Open Access

Strategies and innovations for combatting diseases in animals (Review)

  • Authors:
    • Muhammad Ahmad
    • Ishtiaq Ahmed
    • Tayyaba Akhtar
    • Muhammad Amir
    • Shakeela Parveen
    • Edward Narayan
    • Hafiz M.n. Iqbal
    • Saif Ur Rehman
  • View Affiliations

  • Published online on: August 1, 2024     https://doi.org/10.3892/wasj.2024.270
  • Article Number: 55
  • Copyright : © Ahmad et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Emerging and re‑emerging infectious diseases pose significant challenges to animal and public health, and are aligning with global food security concerns. Anthropogenic activities, such as changes in climate, agricultural procedures and farming practices contribute to the spread of zoonotic infections in new geographical areas. Overall diagnostic quality and treatment options have improved over the past few decades. The re‑emergence of old and new lethal diseases, such as highly pathogenic avian influenza in animals and humans, along with recurring epidemics, is increasingly threatening different sectors, including the animal care industry and veterinary medicine. The rapid pace of the animal industry, environmental change and socioeconomic burdens are influencing and challenging veterinarians, farmers and epidemiologists in their capacity to survey and control the spread of diseases in animals, and signify the useful impacts on their livelihoods and environment. There is high research interest in engineering various types of drug‑loaded cues with multiple functionalities to tackle several infectious and non‑infectious diseases. Additionally, the issue can be resolved with international‑level awareness efforts, the deployment of proper design and reliable reporting standards, precise test protocols, and acceptance of the utilization of latent class models to account for, interpret and justify imperfect reference tests. The present review discusses the challenges and strategies for preventing and combating infectious diseases in livestock. Furthermore, the ongoing challenges are summarized, and future considerations, concluding remarks and recommendations are also provided for progress in the animal care and medicine sectors.

1. Introduction

Infection is a type of parasitism that implies an interaction between a parasite and its hosts. Parasites are typically microscopic pathogens that include viruses, bacteria, fungi, protozoans, worms and arthropods. The virulence factor of a disease-causing agent reveals the severity of the infection and is classified as infectious or non-infectious. In developing countries, emerging and re-emerging infectious diseases (IDs) among livestock pose serious challenges to animal and public health, and ultimately align with the international food security agenda (1-6). These emerging new viral infections include diseases, such as Rift Valley fever, West Nile fever, SARS coronavirus, Hendra virus, avian influenza A (H5N1), Nipah virus, Zika virus, Ebola virus, Lassa virus, Marburg virus and swine influenza A (H1N1) virus. These diseases are not only threatening to animals, but also to humans, causing massive economic losses in the veterinary domain and public health concerns globally. The complex association between host populations and other environmental factors creates favorable conditions for the emergence of such diseases. This is mainly due to anthropogenic activities, so-called ‘man-made’, since the entire living system is faced with a series of devastating changes. Such devastating changes occur in the global climate, agricultural procedures and farming practices, among others, due to malpractices, imperfect design, and inefficient handling. All these are major contributors to widespread zoonotic infections in new geographical areas that are further transmissible between animals and humans, either directly or indirectly (7-17).

For instance, the spread of influenza virus A (H1N1), an emerging virus after virus gene recombination is a typical example and poses some significant threats to human health from 2009 when the first case of influenza-like infection in humans was reported in Mexico, and spread to 91 countries until July, 2009 (4,11,18,19). Similarly, in March, 2024, a young goat in Minnesota tested positive for highly pathogenic avian influenza (H5N1). Shortly thereafter, dairy cattle in Texas and Kansas were infected. The virus had likely been present since December, 2023 and had spread to 52 herds across nine states in the USA by May, 2024, causing symptoms, such as a thicker milk consistency and reduced output in affected cows, with evidence of high virus levels in raw milk (12,20-23).

Since 1997, 909 human cases of influenza A (H5N1) have been reported globally, with a 52% mortality rate. From January, 2022 to April, 2024, 26 cases were recorded in eight countries, resulting in seven deaths. In the USA, two cases were recently confirmed among dairy farm workers in Michigan and Texas, marking the first documented instance of human transmission from infected cattle. The virus, identified as Clade 2.3.4.4b, has been detected in various mammals since 2022, emphasizing the need for enhanced surveillance and preventive measures to mitigate international transmission risk (24-26).

According to one estimate, ~75% of new or emerging IDs spread from animals and >60% of IDs spread in humans (27,28). Therefore, effective control and treatment measures for IDs and non-infectious diseases (NIDs) are of high priority to safeguard and secure animals and humans, along with national and international food supplies (4,29-33).

From the world population hike prospects, according to the United Nations (UN), Department of Economic and Social Affairs, the current world population of 7.8 billion is expected to reach 8.6 billion by 2030, 9.8 billion by 2050 and 11.2 billion by 2100(34). Such a rapid pace in the world population growth rate also poses significant challenges, such as an increasing food demand, with a massive burden on the livestock industry to fulfill the increasing food demand for meat or other dairy products (35-37).

In addition, statistics on livestock populations, growth rates, and quality vary considerably from source to source and from place to place. To equalize this fast-paced demand and production ratio, methods or strategies in farm production are extremely varied and pose risks related to the transmission of IDs owing to malpractice. This type of known or unknown vulnerability demonstrates the importance of infectious livestock diseases in global food security (38-43). More specifically, and as aforementioned, the emergence of new pathogenic variants or the re-emergence of old infections and NIDs in the animal industry has devastated national flocks, as has occurred with strains of highly pathogenic avian influenza and others (7,44-49).

Historically, both domestic and commercially raised animals have been affected by IDs and NIDs. According to the World Organization for Animal Health (OIE) report, for the year 2024, the list included 173 animal diseases, infections and infestations (50). With advancements being made in scientific understanding and increased societal awareness, consumers are becoming increasingly concerned about issues related to previous epidemics and current outbreaks (48,49). These concerns have intensified as outbreaks, such as foot-and-mouth disease (FMD), Query fever (Q fever), hemorrhagic septicemia, avian influenza, Newcastle disease, Bluetongue, bovine virus diarrhea, African swine fever, classical swine fever, brucellosis, Peste des petits ruminants, contagious bovine pleuropneumonia, contagious caprine pleuropneumonia, East Coast fever, heartwater, African horse sickness, Aujeszky's disease, tuberculosis, sylvatic rabies, and trichinellosis have become more severe, as shown in Table I (26,49,51-66).

Table I

Some examples of infectious and/or non-infectious diseases in the animal industry along with their characteristic features.

Table I

Some examples of infectious and/or non-infectious diseases in the animal industry along with their characteristic features.

Disease nameAnimals affectedSigns/symptomsTransmission source
Foot-and-mouth disease (FMD)All cloven-hoofed animalsSore feet; thus, they lie down or kneel, saliva dripping from the mouth, blisters in the mouth, ulcers around the feetIngestion of contaminated forage and water; all infective body excretions, including urine, milk, feces, and semen
Highly pathogenic avian Influenza (HPAI)Chicken, ducksSwollen wattles, blue/purple comb, red streaks on legs, bleeding under the skin, accumulation of fluid/bloodFeces or saliva
Newcastle disease (ND)Numerous species of birdsSwelling and redness of the eyelids, diarrhea, crooked neck, paralyzed, speckled spleen, hemorrhage in some parts of the intestineFeces and saliva, infected cages, footwear, or equipment
African swine fever (ASF)All kinds of swineHigh fever, red skin on ears and tail, ulcer, enlarged spleenFeces, saliva, and blood
Classical swine fever (CSF)PigsHigh fever, weight loss, convulsions, crusty bloody fluids around the eyesFeces and saliva, infected equipment
Peste des petits ruminants (PPR)Sheep and goatsFoul-smelling diarrhea, crusting around the mouth and nose, ulcers in the mouth, reddened lungsFeces and saliva, infected food or equipment
Contagious bovine pleuropneumonia (CBPP)All breeds of cattleRunny nose, extended neck, yellow covering around the lungsSaliva or mucus
Contagious caprine pleuropneumonia (CCPP)GoatsFever, breathing difficulty, coughing, weight loss, frothy nasal discharges, enlarged and bloody lungsSaliva or mucus
East Coast fever (ECF)Cattle and buffaloHigh fever, breathing difficulty, swelling/enlarged lymph nodes Tick-transmission
HeartwaterCattle, sheep, and goatsHigh fever, convulsions, crooked necks, excess fluid around the heart/lungs, engorged Amblyomma ticks on the skin,Bitten by an infected tick (vector)
African horse sickness (AHS)HorsesHigh fever, excess sweating, coughing, swollen around the eye area, Accumulation of fluid in the lungsAHS virus is transmitted by small biting insects
Hemorrhagic septicemia (HS)Buffalos and cattle; other goats, camels, horses, and donkeys, while dogs and fowls are not susceptibleRapid onset, high fever, dullness, lacrimation, dyspnea, swelling in the head-throat-brisket regionIngestion or inhalation of causative agents by contaminated feed and water that occurs during wet condition
Bovine viral diarrheaCattleBloody diarrhea, high fever, mouth ulcer, off-feed, ataxiaIngestion of contaminated fomites or either through congenital infection
TrichinellosisPigs, horses, carnivores and other animals (zoonotic disease)Eye swelling, cough, itching, diarrhea or constipation, achy joints, fever, nausea, vomitingIngestion of contaminated meat or meat products because infected animals or humans have nematode larvae lodged in their muscles

Efforts have been made from a scientific standpoint to prevent and combat the further invasion of highly infectious diseases. The objective of the present review was to illustrate and discuss certain drug-loaded cues and their smart exploitation for IDs and NIDs in animals. The present review focuses on various IDs and NIDs, along with the detection measures, forecasting and control of infectious disease epidemics.

