses in transformed lymphocytes. The Journal of General Virology. 2018;99:321-327. DOI: 10.1099/jgv.0.001011
  76. 76. Witter RL. Increased virulence of Marek’s disease virus field isolates. Avian Diseases. 1997;41:149-163
  77. 77. Gimeno IM, Witter RL, Neumann U. Neuropathotyping: A new system to classify Marek’s disease virus. Avian Diseases. 2002;46:909-918. DOI: 10.1637/0005-2086(2002)046[0909:NANSTC]2.0.CO;2
  78. 78. Calnek BW. Marek’s disease – a model for herpesvirus oncology. Critical Reviews in Microbiology. 1986;12:293-320. DOI: 10.3109/10408418509104432
  79. 79. Schat KA. Marek’s disease: A model for protection against herpesvirus-induced tumours. Cancer Surveys. 1987;6(1):1-37
  80. 80. Xing Z, Schat KA. Expression of cytokine genes in Marek’s disease virus-infected chickens and chicken embryo fibroblast cultures. Immunology. 2000;100:70-76. DOI: 10.1046/j.1365-2567.2000.00008.x
  81. 81. Jarosinski KW, O, Connell PH, Schat KA. Impact of deletions within the Bam HI-L fragment of attenuated Marek’s disease virus on vIL-8 expression and the newly identified transcript of open reading frame LORF4. Virus Genes. 2003;26:255-269. DOI: 10.1023/a:1024447230464
  82. 82. Jarosinski KW, Schat KA. Multiple alternative splicing to exons II and III of viral interleukin-8 (vIL-8) in the Marek’s disease virus genome: The importance of vIL-8 exon I. Virus Genes. 2007;34:9-22. DOI: 10.1007/s11262-006-0004-9
  83. 83. Morimura T, Hattori M, Ohashi K, Sugimoto C, Onuma M. Immunomodulation of peripheral T cells in chickens infected with Marek’s disease virus: Involvement in immunosuppression. The Journal of General Virology. 1995;76:2979-2985. DOI: 10.1099/0022-1317-76-12-2979
  84. 84. Morimura T, Ohashi K, Kon Y, Hattori M, Sugimoto C, Onuma M. Apoptosis and CD8-down-regulation in the thymus of chickens infected with Marek’s disease virus. Archives of Virology. 1996;141:2243-2249. DOI: 10.1007/BF01718230
  85. 85. Baigent SJ, Ross LJ, Davison TF. Differential susceptibility to Marek’s disease is associated with differences in number, but not phenotype or location, of pp38+ lymphocytes. The Journal of General Virology. 1998;79:2795-2802. DOI: 10.1099/0022-1317-79-11-2795
  86. 86. Sun A, Luo J, Wan B, Du Y, Wang X, Weng H, et al. Lorf9 deletion significantly eliminated lymphoid organ atrophy induced by meq-deleted very virulent Marek’s disease virus. Veterinary Microbiology. 2019;235:164-169. DOI: 10.1016/j.vetmic.2019.06.020
  87. 87. Lee LF, Sharma JM, Nazerian K, Witter RL. Suppression of mitogen-induced proliferation of normal spleen cells by macrophages from chickens inoculated with Marek’s disease virus. Journal of Immunology. 1978;120:1554-1559
  88. 88. Heidari M, Delekta PC. Transcriptomic analysis of host immune response in the skin of chickens infected with Marek’s disease virus. Viral Immunology. 2017;30:377-387. DOI: 10.1089/vim.2016.0172
  89. 89. Islam AF, Wong CW, Walkden-Brown SW, Colditz IG, Arzey KE, Groves PJ. Immunosuppressive effects of Marek’s disease virus (MDV) and herpesvirus of turkeys (HVT) in broiler chickens and the protective effect of HVT vaccination against MDV challenge. Avian Pathology. 2002;31:449-461. DOI: 10.1080/0307945021000005824
  90. 90. Murugesan GR, Ledoux DR, Naehrer K, Berthiller F, Applegate TJ, Grenier B, et al. Prevalence and effects of mycotoxins on poultry health and performance, and recent development in mycotoxin counteracting strategies. Poultry Science. 2015;94:298-1315. DOI: 10.3382/ps/pev075