2. Detective measures and epidemiological modeling

The implementation of epidemiological modeling is not new and has a long progressive history (67-74). The use of real-time epidemiological modeling to design robust policies during serious outbreaks has increased over the past 20 years (75-78).

For instance, there are recent examples of the real-time epidemiological modeling of animal diseases, such as FMD epidemics (75,76,78-80). Among the various deployed modeling systems, compartmental modeling is the most commonly used and has been further categorized as deterministic or stochastic (81). Various parameters, such as infection type, symptoms and status, are considered critical when implementing compartmental modeling (82-84). Protective measures are typically considered before a pathogen enters a new population, using modeling analytics to guide targeted surveillance. Comprehensive surveillance systems integrating livestock, wildlife and human components are crucial, and have been recommended in several reports (29,85-88).

Despite the usefulness of modeling, several challenges remain. Numerous articles have highlighted issues in modeling infectious disease dynamics, including applications in public health policy and livestock diseases (89,90). From the public health policy perspective (90), six main challenges have been described: i) Communicating the limits of modeling; ii) maintaining the value of models in the face of long time horizons; iii) usefully deploying modeling in the context of ‘black swans’; iv) integrating modelers and model-building into the policy process; v) economic analysis and decision support; and vi) creating a cycle where results inform decisions and vice versa. Similarly, another study (89), presented eight challenges: i) Linking models to transmission experiments; ii) disease control by selective breeding; iii) applying models to data/resource-poor settings; iv) how best to exploit rich livestock data; v) improving spatial models; vi) unifying multiple scales of transmission; vii) linking livestock populations to other species; and viii) modeling livestock with political and economic constraints in modeling infectious livestock diseases (91).

3. Forecasting and control of infectious disease epidemics

Once the initial phase of an outbreak is identified, the effective management and control of the causative agent are often required to minimize its overall impact on animal and human health. Upon this initial detection in the host population, a considerable challenge which is often encountered is forecasting whether this initial phase will dissipate or will progress to a major epidemic concern (83,92,93). For the effective control of IDs or epidemics or to help manage diseases, the implementation of surveillance programs is of utmost importance. The most advanced countries/jurisdictions have executed routine surveillance programs in direct response to major disease outbreaks that have caused catastrophic losses. The implementation of surveillance programs or surveys combined with a range of biosafety and biosecurity measures have allowed these jurisdictions to effectively eradicate certain diseases of concern (94). In this context, lessons for underdeveloped countries lie in these directions. Herein, six key drivers are proposed, which should be taken into consideration: i) The notifiable reporting of major disease outbreaks; ii) the protection of wild and native species; iii) the control of trade, border biosecurity and biosafety to prevent undesirable disease epidemics; iv) the implementation of legislative measures; v) socioeconomic contribution and public pressure; and vi) routine-based inspections of in-practice surveillance programs (95,96). Routine-based inspections of in-practice surveillance programs have been further subcategorized into six categories: i) The reporting notifiable diseases and elevated mortality; ii) routine testing for pathogens; iii) regular inspections of farm areas; iv) reliable quality tests before trade; v) the maintenance of a proper hygienic environment; and vi) onsite reporting requirements (97).

To date, various treatment strategies have been developed and exploited to control various diseases and disease-causing agents. Comprehensive guidelines for the judicious therapeutic use of antimicrobials in poultry have been reported by the American Veterinary Medical Association (AVMA) (for further details, please visit AVMA online: www.avma.org). A range of antibacterial drugs, such as amoxicillin, metronidazole, clindamycin, doxycycline, spectinomycin, bacitracin, chlortetracycline, cephalexin, ciprofloxacin, sulfamethoxazole and trimethoprim, and antiviral therapeutics, such as oseltamivir and zanamivir, are currently being used. However, direct and excessive drug use has raised concerns regarding drug resistance (98,99). Of note, the effectiveness of treatment with antiviral drugs, such as oseltamivir and zanamivir, is becoming increasingly reduced due to the development of resistance. Oseltamivir is a selective antiviral prodrug that is used in the treatment of influenza. Zanamivir is another drug which is used as an inhibitor of neuraminidase; this drug is used in the treatment of the common flu and to protect against viruses A and B (100). The development of antiviral drugs which are highly effective and which can protect against a wide range of viral pathogens is an arduous task; this is due to the fact that viruses use host cells for replication. For this reason, the global research community is aiming to expand the range of currently available antiviral drugs to other pathogen families (99). Drug resistance is continually becoming more of a challenge; thus, the development of novel drugs which are able to combat a wide variety of viral infections, is of utmost importance. Another key issue is the fact that the genetic composition of viruses may change and this may subsequently render the virus resistant to several therapeutics (101). Viruses may become resistant to antiviral drugs primarily due to spontaneous and intermittent mechanisms. A previous study isolated three types of influenza A viruses; each of these types was found to have a distinct neuraminidase genetic mutation and all types were found to be resistant to the neuraminidase inhibitor, oseltamivir (102). Due to viral evolution, viruses are becoming more resistant to antiviral drugs; thus, continuous monitoring is required. (103).

In this context, materials-based cues/constructs with antimicrobial ‘non-drugs’ e.g., nanoparticles or effective drug-loaded composite materials have found widespread biomedical applications (104,105). The importance of such drug-loaded interventions (also known as Trojan horse strategies of drug delivery) has been established through various reports, expanding the efficacy of conventional drugs, helping to reduce the dosage and frequency of drug usage, and preventing antimicrobial resistance against drugs (106). Specific strategies, such as antibiotic stewardship programs and alternatives to antibiotics have improved animal husbandry practices, legislation and regulatory measures, surveillance and monitoring, public awareness and education, and research and development are being used to combat antimicrobial resistance.

Material-based cues with or without loaded drugs provide additional advantages, such as targeted drug delivery, slow and controlled drug release, low or no toxicity, internal/external stimuli-responsive behavior and controlled degradation or effective release rate in a sophisticated manner. With these added values, substantial research efforts have been made to engineer material-based cues/constructs with or without loaded drugs with antibacterial and/or antiviral activity (107).

4. Quest for materials-based cues

With key scientific advances being made in the field of materials science and the added value of nanotechnology, the quest for material-based cues/constructs in numerous geometries with multifunctional activities (Fig. 1), such as antibacterial, antifungal and antiviral activities, has increased over the past several years (99,104,108-112). In addition, material-based cues possess exceptional biocompatibility, biodegradability, and internal and external stimuli-responsive modes; thus, they are broadly used in various sectors of the modern world, such as clinical, pharmaceutical, cosmeceutical, nutraceutical, biomedical engineering, tissue engineering and veterinary medicine (109,112-115). Biocompatible and stimuli-responsive materials can lead to the development of more effective treatments by ensuring that they positively interact with animal tissues and adapt to changing physiological conditions. Biocompatibility minimizes the risk of adverse immune responses and other complications, thereby making treatment safer for a wide range of animal species. Stimuli-responsive materials enable precise control over the release and action of therapeutic agents, enhance treatment efficiency, and reduce the need for frequent interventions. These materials can be tailored to the specific needs of different veterinary applications, allowing for customized treatments that cater to the unique requirements of various animal species and conditions. The majority of infectious or non-infectious diseases in humans and animals are mainly caused by viruses, bacteria, fungi, protozoans, worms and arthropods. Thus, materials science has provided a new wave of pristine or drug-loaded cues/constructs with antibacterial, antifungal, and/or antiviral potentials.

Currently, there is an increasing demand for antibiotic alternatives for the treatment of infections, and advances in drug delivery can play a crucial role in enhancing the efficacy of herbal-derived active principles, such as curcumin and flavonoids. It has been reported that curcumin encapsulated in nanocapsules can effectively reduce the extent of listeriosis in gerbils experimentally infected with Listeria monocytogenes (116). Another study reported the improved bactericidal and biofilm formation inhibitory effects of flavonoids against Escherichia coli upon nanoencapsulation (117). In another study, the nano-delivery of Origanum majorana essential oil required lower doses of therapeutic oil to treat fish infected with Aeromonas hydrophila (118).

Another potential application of this drug delivery system is in biofilms, which are a public health hazard. Nano-encapsulated glycerol monolaurate has been reported to reduce viable bacteria within the biofilm of Pseudomonas aeruginosa, providing a positive response to nano-drug delivery against biofilms (119,120). Nano-loaded antibiotics have also been reported to be more efficient against Pseudomonas aeruginosa (121). However, findings obtained in vitro must also be able to be produced in vivo; some in vitro findings have not been observed in in vivo biofilm therapeutic studies (122).

For this purpose, an array of novel materials, such as chitosan, alginate, cellulose and graphene, has been engineered to exhibit potent bioactivity towards different pathogens on contact without releasing any toxic biocides for healthy cells. A facile in situ procedure for fabricating hydrogel-silver nanocomposites involves the formation of silver nanoparticles within swollen poly (acrylamide-co-acrylic acid) hydrogels (123). Apart from antibiotic-nanoparticle combinations, therapeutic interventions using antimicrobial peptide-nanoparticle combinations against infectious agents are also being investigated, and a number of these have exhibited promising outcomes (124). A recent study reported thermo-magnetically responsive drug-eluting grippers as a convenient means of drug delivery with minimal invasiveness and for biomedical engineering, as they can be moved using magnetic fields after doping with magnetic nanoparticles (125). The effective targeted delivery of drugs can also reduce the dose requirement, particularly as regards the treatment of resistant bacterial forms, such as mycobacteria. A previous study reported that the nano-delivery of an antibiotic combination against Mycobacterium avium in mice elicited sustained and effective drug release compared to free drugs (126). This opens a promising platform for the further advanced research on drugs against bovine tuberculosis. Novel interventions, such as ultrasmall tungsten disulfide quantum dots with catalytic as well as photoluminescence properties, have been tested in biomedicine for their application as drug delivery nanoparticles against infection and biofilms, and ultimately provide a promising platform for checking infections (57,127).

Based on the antibacterial activity analysis results, the developed hydrogel-silver nanocomposites have demonstrated excellent antibacterial effects against Escherichia coli. A previous study prepared a polyelectrolyte complex (PEC) hydrogel using chitosan as the cationic polyelectrolyte and γ-poly (glutamic acid) (γ-PGA) as the anionic polyelectrolyte. The chitosan-based PEC gel exhibited antibacterial activity against Escherichia coli and Staphylococcus aureus (128). Additionally, the PEC hydrogel was effective in promoting cell proliferation when tested in an in vitro culture of 3T3 fibroblasts. According to that study (128), based on the results, the PEC hydrogel appears to have potential as a new material for biomedical applications and has been exploited as a wound-dressing material (129).

The development of silver nanoparticles loaded with chitosan-alginate constructs has been reported with antibacterial activities against six bacterial strains, i.e., Staphylococcus aureus, Pseudomonas aeruginosa, Klebsiella pneumoniae, Acinetobacter baumannii, Morganella morganii and Haemophilus influenza (130,131). In 2019, Bilal et al (132) developed and characterized the biogenic nanoparticle-chitosan conjugates with antimicrobial (against Gram +ve and Gram -ve bacterial isolates, i.e., Staphylococcus aureus, and Escherichia coli), antibiofilm (against Pseudomonas aeruginosa) and anticancer (against the MCF-7 breast cancer cell line) potential. In another study, a nanocomposite containing ceftriaxone loaded into bioactive chitosan (obtained from the fungus Cunninghamella elegans) sustained the antibacterial effects of ceftriaxone against several drug-resistant bacterial strains (133). Sepsis due to severe bacterial infections is at the forefront of livestock mortality. The management of systemic sepsis through the strategic management of the infectious microenvironment using bioresponsive nanoparticles in murine models has shown promise for the future development of nanomedicine for sepsis (134). In the case of antimicrobial peptide-loaded topical antibacterial therapy, it has been found that a peptide applied through poloxamer gel can effectively reduce the Staphylococcus aureus count, whereas application as a lipid nanocarrier was not found to be very effective (135).

As regards antiviral drugs, a previous study reported that the antiviral activity of monocaprin solutions containing 5% propylene glycol was markedly suppressed by 5% polysorbate 20(136). The same study demonstrated that the antiviral activity of solutions containing 7.5% propylene glycol and polysorbate 20 (0.75 to 1.5% concentration) was comparable to that of pure monocaprin (136). Monocaprin is a monoglyceride that has antiviral activity. It has been demonstrated that monocaprin is effective against enveloped viruses, such as vesicular stomatitis virus, herpes simplex virus (HSV), visna virus and human immunodeficiency virus (HIV) in vitro (137,138).

Similarly, previous studies have demonstrated that hydrophilic gels containing monocaprin at a concentration of 20 mM were able to inactivate HSV-2 and HSV-1 by >100,000-fold (137,138). In their study, Rokhade et al (139) developed semi-interpenetrating polymer network microspheres of acrylamide-grafted dextran and chitosan-based hydrogels for the controlled release of the antiviral drug, acyclovir. Among the antiviral therapeutics, antiretroviral drug research is the most critical, particularly for combating HIV infections. Progress has been made in the development of effective nanoparticle-based technologies for the delivery of drugs to the central nervous system to combat retroviral progression (140). For instance, an antiretroviral peptide drug (enfuvirtide), loaded through an iron oxide nanoparticle coated with an amphiphilic polymer, has been shown to be successful in crossing the blood-brain barrier for effective drug delivery to the central nervous system (141,142).

Other examples of the smart exploitation of various drug-loaded cue formulations against infectious pathogens and diseases with considerable potential in veterinary medicine are summarized in Table II.

Table II

Smart exploitation of various drug-loaded cues/constructs formulations against infectious pathogens with considerable potentialities for veterinary medicine.

Table II

Smart exploitation of various drug-loaded cues/constructs formulations against infectious pathogens with considerable potentialities for veterinary medicine.

Infectious pathogen or diseaseLoaded drugCo-material usedAnimal modelBioactivity or proposed applications(Refs.)
Cutaneous leishmaniasis (L. amazonensis and L. braziliensis)Amphotericin BPoly(vinyl alcohol) (PVA) hydrogels-Antifungal and leishmanicidal activity, functional wound dressing(147)
Candida albicansMiconazoleChitosan-based nanoparticlesYesAntifungal activity, Promising therapy for the treatment of vulvovaginal candidiasis(148)
Cutaneous leishmaniasisAmphotericin B Poly(lactic-co-glycolic acid) (PLGA) nanoparticles-Topical therapy, useful for the local treatment of Cutaneous leishmaniasis(149)
Cryptococcus neoformansAmphotericin BMethoxy poly(ethylene glycol)- poly(lactide)-poly(β-amino ester) nanocarriersYesReduce the C. neoformans burden in the lungs, liver and spleen, based on its improved biodistribution(150)
Leishmania majorAmphotericin BNano-sized chitosanYesWound healing and parasite inhibition, successful treatment of Leishmania major pathological effects(151)
Leishmania majorAmphotericin BAnionic linear globular dendrimerYesSuccessful treatment of Leishmania's major pathological effects(151)
Visceral leishmaniasis (Leishmania donovani)Amphotericin BPeptide-coated iron oxide nanoparticles-Reduce total parasite burden in spleen, treatment of visceral leishmaniasis(152)
Leishmaniasis (Leishmania tropica)ParomomycinSolid lipid nanoparticlesYesInhibit the parasite propagation, treatment of leishmaniasis(153)
Leishmania donovaniAmphotericin BGlycol chitosan-stearic acid copolymerYesEffective antileishmanial properties, efficacious drug delivery system(154)
Fusarium solaniAmphotericin BEudragit RL 100 nanoparticlesYesAntifungal activity, suitable against eye irritation, Ophthalmic application(155)
Paracoccidioides brasiliensisDesoxycholate amphotericin B Poly(lactic-co-glycolic acid) (PLGA) and dimercaptosuccinic acid (DMSA) polymeric blendsYesUseful to treat systemic fungal infections such as paracoccidioidomycosis, candidiasis, aspergillosis, and cryptococcosis(149)
Systemic candidiasisAmphotericin BSodium alginate nanospheresYesTargeted antifungal therapy(156)
Systemic candidiasis, Candida albicansCaspofungin-YesPersistence of caspofungin in tissues to understand drug activity(157)

The therapeutic targeting of immune cells using drug-loaded constructs is gaining increasing attention, particularly for combating cancerous, inflammatory and autoimmune conditions. A previous study reported a novel nanocapsule based on silica that can be loaded with a large dose of drugs targeting immune cells, such as macrophages, which can be used in tumor therapy without systemic toxicity (143).

5. Future perspectives

Literature with suitable examples encourages veterinarians toward material-based cues to combat diseases in the animal sector. The current advancements in materials science, along with the involvement of nanotechnology in numerous clinical applications, have significantly improved the detection, diagnosis and therapy of diseases that have been difficult to manage in the past (144-146). Furthermore, with ever-increasing scientific knowledge and common awareness, consumers and producers are on common ground and are highly concerned about the overuse/overdosing/misuse of antibiotics. Such a heavy consumption of antibiotics has also resulted in the emergence of multidrug-resistant pathogens. In this context, the absence of highly effective and 100% reliable treatment against various concerning and resistant diseases makes material-based cues, with or without loaded drugs in their active form, a promising option for veterinarians. The conceptual framework of antibiotic resistance development associated with antibiotic residues in the environment that lead to treatment failure is illustrated in Fig. 2.