  91. 91. Sun Y, Song Y, Long M, Yang S. Immunotoxicity of three environmental mycotoxins and their risks of increasing pathogen infections. Toxins. 2023;15:187. DOI: 10.3390/toxins15030187
  92. 92. Ganesan AR, Mohan K, Karthick Rajan D, Pillay AA, Palanisami T, Sathishkumar P, et al. Distribution, toxicity, interactive effects, and detection of ochratoxin and deoxynivalenol in food: A review. Food Chemistry. 2022;378:131978. DOI: 10.1016/j.foodchem.2021.131978
  93. 93. Pandya JP, Arade PC. Mycotoxin: A devil of human, animal and crop health. Advanced Life Science. 2016;5:3937-3941
  94. 94. Wang W, Zhai S, Xia Y, Wang H, Ruan D, Zhou T, et al. Ochratoxin A induces liver inflammation: Involvement of intestinal microbiota. Microbiome. 2019;7:151. DOI: 10.1186/s40168-019-0761-z
  95. 95. Cao W, Yu P, Yang K, Cao D. Aflatoxin B1: Metabolism, toxicology, and its involvement in oxidative stress and cancer development. Toxicology Mechanisms and Methods. 2022;32:395-419. DOI: 10.1080/15376516.2021.2021339
  96. 96. Rushing BR, Selim MI. Aflatoxin B1: A review on metabolism, toxicity, occurrence in food, occupational exposure, and detoxification methods. Food and Chemical Toxicology. 2019;124:81-100. DOI: 10.1016/j.fct.2018.11.047
  97. 97. Diaz GJ, Murcia HW, Cepeda SM. Cytochrome P450 enzymes involved in the metabolism of aflatoxin B1 in chickens and quail. Poultry Science. 2010;89:2461-2469. DOI: 10.3382/ps.2010-00864
  98. 98. Yunus AW, Razzazi-Fazeli E, Bohm J. Aflatoxin B(1) in affecting broiler’s performance, immunity, and gastrointestinal tract: A review of history and contemporary issues. Toxins. 2011;3:566-590. DOI: 10.3390/toxins3060566
  99. 99. Qureshi MA, Brake J, Hamilton PB, Hagler WM, Nesheim S. Dietary exposure of broiler breeders to aflatoxin results in immune dysfunction in progeny chicks. Poultry Science. 1998;77:812-819. DOI: 10.1093/ps/77.6.812
  100. 100. Ringot D, Chango A, Schneider YJ, Larondelle Y. Toxicokinetics and toxicodynamics of ochratoxin A, an update. Chemico-Biological Interactions. 2006;159:18-46. DOI: 10.1016/j.cbi.2005.10.106
  101. 101. Bhatti SA, Khan MZ, Saleemi MK, Hassan ZU. Impact of dietary Trichosporonmycotoxinivorans on ochratoxin A induced immunotoxicity; in vivo study. Food and Chemical Toxicology. 2019;132:110696. DOI: 10.1016/j.fct.2019.110696
  102. 102. Grenier B, Applegate TJ. Modulation of intestinal functions following mycotoxin ingestion: Meta-analysis of published experiments in animals. Toxins. 2013;5(2):396-430. DOI: 10.3390/toxins5020396
  103. 103. Oswald IP, Marin DE, Bouhet S, Pinton P, Taranu I, Accensi F. Immunotoxicological risk of mycotoxins for domestic animals. Food Additives and Contaminants. 2005;22:354-360. DOI: 10.1080/02652030500058320
  104. 104. Swamy HV, Smith TK, Karrow NA, Boermans HJ. Effects of feeding blends of grains naturally contaminated with Fusarium mycotoxins on growth and immunological parameters of broiler chickens. Poultry Science. 2004;83:533-543. DOI: 10.1093/ps/83.4.533
  105. 105. Girgis GN, Barta JR, Brash M, Smith TK. Morphologic changes in the intestine of broiler breeder pullets fed diets naturally contaminated with Fusarium mycotoxins with or without coccidial challenge. Avian Diseases. 2010;54:67-73. DOI: 10.1637/8945-052809-Reg.1
  106. 106. Awad W, Ghareeb K, Böhm J, Zentek J. The toxicological impacts of the Fusarium mycotoxin, deoxynivalenol, in poultry flocks with special reference to immunotoxicity. Toxins. 2013;5(5):912-925. DOI: 10.3390/toxins5050912