Studies developed at a laboratory scale using animal models for different disease-causing agents have stimulated employment in routine practice and can certainly be extrapolated for veterinary medicine in the future. For instance, skin-related infections and NIDs can be efficiently diagnosed in infected animals or companion animals. Thus, material-based drug-loaded hydrogels could be a straightforward solution for improving Veterinary Dermatology. Other examples include udder and ear helminthiases in cows, which can be effective in tackles in which topical drugs are ineffective. Thus, drug-material-based combined therapy with topical bioactive agents can be considered an alternative to topical drugs alone.

6. Current challenges, concluding remarks and outlook

As aforementioned, in different respective sections, the (re)-emergence of old and new lethal diseases along with recurring epidemics remains a massive challenge to the development of the animal sector in developing countries. Several critical factors play key roles in the impact and spread of the disease in various resource-limiting countries. Among these, poor animal care systems, non-hygienic maintenance, unsatisfactory livelihoods, unending intensification, reliance on external trade, and biosafety and biosecurity issues are the major examples. Under such unavoidable circumstances, the following question arises: That is, if the proximity of workers, along with their observational skills, could be used as a route for syndromic surveillance. Considering this thoughtful scenario, there is no reason to believe that such skillful workers could not notice the basic symptoms (e.g., as summarized in Table I) and deviations from the norm, such as first-hand contact, to highlight existing and emerging diseases. To tackle this challenge effectively, robust and aware-full epidemiological modeling and the design of surveillance, detection, and protection measures need to be deployed at the grassroots level to better understand the factors that influence individuals or collectively. To further strengthen such surveillance strategies, veterinarians should work in interdisciplinary and multidisciplinary areas of veterinary science, epidemiology, medicine, materials science, biology, chemistry, microbiology and social science to design robust systems that are inclusive of skillful individuals and state-of-the-art technology.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

All authors (MAh, IA, TA, MAm, SP, EN, HMNI and SUR) substantially contributed to the conception, design, analysis and interpretation of the data to be included in the review. All authors have read and approved the final version of the manuscript, and agree to be accountable for its contents. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Rist CL, Arriola CS and Rubin C: Prioritizing zoonoses: A proposed one health tool for collaborative decision-making. PLoS One. 9(e109986)2014.PubMed/NCBI View Article : Google Scholar

2 

Tiwari R, Kumar H, Dutt T, Singh BP, Pachaiyappan K and Dhama K: Future challenges of food security and sustainable livestock production in India in the changing climatic scenario. Asian J Anim Vet Adv. 9:367–384. 2014.

3 

Lindahl JF and Grace D: The consequences of human actions on risks for infectious diseases: A review. Infect Ecol Epidemiol. 5(30048)2015.PubMed/NCBI View Article : Google Scholar

4 

Tomley FM and Shirley MW: Livestock infectious diseases and zoonoses. Philos Trans R Soc B Biol Sci. 364:2637–2642. 2009.PubMed/NCBI View Article : Google Scholar

5 

Esposito MM, Turku S, Lehrfield L and Shoman A: The impact of human activities on zoonotic infection transmissions. Animals (Basel). 13(1646)2023.PubMed/NCBI View Article : Google Scholar

6 

Sm E, Altilmisani NM, Albishri F, Gad HA, Al-Dubai TA and Al-Wesabi EO: Overview of quality control and safety in public health pest laboratory in Jeddah, Saudi Arabia. Int J Agric Biosci. 13:92–100. 2024.

7 

Dhama K, Dhama K, Chakraborty S, Tiwari R, Kumar A, Rahal A, Latheef SK, Wani MY and Kapoor S: Avian/Bird flu virus: Poultry pathogen having zoonotic and pandemic threats: A review. J Med Sci. 13:301–315. 2013.

8 

Han BA, Kramer AM and Drake JM: Global patterns of zoonotic disease in mammals. Trends Parasitol. 32:565–577. 2016.PubMed/NCBI View Article : Google Scholar

9 

Sayed E, Altilmisani NM, Albishri F, Ahmed A, Elkhalifa SM, Al-Dubai TA and Al-Wesabi EO: Prevalence and zoonotic potential of parasites in wild rats in Jeddah City, Saudi Arabia. Int J Vet Sci. 13:232–240. 2024.

10 

Morwal S and Sharma SK: Bacterial zoonosis-A public health importance. J Dairy Vet Anim Res. 5:56–59. 2017.

11 

Tounta DD, Nastos PT and Tesseromatis C: Human activities and zoonotic epidemics: A two-way relationship. The case of the COVID-19 pandemic. Glob Sustain. 5(e19)2022.

12 

Tazerji SS, Nardini R, Safdar M, Shehata AA and Duarte PM: An overview of anthropogenic actions as drivers for emerging and re-emerging zoonotic diseases. Pathogens. 11(1376)2022.PubMed/NCBI View Article : Google Scholar

13 

Subedi D, Farhan MHRF, Niraula A, Shrestha P, Chandran D, Acharya KP and Ahmad M: Avian influenza in low and middle-income countries (LMICs): Outbreaks, vaccination challenges and economic impact. Pak Vet J. 44:9–17. 2024.

14 

Mujahid U, Ahmad M, Mujahid A, Narayan E, Rehman SU, Iqbal HMN and Ahmed I: Recent outbreak of Marburg virus; a global health concern and future perspective. Eur J Clin Microbiol Infect Dis. 43:209–211. 2024.PubMed/NCBI View Article : Google Scholar

15 

Ahmad M, Ahmed I, Satapathy P, Asumah MN and Padhi BK: Re-emergence of the Lassa virus in Africa: A global health concern. Int J Surg. 109:1044–1045. 2023.PubMed/NCBI View Article : Google Scholar

16 

Hing S, Narayan EJ, Thompson RCA and Godfrey SS: The relationship between physiological stress and wildlife disease: Consequences for health and conservation. Wildl Res. 43:51–60. 2016.

17 

Alotaibi BA, Muddassir M, Alotaibi MR, Azeem MI and Alsanhani A: Assessing university students knowledge and awareness about COVID-19 infection symptoms and preventive measures in, Saudi Arabia. Int J Agric Biosci. 12:208–215. 2023.

18 

Okamatsu M, Hiono T, Kida H and Sakoda Y: Recent developments in the diagnosis of avian influenza. Vet J. 215:82–86. 2016.PubMed/NCBI View Article : Google Scholar

19 

Islam MT, Roy S, Talukdar H, Shammi SA and Ahmed S: Sero-prevalence and associated risk factors of avian influenza virus infection in backyard chicken at Sylhet region, Bangladesh. Int J Vet Sci. 13:362–368. 2024.

20 

Stone H, Jindal M, Lim S, Dawson R, Quigley A, Scotch M and MacIntyre CR: Potential pathways of spread of highly pathogenic avian influenza A/H5N1 clade 2.3.4.4b across dairy farms in the United States medRxiv: 2024.05.02.24306785, 2024.

21 

Ahmed A, Saqlain S, Rasool A, Muhammad S and Umar S: Avian influenza virus (H5N1) was not detected among dairy cattle and farm workers in Pakistan. Influenza Other Respi Viruses. 18(e13317)2024.PubMed/NCBI View Article : Google Scholar

22 

Hu X, Saxena A, Magstadt DR, Gauger PC, Burrough E, Zhang J, Siepker C, Mainenti M, Gorden PJ, Plummer P and Li G: Highly pathogenic avian influenza A (H5N1) clade 2.3.4.4b virus detected in dairy cattle bioRxiv: 2024.04.16.588916, 2024.

23 

Briand FX, Souchaud F, Pierre I, Beven V, Hirchaud E, Hérault F, Planel R, Rigaudeau A, Bernard-Stoecklin S, Van der Werf S, et al: Highly pathogenic avian influenza A(H5N1) clade 2.3.4.4b virus in domestic cat, France, 2022. Emerg Infect Dis. 29:1696–1698. 2023.PubMed/NCBI View Article : Google Scholar

24 

Uyeki TM, Milton S, Hamid CA, Webb CR, Presley SM, Shetty V, Rollo SN, Martinez DL, Rai S, Gonzales ER, et al: Highly pathogenic avian influenza A(H5N1) virus infection in a dairy farm worker. N Engl J Med. 390:2028–2029. 2024.PubMed/NCBI View Article : Google Scholar

25 

Putri DD, Handharyani E, Soejoedono RD, Setiyono A and Etriwati  : Newcastle disease polyclonal antibodies as candidate reagents in immunohistochemistry diagnostic test and passive immunization. Int J Vet Sci. 13:259–265. 2024.

26 

Reynolds DL, Simpson EB and Hille MM: Evidence for antibody dependent enhancement for an avian coronavirus. Int J Vet Sci. 13:707–711. 2024.

27 

Chomel BB, Belotto A and Meslin FX: Wildlife, exotic pets, and emerging zoonoses. Emerg Infect Dis. 13:6–11. 2007.PubMed/NCBI View Article : Google Scholar

28 

World Zoonoses Day: Most emerging infectious diseases originate in animals. Source Texas Vet Med Assoc, 2018.

29 

Dhama K, Chakraborty S, Tiwari R, Verma AK, Saminathan M, Amarpal Malik YS, Nikousefat Z, Javdani M and Khan RU: A concept paper on novel technologies boosting production and safeguarding health of humans and animals. Res Opin Anim Vet Sci. 4:353–370. 2014.

30 

Christaki E: New technologies in predicting, preventing and controlling emerging infectious diseases. Virulence. 6:558–565. 2015.PubMed/NCBI View Article : Google Scholar

31 

Rukambile E, Sintchenko V, Muscatello G, Kock R and Alders R: Infection, colonization and shedding of Campylobacter and Salmonella in animals and their contribution to human disease: A review. Zoonoses Public Health. 66:562–578. 2019.PubMed/NCBI View Article : Google Scholar

32 

Williamson ED: Vaccines for emerging pathogens: From research to the clinic. Clin Exp Immunol. 196:155–156. 2019.PubMed/NCBI View Article : Google Scholar

33 

Feng Z, Hag M El, Wang N, Qin T, Chen S and Peng D: Negative regulation of RpoS-mediated STM1703 in biofilm formation of salmonella pullorum. Pak Vet J. 43:25–32. 2023.

34 

Nations United: World population prospects. United Nations, 2017.

35 

Delgado C, Rosegrant M, Steinfeld H, Ehui S and Courbois C: Livestock to 2020: The next food revolution. Outlook Agric. 30:27–29. 2001.

36 

Jones PG and Thornton PK: Croppers to livestock keepers: Livelihood transitions to 2050 in Africa due to climate change. Environ Sci Policy. 12:427–437. 2009.

37 

Mottet A, Teillard F, Boettcher P, Besi GD and Besbes B: Review: Domestic herbivores and food security: Current contribution, trends and challenges for a sustainable development. Animal. 12:S188–S198. 2018.PubMed/NCBI View Article : Google Scholar

38 

Bengis RG, Kock RA and Fischer J: Infectious animal diseases: The wildlife/livestock interface. Rev Sci Tech. 21:53–65. 2002.PubMed/NCBI View Article : Google Scholar

39 

Miller RS, Farnsworth ML and Malmberg JL: Diseases at the livestock-wildlife interface: Status, challenges, and opportunities in the United States. Prev Vet Med. 110:119–132. 2013.PubMed/NCBI View Article : Google Scholar

40 

Wobeser GA: Essentials of Disease in Wild Animals. Wiley-Blackwell, Hoboken, NJ, 2013.

41 

Wiethoelter AK, Beltrán-Alcrudo D, Kock R and Mor SM: Global trends in infectious diseases at the wildlife-livestock interface. Proc Natl Acad Sci USA. 112:9662–9667. 2015.PubMed/NCBI View Article : Google Scholar

42 

Cárdenas L, Awada L, Tizzani P, Cáceres P and Casal J: Characterization and evolution of countries affected by bovine brucellosis (1996-2014). Transbound Emerg Dis. 66:1280–1290. 2019.PubMed/NCBI View Article : Google Scholar

43 

Dik I, Bulut O, Avci O, Hasoksuz M, Palanci HS, Aslim HP and Bulut Z: Molecular detection and characterization of bovine noroviruses from cattle in Konya, Turkey. Pak Vet J. 43:67–72. 2023.

44 

Alexander DJ: A review of avian influenza in different bird species. Vet Microbiol. 74:3–13. 2000.PubMed/NCBI View Article : Google Scholar

45 

Velkers FC, Bouma A, Matthijs MGR, Koch G, Westendorp ST and Stegeman JA: Outbreak of avian influenza H7N3 on a Turkey farm in the Netherlands. Vet Rec. 159:403–405. 2006.PubMed/NCBI View Article : Google Scholar

46 

Alexander DJ: An overview of the epidemiology of avian influenza. Vaccine. 25:5637–5644. 2007.PubMed/NCBI View Article : Google Scholar

47 

Mourya DT, Yadav PD, Ullas PT, Bhardwaj SD, Sahay RR, Chadha MS, Shete AM, Jadhav S, Gupta N, Gangakhedkar RR, et al: Emerging/re-emerging viral diseases & new viruses on the Indian horizon. Indian J Med Res. 149:447–467. 2019.PubMed/NCBI View Article : Google Scholar

48 

Hayek MN: The infectious disease trap of animal agriculture. Sci Adv. 8(eadd6681)2022.PubMed/NCBI View Article : Google Scholar

49 

Rohr JR, Barrett CB, Civitello DJ, Craft ME, Delius B, DeLeo GA, Hudson PJ, Jouanard N, Nguyen KH, Ostfeld RS, et al: Emerging human infectious diseases and the links to global food production. Nat Sustain. 2:445–456. 2019.PubMed/NCBI View Article : Google Scholar

50 

World Organisation for Animal Health (OIE): Information on aquatic and terrestrial animal diseases: OIE, Paris, 2021.

51 

Abusalab S and Hamid M: Haemorrhagic septicaemia: A general review. Sudan J Vet Res. 18:1–14. 2003.

52 

Jelsma T, Wijnker JJ, Smid B, Verheij E, van der Poel WHM and Wisselink HJ: Determination of intestinal viral loads and distribution of bovine viral diarrhea virus, classical swine fever virus, and peste des petits ruminants virus: A pilot study. Pathogens. 10(1188)2021.PubMed/NCBI View Article : Google Scholar

53 

Karakurt E, Nuhoğlu H, Dağ S, Çelebi Ö, Büyük F, Beytut E, Yıldız A, Kuru M and Akça D: Immunohistochemical investigation of TNF-α and IFN-γ expressions in sheep fetuses with brucellosis. Pak Vet J. 43:85–90. 2023.

54 

Yi SW, Bui NA, Lee HS, Bui VN, Dao DT, Nguyen TH, Lee HG, Jung YH, Hur TY and Oh SI: Age-dependent cytokine expression in response to foot-and-mouth disease virus in bovine peripheral blood mononuclear cells. Pak Vet J. 43:209–212. 2023.

55 

Akhtar T, Shahid S, Asghar A, Naeem MI, Aziz S and Ameer T: Utilisation of herbal bullets against newcastle disease in poultry sector of Asia and Africa (2012-2022). Int J Agric Biosci. 12:56–65. 2023.

56 

Du X, Gul ST, Ahmad L, Hussain R and Khan A: Fowl typhoid: Present scenario, diagnosis, prevention and control measures. Int J Agric Biosci. 12:172–179. 2023.

57 

Afzal Z, Javed MT, Mohsin M, Ahmad HMW, Saeed Z, Taimoor M, Aleem RA, Raza A, Ayub A, Israr F, et al: The usefulness of glutaraldehyde coagulation test as a conjuncture test in the diagnosis of tuberculosis in humans and animals. Agrobiol Rec. 15:34–40. 2024.

58 

Sarmykova M, Yespembetov B, Sambetbayev A, Tileukhanov K, Kaldyrkaev A, Shestakov A, Melisbek A, Burashev Y, Usserbayev B and Syrym N: Isolation and characterization of bacteriophage streptococcus equi for application against horse strangles. Int J Vet Sci. 13:691–699. 2024.

59 

Verma AK, Dhama K, Chakraborty S, Kumar A, Tiwari A, Rahal A, Mahima   and Singh SV: Strategies for combating and eradicating important infectious diseases of animals with particular reference to India: Present and future perspectives. Asian J Anim Vet Adv. 9:77–106. 2014.

60 

Nii-Trebi NI: Emerging and neglected infectious diseases: Insights, advances, and challenges. Biomed Res Int. 2017(5245021)2017.PubMed/NCBI View Article : Google Scholar

61 

Walker JW, Han BA, Ott IM and Drake JM: Transmissibility of emerging viral zoonoses. PLoS One. 13(e0206926)2018.PubMed/NCBI View Article : Google Scholar

62 

Watkins K: Emerging infectious diseases: A review. Curr Emerg Hosp Med Rep. 6:86–93. 2018.PubMed/NCBI View Article : Google Scholar

63 

Singh RK, Sharma GK, Mahajan S, Dhama K, Basagoudanavar SH, Hosamani M, Sreenivasa BP, Chaicumpa W, Gupta VK and Sanyal A: Foot-and-Mouth disease virus: Immunobiology, advances in vaccines and vaccination strategies addressing vaccine failures-an indian perspective. Vaccines (Basel). 7(90)2019.PubMed/NCBI View Article : Google Scholar

64 

Yatoo MI, Parray R, Bashir ST, Bhat RA, Gopalakrishnan A, Karthik K, Dhama K and Singh SV: Veterinary quarterly contagious caprine pleuropneumonia-a comprehensive review contagious caprine pleuropneumonia-a comprehensive review. Vet Q. 39:1–25. 2019.PubMed/NCBI View Article : Google Scholar

65 

Kabir A, Kalhoro DH, Abro SH, Kalhoro MS, Yousafzai1 HA, Shams S, Khan IU, Lochi GM, Mazari MQ, Baloch MW, et al: Peste des petits ruminants: A review. Pure Appl Biol. 8:1214–1222. 2019.