  107. 107. Dänicke S, Pappritz J, Goyarts T, Xu B, Rautenschlein S. Effects of feeding a Fusarium toxin-contaminated diet to infectious bursal disease virus-infected broilers on the protein turnover of the bursa of Fabricius and spleen. Achieves in Animal Nutrition. 2011;65:1-20. DOI: 10.1080/1745039x.2010.541191
  108. 108. Xue CY, Wang GH, Chen F, Zhang XB, Bi YZ, Cao YC. Immunopathological effects of ochratoxin A and T-2 toxin combination on broilers. Poultry Science. 2010;89(6):1162-1166. DOI: 10.3382/ps.2009-00609
  109. 109. Grace D, Knight-Jones TJD, Melaku A, Alders R, Jemberu WT. The public health importance and management of infectious poultry diseases in smallholder systems in Africa. Food. 2024;13:411. DOI: 10.3390/foods13030411
  110. 110. Campbell AM, Johnson AM, Persia ME, Jacobs L. Effects of housing system on anxiety, chronic stress, fear, and immune function in Bovan Brown laying hens. Animals (Basel). 2022;12(14):1803. DOI: 10.3390/ani12141803
  111. 111. Wang K, Shen D, Dai P, Li C. Particulate matter in poultry house on poultry respiratory disease: A systematic review. Poultry Science. 2023;102:102556. DOI: 10.1016/j.psj.2023.102556
  112. 112. Gomes AV, Quinteiro-Filho WM, Ribeiro A, Ferraz-de-Paula V, Pinheiro ML, Baskeville E, et al. Overcrowding stress decreases macrophage activity and increases Salmonella Enteritidis invasion in broiler chickens. Avian Pathology. 2014;43:82-90. DOI: 10.1080/03079457.2013.874006
  113. 113. He P, Chen Z, Yu H, Hayat K, He Y, Pan J, et al. Research progress in the early warning of chicken diseases by monitoring clinical symptoms. Applied Sciences. 2022;12:5601. DOI: 10.3390/app12115601
  114. 114. Papageorgiou M, Goliomytis M, Tzamaloukas O, Miltiadou D, Simitzis P. Positive welfare indicators and their association with sustainable management systems in poultry. Sustainability. 2023;15:10890. DOI: 10.3390/su151410890
  115. 115. Landmann M, Scheibner D, Graaf A, Gischke M, Koethe S, Fatola OI, et al. A semiquantitative scoring system for histopathological and immunohistochemical assessment of lesions and tissue tropism in avian influenza. Viruses. 2021;13:868. DOI: 10.3390/v13050868
  116. 116. Alvarez KLF, Poma-Acevedo A, Fernández-Sánchez M, Fernández-Díaz M. An EdU-based flow cytometry assay to evaluate chicken T lymphocyte proliferation. BMC Veterinary Research. 2020;16:230. DOI: 10.1186/s12917-020-02433-0
  117. 117. Onbaşılar EE, Aksoy FT. Stress parameters and immune response of layers under different cage floor and density conditions. Livestock Production Science. 2005;95:255-263. DOI: 10.1016/j.livprodsci.2005.01.006
  118. 118. Adedokun SA, Olojede OC. Optimizing gastrointestinal integrity in poultry: The role of nutrients and feed additives. Frontiers in Veterinary Science. 2019;5:348. DOI: 10.3389/fvets.2018.00348
  119. 119. Butucel E, Balta I, McCleery D, Morariu F, Pet I, Popescu CA, et al. Farm biosecurity measures and interventions with an impact on bacterial biofilms. Agriculture. 2022;12:1251. DOI: 10.3390/agriculture12081251
  120. 120. Marangon S, Busani L. The use of vaccination in poultry production. Revue Scientifique et Technique. 2007;26:265-274
  121. 121. Koopmans M. Surveillance strategy for early detection of unusual infectious disease events. Current Opinion in Virology. 2013;3:185-191. DOI: 10.1016/j.coviro.2013.02.003

Written By

Amra Alispahic, Adis Softic, Aida Kustura, Jasmin Omeragic and Teufik Goletic

Submitted: 15 April 2024 Reviewed: 07 May 2024 Published: 10 June 2024