66 

World Health Organization (WHO): Influenza A (H1N2) variant virus-Brazil. WHO, Geneva, 2021.

67 

McKendrick AG: Studies on the theory of continuous probabilities, with special reference to its bearing on natural phenomena of a progressive nature. Proc London Math Soc. S2-S13:401–416. 1914.

68 

McCulloch K, Romero N, MacLachlan J, Allard N and Cowie B: Modeling progress toward elimination of hepatitis B in Australia. Hepatology. 71:1170–1181. 2020.PubMed/NCBI View Article : Google Scholar

69 

Farr W: On the cattle plague. J Soc Sci. 1:349–351. 1866.

70 

Snow J: On continuous molecular changes, more particularly in their relation to epidemic diseases. Rev Infect Dis. 7:441–447. 1985.

71 

Kermack WO and Mckendrick AG: A contribution to the mathematical theory of epidemics. Proc R Soc London Ser A, Contain Pap a Math Phys Character. 115:700–721. 1927.

72 

Kermack WO and Mckendrick AG: Contributions to the mathematical theory of epidemics. II. -The problem of endemicity. Proc R Soc London Ser A, Contain Pap a Math Phys Character. 138:55–83. 1932.

73 

Kermack WO and McKendrick AG: Contributions to the mathematical theory of epidemics. III.-Further studies of the problem of endemicity. Proc R Soc London Ser A, Contain Pap a Math Phys Character. 141:94–122. 1933.

74 

Kermack WO and McKendrick AG: Contributions to the mathematical theory of epidemics-III. Further studies of the problem of endemicity. Bull Math Biol. 53:89–118. 1991.PubMed/NCBI View Article : Google Scholar

75 

Ferguson NM, Donnelly CA and Anderson RM: The foot-and-mouth epidemic in Great Britain: Pattern of spread and impact of interventions. Science. 292:1155–1160. 2001.PubMed/NCBI View Article : Google Scholar

76 

Anderson I: Foot and mouth disease 2007: A review and lessons learned., 2008.

77 

Funk S, Camacho A, Kucharski AJ, Eggo RM and Edmunds WJ: Real-time forecasting of infectious disease dynamics with a stochastic semi-mechanistic model. Epidemics. 22:56–61. 2018.PubMed/NCBI View Article : Google Scholar

78 

Keeling MJ, Woolhouse MEJ, Shaw DJ, Matthews L, Chase-Topping M, Haydon DT, Cornell SJ, Kappey J, Wilesmith J and Grenfell BT: Dynamics of the 2001 UK foot and mouth epidemic: Stochastic dispersal in a heterogeneous landscape. Science. 294:813–817. 2001.PubMed/NCBI View Article : Google Scholar

79 

Hayama Y, Firestone SM, Stevenson MA, Yamamoto T, Nishi T, Shimizu Y and Tsutsui T: Reconstructing a transmission network and identifying risk factors of secondary transmissions in the 2010 foot-and-mouth disease outbreak in Japan. Transbound Emerg Dis. 66:2074–2086. 2019.PubMed/NCBI View Article : Google Scholar

80 

Picado A, Guitian FJ and Pfeiffer DU: Space-time interaction as an indicator of local spread during the 2001 FMD outbreak in the UK. Prev Vet Med. 79:3–19. 2007.PubMed/NCBI View Article : Google Scholar

81 

Keeling MJ and Rohani P: Modeling infectious diseases in humans and animals. Clin Infect Dis. 47:864–866. 2008.

82 

Kleczkowski A, Hoyle A and Mcmenemy P: One model to rule them all? Modelling approaches across OneHealth, for human, animal and plant epidemics. Philos Trans R Soc Lond B Biol Sci. 374(20180255)2019.PubMed/NCBI View Article : Google Scholar

83 

Thompson RN and Brooks-Pollock E: Detection, forecasting and control of infectious disease epidemics: Modelling outbreaks in humans, animals and plants. Philos Trans R Soc B Biol Sci. 374(20190038)2019.PubMed/NCBI View Article : Google Scholar

84 

Hart WS, Hochfilzer LFR, Cunniffe NJ, Lee H, Nishiura H and Thompson RN: Accurate forecasts of the effectiveness of interventions against Ebola may require models that account for variations in symptoms during infection. Epidemics. 29(100371)2019.PubMed/NCBI View Article : Google Scholar

85 

Looi LM and Chua KB: Lessons from the Nipah virus outbreak in Malaysia. Malays J Pathol. 29:63–67. 2007.PubMed/NCBI

86 

Mörner T, Obendorf DL, Artois M and Woodford MH: Surveillance monitoring of wildlife diseases. Rev Sci Tech. 21:67–76. 2002.PubMed/NCBI View Article : Google Scholar

87 

Gronvall G, Boddie C, Knutsson R and Colby M: One health security: An important component of the global health security agenda. Biosecur Bioterror. 12:221–224. 2014.PubMed/NCBI View Article : Google Scholar

88 

Corning S: World Organisation for animal health: Strengthening veterinary services for effective one health collaboration. Rev Sci Tech. 33:639–650. 2014.PubMed/NCBI View Article : Google Scholar

89 

Brooks-Pollock E, de Jong MCM, Keeling MJ, Klinkenberg D and Wood JLN: Eight challenges in modelling infectious livestock diseases. Epidemics. 10:1–5. 2015.PubMed/NCBI View Article : Google Scholar

90 

Metcalf CJE, Edmunds WJ and Lessler J: Six challenges in modelling for public health policy. Epidemics. 10:93–96. 2015.PubMed/NCBI View Article : Google Scholar

91 

Ezanno P, Andraud M, Beaunée G, Hoch T, Krebs S, Rault A, Touzeau S, Vergu E and Widgren S: How mechanistic modelling supports decision making for the control of enzootic infectious diseases. Epidemics. 32(100398)2020.PubMed/NCBI View Article : Google Scholar

92 

Craft ME, Beyer HL and Haydon DT: Estimating the probability of a major outbreak from the timing of early cases: An indeterminate problem? PLoS One. 8(e57878)2013.PubMed/NCBI View Article : Google Scholar

93 

Thompson RN, Gilligan CA and Cunniffe NJ: Detecting presymptomatic infection is necessary to forecast major epidemics in the earliest stages of infectious disease outbreaks. PLoS Comput Biol. 12(e1004836)2016.PubMed/NCBI View Article : Google Scholar

94 

Thompson PN and Etter E: Epidemiological surveillance methods for vector-borne diseases. Rev Sci Tech. 34:235–247. 2015.PubMed/NCBI View Article : Google Scholar

95 

Saegerman C, Humblet MF, Leandri M, Gonzalez G, Heyman P, Sprong H, L'Hostis M, Moutailler S, Bonnet SI, Haddad N, et al: First expert elicitation of knowledge on possible drivers of observed increasing human cases of tick-borne encephalitis in Europe. Viruses. 15(791)2023.PubMed/NCBI View Article : Google Scholar

96 

Ahmad M, Hussain A, Soomro MH and Jalbani S: Leptospirosis: An overview. In: One Health Triad. vol. 2 Abbas R, Saeed N, Younus M, Aguilar-Marcelino L and Khan A (eds.) Unique Scientific Publishers, Faisalabad, Pakistan, pp41-46, 2023.

97 

Wen N: Establishment and development of the disease surveillance system. Immun Progr China. 3:31–37. 2019.

98 

World Health Organization (WHO): Antiviral use and the risk of drug resistance. WHO, Geneva, 2009.

99 

Iqbal HMN: The quest for materials-based hydrogels with antimicrobial and antiviral potentialities. Open Virol J. 12:69–79. 2018.PubMed/NCBI View Article : Google Scholar

100 

Jones JC, Yen HL, Adams P, Armstrong K and Govorkova EA: Influenza antivirals and their role in pandemic preparedness. Antiviral Res. 210(105499)2023.PubMed/NCBI View Article : Google Scholar

101 

Nijhuis M, Van Maarseveen NM and Boucher CAB: Antiviral resistance and impact on viral replication capacity: Evolution of viruses under antiviral pressure occurs in three phases. Handb Exp Pharmacol. 189:299–320. 2009.PubMed/NCBI View Article : Google Scholar

102 

Herlocher ML, Truscon R, Elias S, Yen HL, Roberts NA, Ohmit SE and Monto AS: Influenza viruses resistant to the antiviral drug oseltamivir: Transmission studies in ferrets. J Infect Dis. 190:1627–1630. 2004.PubMed/NCBI View Article : Google Scholar

103 

Smyk JM, Szydłowska N, Szulc W and Majewska A: Evolution of influenza viruses-drug resistance, treatment options, and prospects. Int J Mol Sci. 23(12244)2022.PubMed/NCBI View Article : Google Scholar

104 

Malmsten M: Antimicrobial and antiviral hydrogels. Soft Matter. 7:8725–8736. 2011.

105 

Prasad M, Lambe UP, Brar B, Shah I, Ranjan JM, Rao R, Kumar S, Mahant S, Khurana SK, Iqbal HMN, et al: Nanotherapeutics: An insight into healthcare and multi-dimensional applications in medical sector of the modern world. Biomed Pharmacother. 97:1521–1537. 2018.PubMed/NCBI View Article : Google Scholar

106 

Pham T, Loupias P, Dassonville-Klimpt A and Sonnet P: Drug delivery systems designed to overcome antimicrobial resistance. Med Res Rev. 39:2343–2396. 2019.PubMed/NCBI View Article : Google Scholar

107 

Le LV, Mkrtschjan MA, Russell B and Desai TA: Hang on tight: Reprogramming the cell with microstructural cues. Biomed Microdevices. 21(43)2019.PubMed/NCBI View Article : Google Scholar

108 

Iqbal HMN and Dhama K: Emerging pathogens and bioactive materials: In Greening The 21st century biomedical sciences. J Exp Biol Agric Sci. 6:296–306. 2018.

109 

Iqbal HMN and Keshavarz T: Bioinspired polymeric carriers for drug delivery applications. In: stimuli responsive polymeric nanocarriers for drug delivery applications: Volume 1: Types and triggers. Elsevier, pp377-404, 2018.

110 

Rasheed T, Bilal M, Abu-Thabit NY and Iqbal HMN: The smart chemistry of stimuli-responsive polymeric carriers for target drug delivery applications. In: Stimuli responsive polymeric nanocarriers for drug delivery applications: Volume 1: Types and Triggers. Elsevier, pp61-99, 2018.

111 

Raza A, Hayat U, Rasheed T, Bilal M and Iqbal HMN: ‘Smart’ materials-based near-infrared light-responsive drug delivery systems for cancer treatment: A review. J Mater Res Technol. 8:1497–1509. 2019.

112 

Raza A, Rasheed T, Nabeel F, Hayat U, Bilal M and Iqbal H: Endogenous and exogenous stimuli-responsive drug delivery systems for programmed site-specific release. Molecules. 24(1117)2019.PubMed/NCBI View Article : Google Scholar

113 

Berger J, Reist M, Mayer JM, Felt O, Peppas NA and Gurny R: Structure and interactions in covalently and ionically crosslinked chitosan hydrogels for biomedical applications. Eur J Pharm Biopharm. 57:19–34. 2004.PubMed/NCBI View Article : Google Scholar

114 

Kalshetti PP, Rajendra VB, Dixit DN and Parekh PP: Hydrogels as a drug delivery system and applications: A review. Int J Pharm Pharm Sci. 4:1–7. 2011.

115 

Rodriguez VAM, Dhama K and Iqbal H: Biomaterials-based hydrogels and their drug delivery potentialities. Int J Pharmacol. 13:864–873. 2017.

116 

Jaguezeski AM, Souza CF, Perin G, Reis JH, Gomes TMA, Baldissera MD, Vaucher RA, de Andrade CM, Stefani LM, Gundel SS, et al: Effect of free and nano-encapsulated curcumin on treatment and energetic metabolism of gerbils infected by Listeria monocytogenes. Microb Pathog. 134(103564)2019.PubMed/NCBI View Article : Google Scholar

117 

Omwenga EO, Hensel A, Shitandi A and Goycoolea FM: Chitosan nanoencapsulation of flavonoids enhances their quorum sensing and biofilm formation inhibitory activities against an E.coli Top 10 biosensor. Colloids Surfaces B Biointerfaces. 164:125–133. 2018.PubMed/NCBI View Article : Google Scholar

118 

da Cunha JA, de Ávila Scheeren C, Fausto VP, de Melo LDW, Henneman B, Frizzo CP, de Almeida Vaucher R, de Vargas AC and Baldisserotto B: The antibacterial and physiological effects of pure and nanoencapsulated Origanum majorana essential oil on fish infected with Aeromonas hydrophila. Microb Pathog. 124:116–121. 2018.PubMed/NCBI View Article : Google Scholar

119 

Lopes LQS, de Almeida Vaucher R, Giongo JL, Gündel A and Santos RCV: Characterisation and anti-biofilm activity of glycerol monolaurate nanocapsules against Pseudomonas aeruginosa. Microb Pathog. 130:178–185. 2019.PubMed/NCBI View Article : Google Scholar

120 

Shin S, Ahmed I, Hwang J, Seo Y, Lee E, Choi J, Moon S and Hong JW: A microfluidic approach to investigating a synergistic effect of tobramycin and sodium dodecyl sulfate on Pseudomonas aeruginosa biofilms. Anal Sci. 32:67–73. 2016.PubMed/NCBI View Article : Google Scholar

121 

Dua K, Gupta G, Rao NK and Bebawy M: Nano-antibiotics: A novel approach in treating P. aeruginosa biofilm infections. Minerva Med. 109(400)2018.PubMed/NCBI View Article : Google Scholar

122 

Rozenbaum RT, Su L, Umerska A, Eveillard M, Håkansson J, Mahlapuu M, Huang F, Liu J, Zhang Z, Shi L, et al: Antimicrobial synergy of monolaurin lipid nanocapsules with adsorbed antimicrobial peptides against Staphylococcus aureus biofilms in vitro is absent in vivo. J Control Release. 293:73–83. 2019.PubMed/NCBI View Article : Google Scholar

123 

Thomas V, Yallapu MM, Sreedhar B and Bajpai SK: A versatile strategy to fabricate hydrogel-silver nanocomposites and investigation of their antimicrobial activity. J Colloid Interface Sci. 315:389–395. 2007.PubMed/NCBI View Article : Google Scholar

124 

Rajchakit U and Sarojini V: Recent developments in antimicrobial-peptide-conjugated gold nanoparticles. Bioconjug Chem. 28:2673–2686. 2017.PubMed/NCBI View Article : Google Scholar

125 

Kobayashi K, Yoon C, Oh SH, Pagaduan JV and Gracias DH: Biodegradable thermomagnetically responsive soft untethered grippers. ACS Appl Mater Interfaces. 11:151–159. 2019.PubMed/NCBI View Article : Google Scholar

126 

Grewal TK, Majeed S and Sharma S: Therapeutic implications of nano-encapsulated rifabutin, azithromycin & ethambutol against experimental Mycobacterium avium infection in mice. Indian J Med Res. 147:594–602. 2018.PubMed/NCBI View Article : Google Scholar

127 

Mohid SA, Ghorai A, Ilyas H, Mroue KH, Narayanan G, Sarkar A, Ray SK, Biswas K, Bera AK, Malmsten M, et al: Application of tungsten disulfide quantum dot-conjugated antimicrobial peptides in bio-imaging and antimicrobial therapy. Colloids Surfaces B Biointerfaces. 176:360–370. 2019.PubMed/NCBI View Article : Google Scholar

128 

Tsao CT, Chang CH, Lin YY, Wu MF, Wang JL, Han JL and Hsieh KH: Antibacterial activity and biocompatibility of a chitosan-gamma-poly(glutamic acid) polyelectrolyte complex hydrogel. Carbohydr Res. 345:1774–1780. 2010.PubMed/NCBI View Article : Google Scholar

129 

Tsao CT, Chang CH, Lin YY, Wu M, Wang JL, Young T, Han JL and Hsieh K: Evaluation of chitosan/γ-poly(glutamic acid) polyelectrolyte complex for wound dressing materials. Carbohydrate Polymers. 84:812–819. 2011.

130 

Bilal M, Rasheed T, Iqbal HMN, Li C, Hu H and Zhang X: Development of silver nanoparticles loaded chitosan-alginate constructs with biomedical potentialities. Int J Biol Macromol. 105:393–400. 2017.PubMed/NCBI View Article : Google Scholar

131 

Naeem MI, Rehman A, Zahid R, Tehseen U, Arbab Z, Aziz S, Akhtar T, Ahmad HM, Ullah MR, Akram Q, et al: Use of nanotechnology to mitigate antimicrobial resistance. Agrobiol Rec. 13:16–33. 2023.

132 

Bilal M, Zhao Y, Rasheed T, Ahmed I, Hassan S, Nawaz M and Iqbal H: Biogenic nanoparticle-chitosan conjugates with antimicrobial, antibiofilm, and anticancer potentialities: Development and characterization. Int J Environ Res Public Health. 16(598)2019.PubMed/NCBI View Article : Google Scholar

133 

Alshubaily FA and Al-Zahrani MH: Appliance of fungal chitosan/ceftriaxone nano-composite to strengthen and sustain their antimicrobial potentiality against drug resistant bacteria. Int J Biol Macromol. 135:1246–1251. 2019.PubMed/NCBI View Article : Google Scholar

134 

Zhang CY, Gao J and Wang Z: Bioresponsive nanoparticles targeted to infectious microenvironments for sepsis management. Adv Mater. 30(e1803618)2018.PubMed/NCBI View Article : Google Scholar

135 

Håkansson J, Ringstad L, Umerska A, Johansson J, Andersson T, Boge L, Rozenbaum RT, Sharma PK, Tollbäck P, Björn C, et al: Characterization of the in vitro, ex vivo, and in vivo efficacy of the antimicrobial peptide DPK-060 used for topical treatment. Front Cell Infect Microbiol. 9(174)2019.PubMed/NCBI View Article : Google Scholar

136 

Thorgeirsdöttir TO, Thormar H and Kristmundsottir T: Effects of polysorbates on antiviral and antibacterial activity of monoglyceride in pharmaceutical formulations. Pharmazie. 58:286–287. 2003.PubMed/NCBI

137 

Kristmundsdóttir T, Árnadóttir SG, Bergsson G and Thormar H: Development and evaluation of microbicidal hydrogels containing monoglyceride as the active ingredient. J Pharm Sci. 88:1011–1015. 1999.PubMed/NCBI View Article : Google Scholar

138 

Thormar H, Bergsson G, Gunnarsson E, Georgsson G, Witvrouw M, Steingrímsson O, De Clercq E and Kristmundsdóttir T: Hydrogels containing monocaprin have potent microbicidal activities against sexually transmitted viruses and bacteria in vitro. Sex Transm Infect. 75:181–185. 1999.PubMed/NCBI View Article : Google Scholar

139 

Rokhade AP, Patil SA and Aminabhavi TM: Synthesis and characterization of semi-interpenetrating polymer network microspheres of acrylamide grafted dextran and chitosan for controlled release of acyclovir. Carbohydr Polym. 67:605–613. 2007.

140 

Nair M, Jayant RD, Kaushik A and Sagar V: Getting into the brain: Potential of nanotechnology in the management of NeuroAIDS. Adv Drug Deliv Rev. 103:202–217. 2016.PubMed/NCBI View Article : Google Scholar

141 

Fiandra L, Colombo M, Mazzucchelli S, Truffi M, Santini B, Allevi R, Nebuloni M, Capetti A, Rizzardini G, Prosperi D and Corsi F: Nanoformulation of antiretroviral drugs enhances their penetration across the blood brain barrier in mice. Nanomedicine. 11:1387–1397. 2015.PubMed/NCBI View Article : Google Scholar

142 

Edagwa B, McMillan JE, Sillman B and Gendelman HE: Long-acting slow effective release antiretroviral therapy. Expert Opin Drug Deliv. 14:1281–1291. 2017.PubMed/NCBI View Article : Google Scholar

143 

Zhang W, Wang M, Tang W, Wen R, Zhou S, Lee C, Wang H, Jiang W, Delahunty IM, Zhen Z, et al: Nanoparticle-laden macrophages for tumor-tropic drug delivery. Adv Mater. 30(e1805557)2018.PubMed/NCBI View Article : Google Scholar

144 

Mobo BHP, Rabinowitz PM, Conti LA and Taiwo OA: Occupational health of animal workers. Human-Animal Med. 343–371. 2010.

145 

Trinity L, Merrill SC, Clark EM, Koliba CJ, Zia A, Bucini G and Smith JM: Effects of social cues on biosecurity compliance in livestock facilities: Evidence from experimental simulations. Front Vet Sci. 7(130)2020.PubMed/NCBI View Article : Google Scholar

146 

Salman MD: The role of veterinary epidemiology in combating infectious animal diseases on a global scale: The impact of training and outreach programs. Prev Vet Med. 92:284–287. 2009.PubMed/NCBI View Article : Google Scholar

147 

Alexandrino-Junior F, Silva KG, Freire MCLC, Lione VD, Cardoso EA, Marcelino HR, Genre J, de Oliveira AG and do Egito EST: A functional wound dressing as a potential treatment for cutaneous leishmaniasis. Pharmaceutics. 11(200)2019.PubMed/NCBI View Article : Google Scholar

148 

Amaral AC, Saavedra PHV, Souza ACO, de Melo MT, Tedesco AC, Morais PC, Felipe MS and Bocca AL: Miconazole loaded chitosan-based nanoparticles for local treatment of vulvovaginal candidiasis fungal infections. Colloids Surfaces B Biointerfaces. 174:409–415. 2019.PubMed/NCBI View Article : Google Scholar

149 

Amaral AC, Bocca AL, Ribeiro AM, Nunes J, Peixoto DL, Simioni AR, Primo FL, Lacava ZG, Bentes R, Titze-de-Almeida R, et al: Amphotericin B in poly (lactic-co-glycolic acid)(PLGA) and dimercaptosuccinic acid (DMSA) nanoparticles against paracoccidioidomycosis. J Antimicrob Chemother. 63:526–533. 2009.PubMed/NCBI View Article : Google Scholar

150 

Yu Y, Peng L, Liao G, Chen Z and Li C: Noncovalent complexation of amphotericin B with poly(β-Amino Ester) derivates for treatment of C. Neoformans infection. Polymers (Basel). 11(270)2019.PubMed/NCBI View Article : Google Scholar

151 

Mehrizi TZ, Ardestani MS, Hoseini MH, Khamesipour A, Mosaffa N and Ramezani A: Novel nano-sized chitosan amphotericin B formulation with considerable improvement against Leishmania major. Nanomedicine (Lond). 13:3129–3147. 2018.PubMed/NCBI View Article : Google Scholar

152 

Kumar R, Pandey K, Sahoo GC, Das S, Das V, Topno RK and Das P: Development of high efficacy peptide coated iron oxide nanoparticles encapsulated amphotericin B drug delivery system against visceral leishmaniasis. Mater Sci Eng C Mater Biol Appl. 75:1465–1471. 2017.PubMed/NCBI View Article : Google Scholar

153 

Heidari-Kharaji M, Taheri T, Doroud D, Habibzadeh S and Rafati S: Solid lipid nanoparticle loaded with paromomycin: In vivo efficacy against Leishmania tropica infection in BALB/c mice model. Appl Microbiol Biotechnol. 100:7051–7060. 2016.PubMed/NCBI View Article : Google Scholar

154 

Gupta PK, Jaiswal AK, Kumar V, Verma A, Dwivedi P, Dube A and Mishra PR: Covalent functionalized self-assembled lipo-polymerosome bearing amphotericin B for better management of leishmaniasis and its toxicity evaluation. Mol Pharm. 11:951–963. 2014.PubMed/NCBI View Article : Google Scholar

155 

Das S, Suresh P and Desmukh R: Design of Eudragit RL 100 nanoparticles by nanoprecipitation method for ocular drug delivery. Nanomedicine. 6:318–323. 2010.PubMed/NCBI View Article : Google Scholar

156 

Sangeetha S, Venkatesh DN, Adhiyaman R, Santhi K and Suresh B: Formulation of sodium alginate nanospheres containing amphotericin b for the treatment of systemic candidiasis. Trop J Pharm Res. 6:653–659. 2007.

157 

Louie A, Deziel M, Liu W, Drusano MF, Gumbo T and Drusano GL: Pharmacodynamics of caspofungin in a murine model of systemic candidiasis: Importance of Persistence of caspofungin in tissues to understanding drug activity. Antimicrob Agents Chemother. 49:5058–5068. 2005.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-December 2024
Volume 6 Issue 6

Print ISSN: 2632-2900
Online ISSN:2632-2919

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ahmad M, Ahmed I, Akhtar T, Amir M, Parveen S, Narayan E, Iqbal HM and Rehman S: Strategies and innovations for combatting diseases in animals (Review). World Acad Sci J 6: 55, 2024
APA
Ahmad, M., Ahmed, I., Akhtar, T., Amir, M., Parveen, S., Narayan, E. ... Rehman, S. (2024). Strategies and innovations for combatting diseases in animals (Review). World Academy of Sciences Journal, 6, 55. https://doi.org/10.3892/wasj.2024.270
MLA
Ahmad, M., Ahmed, I., Akhtar, T., Amir, M., Parveen, S., Narayan, E., Iqbal, H. M., Rehman, S."Strategies and innovations for combatting diseases in animals (Review)". World Academy of Sciences Journal 6.6 (2024): 55.
Chicago
Ahmad, M., Ahmed, I., Akhtar, T., Amir, M., Parveen, S., Narayan, E., Iqbal, H. M., Rehman, S."Strategies and innovations for combatting diseases in animals (Review)". World Academy of Sciences Journal 6, no. 6 (2024): 55. https://doi.org/10.3892/wasj.2024.270